Who Ucn GMP 2023.01 Eng
Who Ucn GMP 2023.01 Eng
Who Ucn GMP 2023.01 Eng
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
This document is a PDF generated from the WHO Guidelines for malaria hosted on the MAGICapp online
platform: https://app.magicapp.org/#/guideline/6860. Each time the content of the platform is updated, a new PDF version of the
Guidelines will be downloadable on the WHO Global Malaria Programme website to facilitate access where the Internet is not available.
Users should note the downloaded PDFs of the Guidelines may be outdated and not contain the latest recommendations. Please
consult with the website for the most up-to-date version of the Guidelines (https://www.who.int/teams/global-malaria-programme).
WHO/UCN/GMP/2023.01
Contact
WHO Global Malaria Programme
Appia Avenue 20, 1202 Geneva, Switzerland
gmpfeedback@who.int
https://www.who.int/teams/global-malaria-programme
Sponsors/Funding
Funding for the development and publication of the Guidelines was gratefully received from the Bill & Melinda Gates Foundation, The
Global Fund, the Spanish Agency for International Development Cooperation, Unitaid, and the United States Agency for International
Development (USAID).
Disclaimer
© World Health Organization 2023
Some rights reserved. This work is available under the Creative Commons AttributionNonCommercial-ShareAlike 3.0 IGO license (CC
BY-NC-SA 3.0 IGO; https://creativecommons.org/licenses/by-nc-sa/3.0/igo).
Under the terms of this license, you may copy, redistribute and adapt the work for noncommercial purposes, provided the work is
appropriately cited, as indicated below. In any use of this work, there should be no suggestion that WHO endorses any specific
organization, products or services. The use of the WHO logo is not permitted. If you adapt the work, then you must license your work
under the same or equivalent Creative Commons license. If you create a translation of this work, you should add the following
disclaimer along with the suggested citation: “This translation was not created by the World Health Organization (WHO). WHO is not
responsible for the content or accuracy of this translation. The original English edition shall be the binding and authentic edition”.
Any mediation relating to disputes arising under the license shall be conducted in accordance with the mediation rules of the World
Intellectual Property Organization.
Suggested citation. WHO Guidelines for malaria, 14 March 2023. Geneva: World Health Organization; 2023 (WHO/UCN/GMP/
2023.01). License: CC BY-NC-SA 3.0 IGO.
Sales, rights and licensing. To purchase WHO publications, see http://apps.who.int/bookorders. To submit requests for commercial use
and queries on rights and licensing, see http://www. who.int/about/licensing.
Third-party materials. If you wish to reuse material from this work that is attributed to a third party, such as tables, figures or images, it
is your responsibility to determine whether permission is needed for that reuse and to obtain permission from the copyright holder. The
risk of claims resulting from infringement of any third-party-owned component in the work rests solely with the user.
General disclaimers. The designations employed and the presentation of the material in this publication do not imply the expression of
any opinion whatsoever on the part of WHO concerning the legal status of any country, territory, city or area or of its authorities, or
concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps represent approximate border lines for
which there may not yet be full agreement.
The mention of specific companies or of certain manufacturers’ products does not imply that they are endorsed or recommended by
WHO in preference to others of a similar nature that are not mentioned. Errors and omissions excepted, the names of proprietary
products are distinguished by initial capital letters.
All reasonable precautions have been taken by WHO to verify the information contained in this publication. However, the published
material is being distributed without warranty of any kind, either expressed or implied. The responsibility for the interpretation and use
of the material lies with the reader. In no event shall WHO be liable for damages arising from its use.
The recommendations contained in this publication are based on the advice of independent experts, who have considered the best
available evidence, a risk–benefit analysis and other factors, as appropriate. This publication may include recommendations on the use
of medicinal products for an indication, in a dosage form, dose regimen, population or other use parameters that are not included in the
approved labelling. Relevant stakeholders should familiarize themselves with applicable national legal and ethical requirements. WHO
does not accept any liability for the procurement, distribution and/or administration of any product for any use.
2 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Sections
1. ABBREVIATIONS .................................................................................................................................................................................................................29
3. INTRODUCTION .................................................................................................................................................................................................................32
4. PREVENTION .......................................................................................................................................................................................................................36
4.2.2 Perennial malaria chemoprevention (PMC) - formerly intermittent preventive treatment of malaria in infants (IPTi) .........97
4.2.6.3 MDA to reduce transmission of P. falciparum in very low to low transmission settings ............................................... 127
4.2.6.4 MDA to reduce transmission of P. falciparum in moderate to high transmission settings ............................................ 132
4.2.6.5 MDA to reduce transmission of P. vivax ................................................................................................................................... 135
4.2.6.6 Mass relapse prevention (MRP) to reduce transmission of P. vivax ................................................................................... 140
5.2.1.3 Reducing the transmissibility of treated P. falciparum infections in areas of low-intensity transmission ................ 164
3 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
5.2.3.3 Monitoring efficacy and safety of antimalarial drugs and resistance ................................................................................ 201
6. INTERVENTIONS IN THE FINAL PHASE OF ELIMINATION AND PREVENTION OF RE-ESTABLISHMENT .......................................... 205
6.1 Interventions recommended for mass implementation in delimited geographical areas ...................................................................... 206
6.2.3 Testing and treatment at points of entry to reduce importation of malaria................................................................................. 215
7. SURVEILLANCE................................................................................................................................................................................................................. 230
10.5 Recommendations for interventions in the final phase of elimination and prevention of re-establishment ................................. 260
4 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Summary of recommendations
1. ABBREVIATIONS
2. EXECUTIVE SUMMARY
3. INTRODUCTION
4. PREVENTION
Pyrethroid-only long-lasting insecticidal nets (LLINs) should be deployed for the prevention and control of malaria
in children and adults living in areas with ongoing malaria transmission.
Remark:
• WHO recommends ITNs that have been prequalified by WHO for deployment in protecting populations at risk of
malaria.
• ITNs are most effective where the principal malaria vector(s) bite predominantly at night after people have retired under
their nets.
• ITNs can be used both indoors and outdoors, wherever they can be suitably hung (though hanging nets in direct sunlight
should be avoided, as sunlight can affect insecticidal activity).
5 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Pyrethroid-PBO ITNs instead of pyrethroid-only LLINs can be deployed for the prevention and control of malaria in
children and adults in areas with ongoing malaria transmission where the principal malaria vector(s) exhibit
pyrethroid resistance.
Remark:
The conditionality of this recommendation is largely driven by the current higher unit cost of pyrethroid-PBO ITNs compared
to pyrethroid-only LLINs and therefore the uncertainty of their cost-effectiveness. Furthermore, as PBO is less wash-resistant
than pyrethroids, its bioavailability declines faster over the three-year estimated life of an ITN; therefore, the added impact of
pyrethroid-PBO ITNs over that of pyrethroid-only LLINs may decline over time. The evidence comes from two sites in
eastern Africa with pyrethroid resistance and not from other geographies where transmission levels and vector characteristics
may vary. PBO acts by inhibiting certain metabolic enzymes, primarily oxidases, and so are likely to provide greater protection
than pyrethroid-only LLINs where mosquitoes display mono-oxygenase-based insecticide resistance mechanisms.
In deciding whether pyrethroid-PBO ITNs may be appropriate in their context, malaria programmes should:
• consider the deployment of pyrethroid-PBO ITNs in areas where resistance to pyrethroids in local vectors has been
detected;
• determine whether resources are adequate to cover the extra cost of pyrethroid-PBO ITNs, while ensuring that
coverage of populations at risk of malaria is not affected;
• note that WHO recommends that ITNs prequalified by WHO be selected for deployment.
Pyrethroid-chlorfenapyr ITNs should be deployed instead of pyrethroid-only LLINs for prevention of malaria in
adults and children in areas with pyrethroid resistance.
Remark:
Note: Recommendations on deployment of pyrethroid-chlorfenapyr nets were separated into two distinct recommendations for
better clarity, but share the same evidence to decision, justification, practical info and research needs. Please refer to the following
section.
6 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Pyrethroid-chlorfenapyr ITNs can be deployed instead of pyrethroid-PBO ITNs for prevention of malaria in adults
and children in areas with pyrethroid resistance.
Remark:
The conditionality of the recommendation to deploy pyrethroid-chlorfenapyr ITNs instead of pyrethroid-PBO ITNs is based
on the GDG’s judgement that the balance of desirable and undesirable effects probably favours pyrethroid-chlorfenapyr ITNs
over pyrethroid-PBO ITNs. However, the evidence for this recommendation is from only one trial in Africa.
In deciding whether to deploy pyrethroid-chlorfenapyr ITNs instead of pyrethroid-only LLINs or pyrethroid-PBO ITNs,
malaria programmes should:
• determine whether resources are adequate to cover the extra costs compared to pyrethroid-only LLINs or pyrethroid-
PBO ITNs, while ensuring optimal coverage of populations at risk of malaria;
• generate additional information or conduct analyses with the aim of maximizing impact through targeted deployment
(e.g. stratification of malaria risk, assessment of the characteristics of local vectors, such as pyrethroid resistance
mechanisms). ITNs for prevention of malaria in adults and children in areas with pyrethroid resistance; and
• note that WHO recommends that ITNs prequalified by WHO be selected for deployment.
Pyrethroid-pyriproxyfen ITNs can be deployed instead of pyrethroid-only LLINs for prevention of malaria in adults
and children in areas with pyrethroid resistance.
Remark:
The conditionality of the recommendation to deploy pyrethroid-pyriproxyfen ITNs instead of pyrethroid-only LLINs is based
on the GDG’s concerns that the available evidence indicates poor cost-effectiveness of pyrethroid-pyriproxyfen ITNs
compared to pyrethroid-only LLINs and that the extra resources currently required to purchase these ITNs may negatively
impact on coverage and equity.
In deciding whether pyrethroid-pyriproxyfen ITNs should be deployed instead of pyrethroid-only LLINs, malaria programmes
should:
• determine whether resources are adequate to cover the extra cost compared to pyrethroid-only LLINs, while ensuring
optimal coverage of populations at risk of malaria;
• generate additional information or conduct analyses with the aim of maximizing impact through targeted deployment
(e.g. stratification of malaria risk, assessment of the characteristics of local vectors, such as pyrethroid resistance
mechanisms); and
• note that WHO recommends that ITNs prequalified by WHO be selected for deployment.
Note: Recommendations on deployment of pyrethroid-pyriproxifen nets were separated into two distinct recommendations for
better clarity, but share the same evidence to decision, justification, practical info and research needs. Please refer to the following
section.
7 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Pyrethroid-pyriproxyfen ITNs are not recommended for deployment over pyrethroid-PBO ITNs for prevention of
malaria in adults and children in areas with pyrethroid resistance.
Remark:
The conditionality of the recommendation against the deployment of pyrethroid-pyriproxyfen ITNs instead of pyrethroid-
PBO ITNs is based on the GDG’s judgement that the balance of effects favours pyrethroid-PBO ITNs over pyrethroid-
pyriproxyfen ITNs and that, based on current cost and efficacy data, pyrethroid-PBO ITNs are more cost-effective. The GDG
acknowledged that evidence to support this recommendation is derived from only a single trial in Africa.
Insecticide-treated nets (ITNs) should be deployed for the prevention and control of malaria in children and adults
in areas with ongoing malaria transmission affected by a humanitarian emergency.
Remark:
This recommendation is limited to classes of ITNs currently recommended by WHO. As with ITNs deployed in more stable
settings, WHO recommends that ITNs that are prequalified by WHO be selected for use in humanitarian emergencies.
When considering deployment of ITNs in humanitarian emergencies, the infrastructure, access, logistical capacity and
resources available must be taken into account, as these may influence the feasibility and cost of procuring and deploying
nets.
Achieving and maintaining optimal coverage with ITNs for malaria prevention and control (2019)
To achieve and maintain optimal ITN coverage, countries should apply mass free net distribution through
campaigns, combined with other locally appropriate delivery mechanisms such as continuous distribution using
antenatal care (ANC) clinics and the Expanded Programme on Immunization (EPI).
Recipients of ITNs should be advised (through appropriate communication strategies) to continue using their nets
beyond the three-year expected lifespan, irrespective of the condition and age of the net, until a replacement net is
available.
Old ITNs should only be collected where there is assurance that: i) communities are not left without nets, i.e. new
ITNs are distributed to replace old ones; and ii) there is a suitable and sustainable plan in place for safe disposal of
the collected material.
If ITNs and their packaging (bags and baling materials) are collected, the best option for disposal is high-
temperature incineration. They should not be burned in the open air. In the absence of appropriate facilities, they
should be buried away from water sources and preferably in non-permeable soil.
Recipients of ITNs should be advised (through appropriate communication strategies) not to dispose of their nets in
any water body, as the residual insecticide on the net can be toxic to aquatic organisms (especially fish).
8 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
IRS should be deployed for the prevention and control of malaria in children and adults living in areas with ongoing
malaria transmission.
Remark:
WHO recommends that WHO-prequalified insecticidal products be selected for IRS use and that these be selected based on
the insecticide susceptibility of the local malaria vector(s). IRS is considered an appropriate intervention where:
IRS can be deployed for the prevention and control of malaria in children and adults in areas with ongoing malaria
transmission affected by a humanitarian emergency.
Remark:
The conditionality of this recommendation is largely driven by the very low certainty of the evidence that IRS reduces malaria
in such settings and due to concerns around feasibility and cost.
When deciding whether IRS may be appropriate for prevention and control of malaria in humanitarian emergency settings,
programmes should consider:
• whether the structures are suitable for spraying. Some shelters provided in emergency settings may not be suitable for
application of insecticides, such as open-sided structures and those built from materials that affect the residual nature of
the insecticides;
• whether the target coverage of IRS can be feasibly achieved in the setting;
• whether there are sufficient resources to cover the relatively high costs associated with an IRS programme. In such
settings, transport of commodities to hard-to-reach areas, coupled with the need to quickly procure items and establish
human capacity to deliver the intervention, is likely to incur higher costs than when deploying IRS in more stable
settings.
As with the deployment of IRS in more stable settings, WHO recommends that WHO-prequalified insecticides be selected
for IRS use in humanitarian emergencies. It is important to ensure that the vector population is susceptible to the insecticide
selected for spraying.
9 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Prioritize optimal coverage with either ITNs or IRS over combination (2019)
The co-deployment of ITNs and IRS is not recommended for prevention and control of malaria in children and
adults in areas with ongoing malaria transmission. Priority should be given to delivering either ITNs or IRS at
optimal coverage and to a high standard, rather than introducing the second intervention as a means to
compensate for deficiencies in the implementation of the first intervention.
Remark:
In settings where optimal ITN coverage, as specified in the strategic plan, has been achieved and where ITNs remain effective,
additionally implementing IRS may have limited utility in reducing malaria morbidity and mortality. Given the resource
constraints across malaria endemic countries, it is recommended that effort be focused on good-quality implementation of
either ITNs or IRS, rather than deploying both in the same area. However, the combination of these interventions may be
considered for resistance prevention, mitigation or management should sufficient resources be available.
Access to effective vector control using ITNs or IRS at optimal coverage levels should be ensured for all
populations at risk of malaria in most epidemiological and ecological settings.
In areas with ongoing local malaria transmission (irrespective of both the pre-intervention and current level of
transmission), vector control interventions should not be scaled back. Ensuring access to effective malaria vector
control at optimal levels for all inhabitants of such areas should be pursued and maintained.
10 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Larviciding (2019)
Insecticides can be regularly applied to water bodies (larviciding) for the prevention and control of malaria in
children and adults as a supplementary intervention to ITNs or IRS in areas with ongoing malaria transmission
where aquatic habitats are few, fixed and findable.
Remark:
The conditionality of this recommendation is due to the low certainty of evidence, the impact being limited to non-extensive
habitats, and concerns about feasibility.
• Larviciding only reduces vector density and so does not have the same potential for health impact as ITNs and IRS; ITNs
provide protection from biting vectors and both ITNs and IRS reduce adult longevity.
• Larviciding should not be seen as a substitute for ITNs or IRS or a means to fill a coverage gap in areas with significant
malaria risk; rather, larviciding represents a potential supplementary strategy for malaria control.
• Feasibility and cost-effectiveness should be taken into account; larviciding will generally be most cost-effective in areas
where larval habitats are few, fixed and findable, and likely less feasible in areas where the aquatic habitats are
abundant, scattered and variable.
The following settings are potentially the most suitable for larviciding as a supplementary measure implemented alongside
ITNs or IRS:
• urban areas: where breeding sites are relatively few, fixed and findable in relation to houses (which are targeted for ITNs
or IRS);
• arid regions: where larval habitats may be few and fixed throughout much of the year.
No recommendation can be made because the evidence on the effectiveness of a specific larval habitat
modification and/or larval habitat manipulation intervention for the prevention and control of malaria was deemed
to be insufficient.
No recommendation can be made because no evidence on the effectiveness of larvivorous fish for the prevention
and control of malaria was identified.
The deployment of topical repellents in areas with ongoing malaria transmission is not recommended if the aim is
to prevent and control malaria at the community level.
Remark:
The panel recommended against the implementation of topical repellents with the aim of controlling malaria at the
community level, given the lack of evidence of a significant impact. To achieve community-level impact, it is likely that a high
level of individual compliance would be needed. Further work is required to separate out the potential protective effects at
the individual and/or community level and therefore fully assess the potential public health value of topical repellents.
11 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Deployment of insecticide-treated clothing is not recommended for the prevention and control of malaria at the
community level in areas with ongoing malaria transmission; however, insecticide-treated clothing may be
beneficial as an intervention to provide personal protection against malaria in specific population groups.
Remark:
The GDG recommended against the deployment of insecticide-treated clothing due to the lack of evidence of an impact in
the general population. In the absence of ITNs, there is some evidence that insecticide-treated clothing may reduce the risk
of malaria infection in specific populations such as refugees and military personnel.
No recommendation can be made because the evidence on the effectiveness of spatial/airborne repellents for the
prevention and control of malaria was deemed to be insufficient.
Space spraying is not recommended for the prevention and control of malaria in children and adults in areas with
ongoing malaria transmission; IRS or ITNs should be prioritized instead.
Remark:
The panel recommended against the deployment of space spraying to control malaria, given the lack of evidence of impact
against malaria. Due to the short-lived nature of the insecticides used, space spraying is generally costly and wasteful of
resources.
Screening of residential houses can be used for the prevention and control of malaria in children and adults in areas
with ongoing malaria transmission.
Remark:
The GDG determined that a conditional recommendation should be given for house screening because of the low- to
moderate-certainty evidence of an impact against malaria. Furthermore, programmes would need to consider a number of
local contextual factors when considering screening of residential houses as a public health strategy, such as:
Programmes should note that this recommendation addresses the use of screening of windows, ceilings, doors and/or eave
spaces, and does not cover other ways of blocking entry points into houses.
12 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
In malaria-endemic areas, pregnant women of all gravidities should be given antimalarial medicine at
predetermined intervals to reduce disease burden in pregnancy and adverse pregnancy and birth outcomes.
Remark:
• Sulfadoxine-pyrimethamine (SP) has been widely used for malaria chemoprevention during pregnancy and remains
effective in improving key pregnancy outcomes.
• IPTp-SP should start as early as possible in the second trimester and not before week 13 of pregnancy.
• Doses should be given at least one month apart, with the objective of ensuring that at least three doses are received.
• Antenatal care (ANC) contacts remain an important platform for delivering IPTp. Where inequities in ANC service and
reach exist, other delivery methods (such as the use of community health workers) may be explored, ensuring that ANC
attendance is maintained and underlying inequities in ANC delivery are addressed.
• IPTp is generally highly cost-effective, widely accepted, feasible for delivery and justified by a large body of evidence
generated over several decades.
In areas of moderate to high perennial malaria transmission, children belonging to age groups at high risk of severe
malaria can be given antimalarial medicines at predefined intervals to reduce disease burden.
Remark:
• Perennial malaria chemoprevention (PMC) schedules should be informed by the age pattern of severe malaria
admissions, the duration of protection of the selected drug, and the feasibility and affordability of delivering each
additional PMC course (see “Practical info”).
• Sulfadoxine-pyrimethamine (SP) has been widely used for chemoprevention in Africa, including for PMC. Artemisinin-
based combination therapies (ACTs) have been effective when used for PMC, but evidence is limited on their safety,
efficacy, adherence to multi-day regimens, and cost-effectiveness in the context of PMC.
• Previously, PMC was recommended in infants (<12 months of age) as intermittent preventive treatment in infants (IPTi).
Since the initial recommendation, new data have documented the value of malaria chemoprevention in children aged 12
to 24 months.
• The Expanded Programme on Immunization (EPI) platform remains important for delivering PMC. Other methods of
delivery can be explored to optimize access to PMC and integration with other health interventions.
• Moderate to high perennial malaria transmission settings are defined as areas with P. falciparum parasite prevalence
greater than 10% or an annual parasite incidence greater than 250 per 1000 [30]. These thresholds are indicative and
should not be regarded as absolutes for determining applicability of the PMC recommendation.
13 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
In areas of seasonal malaria transmission, children belonging to age groups at high risk of severe malaria should be
given antimalarial medicines during peak malaria transmission seasons to reduce disease burden.
Remark:
• Eligibility for seasonal malaria chemoprevention (SMC) is defined by the seasonality of malaria transmission and age
groups at risk of severe malaria. Thresholds for assessing these criteria change over time and location. Malaria
programmes should assess the suitability of SMC based on the local malaria epidemiology and available funding (see
“Practical info”). The added value of a seasonally targeted intervention is likely to be greatest where transmission is
intensely seasonal.
• Monthly cycles of sulfadoxine-pyrimethamine plus amodiaquine (SP+AQ) have been widely used for SMC in African
children under 5 years old and have been shown to be efficacious, safe, well tolerated, available and inexpensive
(Thwing et al unpublished evidence).
School-aged children living in malaria-endemic settings with moderate to high perennial or seasonal transmission
can be given a full therapeutic course of antimalarial medicine at predetermined times as chemoprevention to
reduce disease burden.
Remark:
• Intermittent preventive treatment in school-aged children (IPTsc) has been evaluated in children aged 5–15 years. The
burden of malaria and benefits of IPTsc may vary across this age range, but evidence is limited.
• National malaria programmes can consider IPTsc if resources allow for its introduction among school-aged children
without compromising chemoprevention interventions for those carrying the highest burden of severe disease, such as
children < 5 years old.
• Schools may provide a low-cost means to deliver chemoprevention to school-aged children. However seasonal variation
in malaria transmission and the timing of school terms, as well as equity concerns, may mean alternative delivery
channels are needed to maximize impact.
• First- and second-line malaria treatments should not be used for IPTsc if safe and effective alternatives are available (see
“Practical info”).
• The dosing schedule for IPTsc should be informed by the local malaria epidemiology and timed to give protection during
the period of greatest malaria risk (see “Practical info”).
• Moderate to high malaria transmission settings are defined as areas with P. falciparum parasite prevalence greater than
10% or an annual parasite incidence greater than 250 per 1000 [30]. These thresholds are indicative and should not be
regarded as absolutes for determining applicability of the IPTsc recommendation.
14 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Children admitted to hospital with severe anaemia living in settings with moderate to high malaria transmission can
be given a full therapeutic course of an antimalarial medicine at predetermined times following discharge from
hospital to reduce re-admission and death.
Remark:
• Post-discharge malaria chemoprevention (PDMC) should be given to children following admission with severe
anaemia [133] that is not due to blood loss following trauma, surgery, malignancy or a bleeding disorder.
• PDMC implementation should be tailored to admissions of children with severe anaemia and consider the duration of
protection of the selected antimalarial, and the feasibility and affordability of delivering each additional PDMC course
(see “Practical info”).
• Moderate to high perennial malaria transmission settings are defined as areas with a P. falciparum parasite prevalence
greater than 10% or an annual parasite incidence greater than 250 per 1000 [30]. These thresholds are indicative and
should not be regarded as absolute for determining applicability of the PDMC recommendation.
Antimalarial medicine can be given as chemoprevention through mass drug administration (MDA) in areas of
moderate to high transmission of P. falciparum to provide short-term reductions in disease burden.
Remark:
• MDA may quickly reduce clinical malaria incidence in settings with moderate to high P. falciparum transmission, but
the effect wanes within 1–3 months. Therefore, if MDA is implemented, it should be one of several components of
a robust malaria control programme (including good coverage of effective case management and appropriate
prevention tools and strategies).
• Malaria programmes should judge the suitability of using MDA in their context based on the desired impact, level of
endemicity, and resources required. MDA for burden reduction should be targeted at moderate to high transmission
settings, regardless of seasonality (see “Practical info”).
• Moderate to high malaria transmission settings are defined as areas with P. falciparum parasite prevalence greater
than 10%, or incidence greater than 250 P. falciparum cases per 1000 population per year [30]. These thresholds
should not be regarded as absolutes for determining applicability of MDA implementation. It is biologically plausible
that MDA in intermediate transmission settings may reduce both disease burden and transmission intensity.
15 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
During emergencies or periods of health service disruption, antimalarial medicine can be used for mass drug
administration (MDA) in defined geographical areas to provide short-term reductions in the burden of disease
caused by P. falciparum.
Remark:
• MDA may quickly reduce clinical malaria incidence in settings with moderate to high P. falciparum transmission, but
the effect wanes within 1–3 months. As far as possible, MDA should be implemented as part of a package of malaria
control measures (including effective case management and appropriate prevention tools and strategies).
• Malaria programmes should judge the suitability of using MDA in their context based on the desired impact, level of
endemicity, and resources required (see “Practical info”).
• There is very limited evidence on the impact of MDA on disease in emergency settings. However, the biological
effects of MDA on disease in non-emergency settings are likely to translate to MDA recipients in emergency
settings. The size of effect will vary according to the type of emergency and level of disruption to health services, as
well as underlying transmission intensity, choice of drug, delivery method and other factors.
4.2.6.3 MDA to reduce transmission of P. falciparum in very low to low transmission settings
Conditional recommendation for , Low certainty evidence
MDA to reduce transmission of P. falciparum in very low to low transmission settings (2022)
In areas with very low to low levels of P. falciparum transmission, antimalarial medicine can be given as
chemoprevention through mass drug administration (MDA) to reduce transmission.
Remark:
• MDA may quickly reduce transmission of P. falciparum in very low to low transmission areas, but the effect wanes
within 1–3 months. Therefore, if MDA is implemented, it should be one of several components of a robust malaria
elimination programme (including, at minimum, good coverage of case-based surveillance with parasitological
diagnosis, effective antimalarial treatment, and appropriate prevention tools and strategies) in order to reduce the
risk of resurgence after the MDA programme has ended.
• MDA should be considered only for geographical areas where there is limited risk of importation of malaria either
from adjacent communities or through travel of the population to endemic areas.
• Malaria programmes should consider whether sufficient resources are available to implement MDA without
affecting other components of a robust malaria elimination programme.
• Very low to low transmission settings are defined as areas with P. falciparum parasite prevalence less than 10%, or P.
falciparum incidence less than 250 cases per 1000 population per year [30]. These thresholds should not be
regarded as absolutes for determining applicability of MDA implementation for transmission reduction. MDA
implemented in areas with levels of transmission near these cut-offs may reduce both disease burden and
transmission intensity.
16 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
In areas with moderate to high levels of P. falciparum transmission, providing antimalarial medicine through
mass drug administration (MDA) to reduce transmission is not recommended.
Remark:
• The studies included in the systematic review did not demonstrate evidence that MDA has either a short- or long-
term effect on P. falciparum transmission in moderate to high transmission settings.
• Recommendations on MDA to reduce the burden of malaria in moderate to high transmission settings can be found
in section 4.2.4.1 MDA for burden reduction. Moderate to high transmission settings are defined as areas with P.
falciparum parasite prevalence greater than 10%, or P. falciparum incidence above 250 cases per 1000 population
per year [30]. These thresholds should not be regarded as absolutes for determining applicability of MDA.
In areas with P. vivax transmission, antimalarial medicine can be given as chemoprevention through mass drug
administration (MDA) to reduce transmission.
Remark:
• MDA may quickly reduce transmission of P. vivax, but the effect wanes within 1–3 months. Therefore, if MDA is
implemented, it should be one of several components of a robust malaria elimination programme (including, at
minimum, good coverage of case-based surveillance with parasitological diagnosis, effective antimalarial treatment
including treatment for hypnozoites, and appropriate prevention tools and strategies) in order to reduce the risk of
resurgence after the MDA programme has ended.
• MDA should be considered only for geographical areas where there is limited risk of importation of malaria either
from adjacent communities or through travel of the population to endemic areas.
• Malaria programmes should consider whether sufficient resources are available to implement MDA without
affecting other components of a robust malaria elimination programme.
• Programmes considering implementing MDA for P. vivax should carefully reflect on how to safely and feasibly
administer treatment to prevent relapses.
17 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Mass treatment with an 8-aminoquinoline medicine alone to reduce the transmission of P. vivax is not
recommended.
Remark:
• Without testing for G6PD deficiency, the GDG noted the potential for severe harm from the use of a therapeutic
dose of an 8-aminoquinoline for radical cure of P. vivax hypnozoites. However, conducting G6PD testing for a large
population would significantly add to the complexity and cost of the intervention.
• The GDG noted that there may be highly exceptional circumstances under which mass relapse prevention (MRP)
may be appropriate, such as during a small focal outbreak of P. vivax in a temperate area. However, under such
circumstances the GDG considered that an MDA programme providing a schizonticide in addition to an
8-aminoquinoline would likely be a better strategy.
4.3 Vaccine
Strong recommendation for , High certainty evidence
The RTS,S/AS01 malaria vaccine should be used for the prevention of P. falciparum malaria in children living in regions
with moderate to high transmission as defined by WHO.
Remark:
• The RTS,S/AS01 malaria vaccine should be provided in a four-dose schedule in children from 5 months of age.
• Countries may consider providing the RTS,S/AS01 vaccine seasonally, with a five-dose strategy, in areas with highly seasonal
malaria or with perennial malaria transmission with seasonal peaks.
• Countries that choose to introduce the vaccine in a five-dose seasonal strategy are encouraged to document their
experiences, including adverse events following immunization.
• RTS,S/AS01 malaria vaccine should be provided as part of a comprehensive malaria control strategy.
18 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
5. CASE MANAGEMENT
All cases of suspected malaria should have a parasitological test (microscopy or RDT) to confirm the diagnosis.
Children and adults with uncomplicated P. falciparum malaria should be treated with one of the following
ACTs*:
• artemether-lumefantrine (AL)
• artesunate-amodiaquine (AS+AQ)
• artesunate-mefloquine (ASMQ)
• dihydroartemisinin-piperaquine (DHAP)
• artesunate + sulfadoxine-pyrimethamine (AS+SP)
• artesunate-pyronaridine (ASPY) (2022)
*Artesunate + sulfadoxine-pyrimethamine and artesunate-pyronaridine are not recommended for use in the
first trimester of pregnancy. For details of treatment using ACTs in the first trimester of pregnancy, see
5.2.1.4.1 below.
Remark:
Artesunate-pyronaridine is now included in the list of options for the treatment of uncomplicated malaria (2022). See the
full recommendation and supporting evidence below.
Remark:
• ASPY should be avoided by individuals with known liver disease (clinically apparent liver disease) because ASPY is
associated with liver transaminitis.
• Pharmacovigilance should be strengthened where ASPY is used for the treatment of malaria.
19 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Children weighing <25kg treated with dihydroartemisinin + piperaquine should receive a minimum of 2.5
mg/kg bw per day of dihydroartemisinin and 20 mg/ kg bw per day of piperaquine daily for 3 days.
In low-transmission areas, a single dose of 0.25 mg/kg bw primaquine should be given with an ACT to patients
with P. falciparum malaria (except pregnant women, infants aged < 6 months and women breastfeeding infants
aged < 6 months) to reduce transmission. G6PD testing is not required.
20 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Pregnant women with uncomplicated P. falciparum malaria should be treated with artemether-lumefantrine
during the first trimester.
Remark:
Infants weighing < 5 kg with uncomplicated P. falciparum malaria should be treated with an ACT at the
same mg/kg bw target dose as for children weighing 5 kg.
In people who have HIV/AIDS and uncomplicated P. falciparum malaria, artesunate + SP is not
recommended if they are being treated with co-trimoxazole, and artesunate + amodiaquine is not
recommended if they are being treated with efavirenz or zidovudine.
21 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Travellers with uncomplicated P. falciparum malaria returning to non-endemic settings should be treated
with an ACT.
Hyperparasitaemia (2015)
People with P. falciparum hyperparasitaemia are at increased risk for treatment failure, severe malaria and
death and should be closely monitored, in addition to receiving an ACT.
If the malaria species is not known with certainty, adults and children should be treated as for uncomplicated P.
falciparum malaria.
In areas with chloroquine-susceptible infections, adults and children with uncomplicated P. vivax, P. ovale, P.
malariae or P. knowlesi malaria should be treated with either an ACT or chloroquine.
In areas with chloroquine-resistant infections, adults and children with uncomplicated P. vivax, P. ovale, P.
malariae or P. knowlesi malaria should be treated with an ACT.
* For details of treatment using ACTs in the first trimester of pregnancy, see section 5.2.1.4.1.
The G6PD status of patients should be used to guide administration of primaquine for preventing relapse.
To prevent relapse, children and adults (except pregnant women, infants aged < 6 months, women
breastfeeding older infants unless they are known not to be G6PD deficient, and people with G6PD deficiency)
should be treated with a 14-day course of primaquine in all transmission settings.
22 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
To prevent relapse, an additional treatment option of using primaquine 0.5 mg/kg/day for seven days is
recommended to treat P. vivax or P. ovale malaria in children and adults (except pregnant women, infants aged <
6 months, women breastfeeding infants aged < 6 months, women breastfeeding older infants unless they are
known not to be G6PD deficient, and people with G6PD deficiency).
Remark:
• As recommended previously, the G6PD status of patients should be used to guide administration of primaquine for
preventing relapse.
• A shorter regimen can lead to better adherence compared to the standard 14-day regimen and thus to fewer
relapses.
To prevent relapse, an additional treatment option of using primaquine 1.0 mg/kg/day for seven days to treat P.
vivax or P. ovale malaria is not recommended.
Remark:
• There is a significantly increased risk of serious adverse events (moderate to large undesirable effect) at this daily
dosing of the standard high dose.
In people with G6PD deficiency, primaquine base at 0.75 mg/kg bw once a week for 8 weeks can be given to
prevent relapse, with close medical supervision for potential primaquine-induced haemolysis.
When G6PD status is unknown and G6PD testing is not available, a decision to prescribe primaquine should be
based on an assessment of the risks and benefits of adding primaquine.
In women who are pregnant or breastfeeding, weekly chemoprophylaxis with chloroquine can be given until
delivery and breastfeeding are completed, then, on the basis of G6PD status, primaquine can be given to
prevent future relapse.
23 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
5.2.2.1 Artesunate
Adults and children with severe malaria (including infants, pregnant women in all trimesters and lactating
women) should be treated with intravenous or intramuscular artesunate for at least 24 h and until they can
tolerate oral medication. Once a patient has received at least 24 h of parenteral therapy and can tolerate oral
therapy, treatment should be completed with 3 days of an ACT.
Children weighing < 20 kg should receive a higher dose of artesunate (3 mg/kg bw per dose) than larger
children and adults (2.4 mg/kg bw per dose) to ensure equivalent exposure to the drug.
*Not evaluated using the GRADE framework; recommendation based on pharmacokinetic modelling
If artesunate is not available, artemether should be used in preference to quinine for treating children and
adults with severe malaria.
Where complete treatment of severe malaria is not possible, but injections are available, adults and children
should be given a single intramuscular dose of artesunate, and referred to an appropriate facility for further
care. Where intramuscular artesunate is not available, intramuscular artemether or, if that is not available,
intramuscular quinine should be used.
Where intramuscular injection of artesunate is not available, children < 6 years should be treated with a single
rectal dose (10mg/kg bw) of artesunate, and referred immediately to an appropriate facility for further care.
Rectal artesunate should not be used in older children and adults.
24 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
National drug and regulatory authorities should ensure that the antimalarial medicines provided in both the
public and the private sectors are of acceptable quality, through regulation, inspection and law enforcement.
All malaria programmes should regularly monitor the therapeutic efficacy of antimalarial drugs using the
standard WHO protocols.
The choice of ACTs in a country or region should be based on optimal efficacy, safety and adherence.
Drugs used as first line treatment should not be used in IPTp, PMC, SMC, IPTsc or MDA.
When possible:
fixed-dose combinations should be used rather than co-blistered or loose, single-agent formulations; andfor young
children and infants, paediatric formulations, with a preference for solid formulations (e.g. dispersible tablets) should be
used rather than liquid formulations.
25 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Mass testing and treatment (MTaT) to reduce the transmission of malaria is not recommended.
Remark:
The GDG noted that there may be exceptional circumstances under which MTaT might be appropriate, such as a transmission
focus in a very low transmission or post-elimination setting where MDA is not an acceptable or feasible strategy.
In areas with very low to low transmission or post-elimination settings preventing re-establishment of transmission,
antimalarial medicine can be given as chemoprevention to people with increased risk of infection relative to the
general population to reduce transmission.
Remark:
• Persons given antimalarials should be those with increased risk of infection compared to the general population and
their infections should constitute a large proportion of the parasite reservoir in the area.
• The factors identifying individuals or groups at increased risk of infection should be easy to recognise, thereby
improving the acceptability and feasibility of the intervention.
• Programmes considering implementing targeted drug administration for P. vivax should carefully consider how to safely
and feasibly administer treatment to prevent relapses.
• Care should be taken to avoid stigmatizing groups at increased risk of infection.
• Additional complementary strategies to eliminate or prevent re-establishment of malaria transmission should be in place.
Testing and treatment of people with an increased risk of infection relative to the general population to reduce the
transmission of malaria is not recommended.
Remark:
The GDG noted that there may be limited circumstances under which targeted testing and treatment (TTaT) could be
beneficial. For example, TTaT could be used when people at a higher risk of infection can be easily identified and
chemoprevention is not acceptable to the population. Additionally, TTaT could be used if safe and effective implementation of
radical cure to prevent P. vivax relapses is only feasible for those with confirmed infections.
26 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Routine malaria testing and treatment of people arriving at points of entry (land, sea or air) to reduce importation is
not recommended.
Remark:
No studies of the impact of testing and treatment at points of entry on the rate of malaria importation were found by the
systematic review. Routine testing and treatment for malaria at points of entry is unlikely to be acceptable or feasible to
implement.
Malaria testing and treatment of organized or identifiable groups arriving or returning from malaria-endemic
areas (2022)
Relatively easy access to these groups within a short time after entry is required for this strategy to be feasible and
acceptable. This strategy may be particularly critical to areas in post-elimination that are working to prevent re-
establishment of transmission.
Remark:
• Programmes implementing reactive drug administration (RDA) should have the capacity to conduct case investigations
at the residence to determine the likely location of infection and to identify those individuals co-exposed with the index
case.
• Programmes implementing RDA should have the capacity to enumerate and provide antimalarials to the people residing
with or near a confirmed malaria case and others that share the same risk of infection.
• The people given antimalarial medicine in an RDA intervention should share the same risk of having acquired infection
as the index case or be at risk of acquiring infection from the index case. This includes residents in the same household
or neighborhood, co-travellers and co-workers. However, if the infection was imported and the residence is not located
in a receptive area, there may be no benefit from RDA.
• Programmes contemplating implementation of RDA for P. vivax should carefully consider how to safely and feasibly
administer treatment to prevent relapses.
27 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Remark:
Until an area is nearing elimination or is post-elimination, it is unlikely that reactive case detection and treatment (RACDT)
will have any effect on malaria transmission. However, RACDT becomes an essential component of surveillance when
countries are nearing interruption of transmission to monitor progress towards elimination. When countries are post-
elimination and working towards certification, RACDT can strengthen a country’s claim that it has reached and maintained
zero indigenous cases. RACDT is an essential part of surveillance and response to prevent re-establishment of malaria.
Remark:
• In areas approaching elimination or post-elimination settings where proactive indoor residual spraying (IRS) is occurring,
programmes can consider switching to reactive IRS only, depending on the receptivity of the area.
• Programmes considering adding reactive IRS on top of proactive IRS should balance the potential added benefit with
increasing cost and the risk of insecticide resistance.
• In areas approaching elimination or post-elimination settings where no IRS is occurring, initiating reactive IRS may be
beneficial, depending on whether IRS is a suitable vector control strategy. IRS is most effective where the vector
population is susceptible to the insecticide(s) being applied, the majority of mosquitoes feed and rest indoors and where
most structures are suitable for spraying.
• If the index infection was imported and the residence is not located in a receptive area, there may be no benefit from
reactive IRS.
28 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
7. SURVEILLANCE
8. METHODS
9. GLOSSARY
10. CONTRIBUTORS AND INTERESTS
10.5 Recommendations for interventions in the final phase of elimination and prevention of re-
establishment
1. ABBREVIATIONS
29 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
30 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
2. EXECUTIVE SUMMARY
The consolidated WHO Guidelines for malaria present all of the school aged children (IPTsc), post-discharge malaria
current WHO recommendations for malaria. These are the product chemoprevention (PDMC) and mass drug administration
of careful evaluation following standardized methods as part of the (MDA);
WHO process for developing guidelines [1]. WHO uses strictly • evidence-based recommendation on the use of the malaria
defined processes to assess the quality, consistency and vaccine;
completeness of evidence to determine the strength of each • evidence-based recommendations on the treatment of
recommendation. uncomplicated and severe malaria in all age groups and
situations, including in young children and pregnant women;
WHO malaria recommendations tend to be short, evidence-based and
statements. They are usually accompanied by supplementary • guidance on interventions in the final phase of elimination
statements which draw attention to contextual and and prevention of re-establishment.
implementation considerations that may influence the
appropriateness and impact of a recommendation in different
No guidance is given on the use of antimalarial agents to prevent
settings. Clearly distinguishing recommendations from their
malaria in people travelling from non-endemic settings to areas of
associated contextual considerations provides a degree of
malaria transmission. This is available in the WHO International
flexibility for national policy-makers to adopt and adapt the
travel and health guidance [2].
strategies that are most appropriate in their settings.
WHO guidelines, recommendations and good practice statements
This online platform and the associated PDF help to distinguish A WHO guideline is any document developed by WHO containing
the formal recommendations from the supplementary statements. recommendations for clinical practice, or public health practice or
The Global Malaria Programme will use this platform to produce health policy. A recommendation informs the intended end-user
“living guidelines”, which can be updated more rapidly than printed what he or she can or should do in specific situations to achieve
documents as new evidence becomes available. The tabs below the best possible health outcomes, individually and/or collectively.
each recommendation enable users to access the research It guides the choice among different interventions or measures to
evidence and evidence-to-decision (EtD) frameworks that ensure a positive impact on health and implications for the use of
informed the recommendation. There is also a feedback tab where resources.
users are encouraged to provide input directly related to each
intervention. In certain situations, good practice statements may be provided.
These statements reflect the consensus of the Guidelines
Scope Development Group (GDG) that the benefits of adhering to the
The consolidated WHO Guidelines for malaria bring together all intervention or course of action are large and unequivocal, and do
recommendations for malaria, including prevention using vector not need to be supported by a systematic evidence review or
control, preventive chemotherapy and the vaccine; diagnosis, could be based on indirect evidence.
treatment and elimination strategies. The Guidelines also provide
links to other resources including unpublished evidence reviewed The primary purpose of these WHO Guidelines is to support
at the time of formulating recommendations, guidance and policy-makers in ministries of health and the managers of national
information on: strategic use of information to drive impact; malaria control programmes in endemic countries to establish
surveillance, monitoring and evaluation; operational manuals, national policies and plans tailored to their local context.
handbooks and frameworks; and a glossary of terms and
definitions. Link to WHO prequalification
When a recommendation is linked to the introduction of a new
The Guidelines provide: tool or product, there is a parallel process managed by the WHO
Prequalification Team to ensure that diagnostics, medicines,
• evidence-based recommendations pertaining to vector
vaccines and vector control products meet global standards of
control tools, technologies and approaches that are currently
quality, safety and efficacy, in order to optimize use of health
available for malaria prevention and control, and for which
resources and improve health outcomes. The prequalification
sufficient evidence on their efficacy is available to support
process consists of a transparent, scientifically sound assessment,
systematic reviews. The Guidelines are intended to provide an
including dossier review, consistency testing or performance
underlying framework for the design of effective, evidence-
evaluation, and site visits to manufacturers. This information, in
based national vector control strategies and their adaptation
conjunction with other procurement criteria, is used by the United
to local disease epidemiology and vector bionomics;
Nations (UN) and other procurement agencies to make purchasing
• evidence-based recommendations on the use of antimalarial
decisions regarding these health products. This parallel process
medicines as preventive chemotherapy in people living in
aims to ensure that recommendations are linked to prequalified
malaria-endemic areas who are at risk of malaria morbidity
products and that prequalified products are linked to a
and mortality. These approaches include intermittent
recommendation for use.
preventive treatment (IPT) in pregnancy (IPTp), perennial
malaria chemoprevention (PMC), seasonal malaria
Expert input is important for the interpretation of the evidence,
chemoprevention (SMC), intermittent preventive treatment in
31 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
and the development of guidance may rely on expert opinion, dissemination channels. From this point forward, these
particularly in areas where the evidence is currently weak, scarce consolidated Guidelines represent the latest and definitive
or absent. For example, the vector control recommendations reference for all WHO guidance on malaria.
presented in the Guidelines are based on a consideration of the
evidence gained from randomized controlled trials (RCTs) and Dissemination
other types of trials and studies, as well as the technical These consolidated WHO Guidelines for malaria are available on the
knowledge and experience of the GDG and External Review Group MAGICapp online platform, linked to the WHO malaria website.
involved in the standard guideline development process. The original English version has been translated into French,
Spanish and Arabic. All research evidence and references are
Updating evidence-based guidance available on the web platform and will be available to download,
The first edition of these consolidated Guidelines was released in and relevant implementation guidance will be linked to the
early 2021 as a compilation of the existing recommendations for recommendations. When recommendations are updated, they will
malaria vector control and treatment. be labelled as such and will always display the date of the most
recent update. Each time there is an update, an updated PDF
This version of the Guidelines includes new WHO version of the Guidelines will be downloadable on the WHO
recommendations on two classes of insecticide-treated nets that Global Malaria Programme website to facilitate access where the
were established following the generation of evidence on the Internet is not reliably available. Users should note that older
epidemiological impact of pyrethroid-chlorfenapyr and pyrethroid- downloaded PDFs of the Guidelines may be outdated and may not
pyriproxyfen ITNs. Given the increasing complexity in contain the latest recommendations.
prioritization of limited resources across different ITN products,
WHO and partners also developed separate guidance on ITN WHO Headquarters will work closely with its regional and country
prioritization [3] under resource constrained conditions. offices to ensure the wide dissemination of the Guidelines to all
malaria-endemic countries. There will also be dissemination
Future updates for treatment include recommendations that are
through regional, sub-regional and country meetings. Member
already in the Guidelines but for which the evidence was not
States will be supported to adapt and implement these Guidelines.
previously subjected to the GRADE process, and new molecules
under development that will be included once the evidence base Feedback
becomes available. The Global Malaria Programme welcomes feedback, either via the
tab associated with each recommendation or by e-mail to
Readers should note the dates of individual recommendations.
gmpfeedback@who.int, to help identify recommendations in need
Revisions to these Guidelines will be communicated via the Global
of update or development.
Malaria Programme website and through WHO’s standard
3. INTRODUCTION
Background (Table 1). The goals focus attention on the need to both reduce
Malaria continues to cause unacceptably high levels of disease and morbidity and mortality, and to progressively eliminate malaria
death, as documented in successive editions of the World malaria from countries that had malaria transmission in 2015. The GTS
report [4]. According to the latest report, there were an estimated presents a framework through which the goals can be achieved
247 million cases and 619 000 deaths globally in 2021. Malaria is (Figure 1).
preventable and treatable, and the global priority is to reduce the
burden of disease and death while retaining the long-term vision Table 1. Goals, milestones and targets for the Global technical
of malaria eradication. Here, we present the WHO Guidelines for strategy for malaria 2016–2030
malaria developed by the WHO Global Malaria Programme as a
comprehensive and inclusive resource for advice on malaria.
32 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Objectives
These consolidated WHO Guidelines for malaria aim to provide the
latest evidence-based recommendations in one reference to
support countries in their efforts to reduce and ultimately
eliminate malaria. The objectives of the Guidelines are:
33 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
the certainty of that evidence using the GRADE (Grading of are diseases that predominantly impact the chronically
Recommendations Assessment, Development, and Evaluation) disadvantaged. While the magnitude and extent of health
approach. The synthesized and graded evidence on the health inequalities remain poorly understood, it is clear that certain
effects of interventions, as well as any evidence on contextual population groups have persistently higher disease mortality and
factors, is used to develop an evidence-to-decision (EtD) morbidity and more limited access to life-saving interventions. The
framework for each recommendation [7]. The judgement on the report documents that the poorest, least educated and rural
different factors in the EtD framework (including the certainty of groups are less likely to seek care for children with fever.
evidence) facilitates the determination of the strength and
direction of each recommendation. In most countries, Member States have not adequately identified
and addressed social and structural barriers to health, or taken
Expert input is important for the interpretation of the evidence, action to ensure gender equality, equity and human rights.
and the development of guidance may rely on expert opinion, Communities are often excluded from health decision-making,
particularly in areas where the evidence is currently weak, scarce even though people are entitled to active, free and meaningful
or absent. For example, the vector control recommendations participation in decisions that directly affect them, such as the
presented in the Guidelines are based on a consideration of the design, implementation and monitoring of health interventions.
evidence gained from randomized controlled trials (RCTs) and Participation increases ownership and helps to ensure that policies
other types of trials and studies, as well as the technical and programmes are responsive to the needs of the people they
knowledge and experience of the GDG and External Review Group are intended to benefit.
involved in the standard guideline development process. Details of
The existing inequities are barriers to achieving global and national
how evidence is considered are presented in Section 8: Methods.
goals and targets on malaria. Successful implementation of malaria
Details of contributors for specific recommendations are
control interventions should, therefore, be viewed through a
presented in Section 10: Contributors and interests.
human rights and health equity lens. This means fully
Target audience acknowledging the importance of engaging people in the design
The primary audience for these Guidelines is policy-makers in and delivery of health and care systems to meet their needs, and
ministries of health and the managers of NMPs in endemic empowering them to make informed decisions about their health
countries. The Guidelines may also be of interest to health care and take action.
practitioners, environmental health service professionals,
As many of the malaria interventions are reliant on broader health
procurement agencies, the private sector, and civil society groups.
care delivery platforms, a rights-based approach is required to
The Guidelines are also intended for use by international
ensure that quality health services and programmes are available,
development partners, donors and funding agencies in order to
accessible and acceptable to all those in need, including nomadic
support decision-making on allocation of resources for
populations, individuals with disabilities, out-of-school youth, and
interventions and procurement of appropriate malaria control
those living in sparsely populated and underserved areas far from
products. In addition, the Guidelines are intended to guide
health services and schools.
researchers, research funders and those interested in the
outcomes of research to address the evidence gaps that are National programmes should address inequity concerns by
constraining the development of guidance or weakening current monitoring the coverage of recommended interventions among
recommendations. individuals in identified risk categories and targeting those most at
risk. Health inequities and barriers to health need to be
Equity, gender and human rights
systematically identified and addressed by Member States and
The right to enjoy the highest attainable standard of physical and
other stakeholders through gender-responsive, equitable and
mental health (commonly referred to as the right to health) is
human rights-based health systems, with a focus on individuals
enshrined in several international human rights treaties, regional
and groups experiencing intersecting forms of discrimination,
agreements, and national constitutions and laws. Member States
marginalization and/or social exclusion.
have minimum “core” obligations that include “the prevention,
treatment and control of epidemic, endemic, occupational and Etiology
other diseases” [8]. Malaria is a life-threatening disease caused by the infection of red
blood cells with protozoan parasites of the genus Plasmodium that
Yet, gender-based discrimination, human rights violations, and
are transmitted to people through the bites of infected female
inequities related to social, economic, environmental, commercial
Anopheles mosquitoes. Four species of Plasmodium (P. falciparum, P.
and political determinants of health deprive billions of people
vivax, P. malariae and P. ovale) most commonly infect humans. P.
around the world of their right to enjoy the highest attainable
falciparum and P. vivax are the most prevalent species and P.
standard of health and well-being. It is of great concern that, over
falciparum is the most dangerous. A fifth species, P. knowlesi (a
the past few years, health inequities have been exacerbated by the
species of Plasmodium that primarily infects non-human primates)
impacts of the ongoing and interlinked crises of the coronavirus
is increasingly being reported in humans inhabiting forested
disease (COVID-19), conflict, climate change, food insecurity and
regions of some countries of South-East Asia and the Western
the global economy.
Pacific regions, and in particular on the island of Borneo.
Too many people are missing out on the interventions they need
to keep them healthy, including interventions to prevent and treat Malaria transmission, acquisition of immunity, and clinical
malaria. According to a WHO report [9], malaria, TB and HIV/AIDS manifestations of disease
34 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The intensity of transmission depends on factors related to the consequence of the pattern and intensity of malaria transmission
parasite, the vector, the human host and the environment. in the area of residence. In areas of moderate to high transmission,
Transmission tends to be more intense in places where the partial immunity to clinical disease and a reduced risk of
mosquito lifespan is longer and where the females prefer to bite developing severe malaria are acquired in early childhood. The
humans rather than other animals. The survival and longevity of pattern of acquired immunity is similar across the Sahel subregion,
female mosquitoes is of critical importance in malaria transmission, where malaria transmission is intense only during the three- or
as the malaria parasite generally requires a period of 7–10 days to four-month rainy season and low at other times. In both these
develop inside the mosquito into a form that is infective to situations, clinical disease is confined mainly to young children,
humans. Female mosquito longevity is dependent on intrinsic, who may develop high parasite densities that can progress rapidly
genetic factors, as well as on environmental factors including to severe malaria. By contrast, in these settings, adolescents and
temperature and humidity. The strong human-biting habit of the adults are partially immune and suffer clinical disease much less
African vector species is one of the reasons why approximately frequently, although they are often infected with low blood-
90% of the world’s malaria cases occur in Africa. parasite densities. Immunity is modified in pregnancy and
gradually lost, at least partially, when individuals move out of the
Transmission intensity is usually assessed as the incidence of cases endemic areas for prolonged periods (e.g. a year or more).
or the prevalence of infection. Most countries have information on
the annual parasite incidence (number of new parasitologically In areas of low and very low transmission, as found in much of
confirmed malaria cases per 1000 population per year) from Asia, Latin America and other malaria-endemic areas, the
routine surveillance and/or on the parasite prevalence from transmission fluctuates widely by season, year, and over relatively
surveys, often conducted during or just after periods of peak small distances. P. vivax is an important cause of malaria in these
transmission [10]. regions. This generally low transmission delays acquisition of
immunity, so that adults and children alike suffer from acute
The following categories of transmission intensity are indicative clinical malaria, with a significant risk for progression to severe
and meant to provide an adaptable framework in which each malaria if left untreated. Epidemics may occur in these low or very
country can conduct a stratification exercise to classify low transmission areas when the inoculation rate increases rapidly
geographical units according to local malaria transmission. because of a sudden increase in vectorial capacity. Epidemics may
result in a very high incidence across all age groups, which can
• Areas of high transmission are characterized by an annual overwhelm health services.
parasite incidence of 450 or more cases per 1000 population
and a P. falciparum prevalence rate of ≥35%. In moderate and high transmission areas with sustained high
• Moderate transmission areas have an annual parasite coverage of vector control and access to treatment, reduced
incidence of 250–450 cases per 1000 population and a exposure to malaria infection may change the population structure
prevalence of P. falciparum/P. vivax malaria of 10–35%. of acquired immunity to reflect that found in low or very low
• Areas of low transmission have an annual parasite incidence transmission areas, resulting in a corresponding change in the
of 100–250 cases per 1000 population and a prevalence of P. clinical epidemiology of malaria and an increasing risk of epidemics
falciparum/P. vivax of 1–10%. It should be noted that the if control measures are not sustained.
incidence of cases or infections is a more useful measure in
geographical units in which the prevalence is low, given the Recommendations and supporting implementation guidance
difficulty of measuring prevalence accurately at low Evidence-informed recommendations are a critical component to
levels [11]. support the development of national malaria strategic plans; they
• Very low transmission areas have an annual parasite incidence are intended to communicate “what to do”. A second critical
of < 100 cases per 1000 population and a prevalence of P. element is the strategic use of local data. This informs an
falciparum/P. vivax malaria that is > 0 but < 1%. understanding of the contextual diversity within each malaria-
endemic country. Local data provide an understanding of the
The relation between parasite incidence, parasite prevalence and different types of settings – or strata – within each country. This is
the number of cases presenting to health facilities per week can an essential prerequisite to identify the optimal mix of
be estimated using models [12]. Differences in transmission from interventions and the best means to deliver them in the different
one area to another may be due to geographical characteristics, subnational strata.
such as altitude, temperature, humidity, rainfall patterns, proximity
to water bodies, land use, vector species and distribution, socio- The Global Malaria Programme is working with countries to
demographic characteristics, access to antimalarial treatment, and strengthen the generation and use of local information for
coverage with vector control. In most endemic areas, seasonal stratification, the definition of optimal mixes of interventions, and
patterns of transmission are observed, with high transmission the rational, safe and ethical prioritization of resources to
during part of the year. Both the intensity and timing of maximize impact. Local data are also essential to understand the
transmission are important considerations in designing elimination impact of the strategies deployed, providing opportunities to
strategies. further refine sub-national strategies and inform global knowledge.
The manifestation of clinical disease depends strongly on the WHO also develops implementation guidance such as operational
background level of acquired protective immunity, which is a and field manuals to support the “how” aspect of delivering the
recommended tools and strategies. Operational manuals and other
35 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
guidance hold practical information for increasing the target every programme has to consider, global guidance is not intended
population's access to interventions. These documents are and should not be used to provide prescriptive guidance on what
referenced and linked to these Guidelines. The Global Malaria should be done in every situation. These Guidelines signal a
Programme is working to align this implementation guidance with paradigm shift towards a problem-solving approach using local
the recommendations in the WHO Guidelines for malaria. However, data to identify recommendations that are relevant at a country
where there are inconsistencies, the Guidelines should be the level and based on local context, defining stratum-
default resource for national decisions. Countries may use the specific packages of interventions that optimize impact and are
implementation guidance to define ways in which a prioritized for resource allocation. This shift moves away from
recommendation can be implemented effectively – for example, overly prescriptive recommendations and will clearly distinguish
intermittent preventive treatment for malaria in pregnancy could evidence-informed recommendations from contextual
be implemented through antenatal care and/or community considerations. The contextual considerations at national and
distribution. The intention of the guidance is to enable delivery, subnational levels will inform how recommendations should be
not to prescribe exactly how it should be done. applied and strategies that may increase access for the target
population.
Strategic information to tailor programmatic response and
selection of interventions Accurate stratification of malaria transmission intensity is essential
As malaria control improves, malaria transmission and risk become for effective targeting of interventions. As countries progress
increasingly heterogeneous, both between and within countries. towards elimination, finer scale mapping is required, and
Thus, a “one-size-fits all” approach to programme decisions on stratification should be more specific, ideally at the level of
intervention selection becomes inefficient. The situation requires localities or health facility catchment areas [13][14]. As
stratification of the country at subnational levels according to past, transmission intensity is progressively reduced, stratification needs
present and future malaria risk, the structure and function of the to include vulnerability and receptivity to malaria, i.e. the risk for
health system, and other contextual factors. Stratification provides importation of malaria cases and the inherent potential of the
a rational basis to identify context-specific packages of vector-human ecosystem to transmit malaria.
interventions to target specific populations in the different
subnational strata. Local data are essential to complete Conclusion
stratification and to inform the selection of the optimal mixes of These Guidelines provide a framework within which NMPs and
interventions to maximize impact. Given that resource constraints their implementing partners may adopt and adapt the
usually limit the implementation of all desirable interventions in all recommendations for use. Good quality surveillance data can also
areas of malaria risk, a prioritization exercise must also be feed into this process by providing the granular local information
conducted to ensure that resource allocation also optimizes needed to inform and evaluate national programme decisions (see
intervention mixes and resultant impact. Guidance on these Section 7: Surveillance). Where the boundaries of current
activities is available in Section 7: Surveillance. knowledge are pushed, it is particularly important to ensure
adequate attention to monitoring and evaluation. The information
The choice of interventions in each stratum should be informed by generated can then feed into updated guidance.
WHO’s recommendations. However, given the complexities of
malaria, with heterogeneity of risk and the unique contexts that
4. PREVENTION
Nearly half of the world’s population is at risk of malaria. In areas prevention tools and strategies – including effective vector control
with high malaria transmission, young children and pregnant and the use of preventive chemotherapies – has had a major
women are particularly vulnerable to malaria infection and death. impact in reducing the global burden of this disease.
Since 2000, expanded access to WHO-recommended malaria
36 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
continue to be pyrethroid-only long-lasting insecticidal nets It is important to note that while an individual species of
(LLINs); and ii) indoor residual spraying (IRS) with a product Anopheles will characteristically exhibit certain biting and resting
prequalified by WHO. Specific product choices within these behaviours, these are not absolute; subpopulations and
broad intervention types should be informed by insecticide individuals may exhibit different behaviours depending on a
resistance data for the target area(s) and other information combination of intrinsic genetic factors, availability of preferred
compiled during sub-national prioritization exercises. Once hosts and availability of suitable resting sites. Environmental and
optimal coverage with one of these interventions has been climatic factors, including rainfall, moonlight, wind speed, etc., as
achieved, supplementary interventions may be considered for well as the deployment of vector control interventions can all
deployment depending on the specifics of the population, influence biting and resting behaviours.
situation or setting. These include personal protection measures
that have a primary use-pattern of protecting individual users, Accurate species identification is crucial for all studies and
although they may have some as yet unproven impact when surveillance activities on field populations of vectors. Many of
deployed at the community level. the vectors belong to species complexes and require advanced
molecular analyses for species identification, necessitating
Vectors, their behaviour and distribution appropriate laboratory resources. Without accurate species
Malaria is transmitted through the bites of infective female identification, the data collected on behaviour, distribution and
Anopheles mosquitoes. Of the more than 400 different species of infection rates will have limited use for decision-making by
Anopheles mosquitoes, only around 40 are malaria vectors of control programmes.
major importance. Anopheles mosquitoes lay their eggs in water.
The eggs hatch to produce larvae, which undergo several moults Background and rationale for vector control
before emerging from the pupal stage as adult mosquitoes. The role of arthropods in the transmission of diseases to humans
Different species of Anopheles mosquitoes have their own was first elucidated in the late 19th and early 20th centuries.
preferred aquatic habitats; for example, some prefer small, Since effective vaccines or drugs were not always available for
shallow collections of fresh water such as puddles and animal the prevention or treatment of these diseases, control of
hoof prints, whereas others prefer large, open water bodies transmission often had to rely principally on control of the
including lakes, swamps and rice fields. vector. Early control activities included the screening of houses,
the use of mosquito nets, the drainage or filling of swamps and
Both male and female mosquitoes feed on plant nectar, but it is other water bodies used by insects for breeding, and the
just the female mosquitoes that feed on blood as they require application of oil or Paris green to breeding places. Following the
protein to develop their eggs. Different mosquito species discovery of the insecticidal properties of
demonstrate preferences for feeding on animals (zoophily) or on dichlorodiphenyltrichloroethane (DDT) in the 1940s and
humans (anthropophily); however, these preferences are not subsequent discovery of other insecticides, the focus of malaria
absolute, and females may take a blood meal from non-preferred vector control shifted to the deployment of insecticides to target
hosts when these are present in the area. Different hosts may be both the larval and adult stages of mosquito vectors.
more or less attractive to mosquitoes than others. Several
factors have been implicated in the attraction of female Nowadays, it is well established that effective vector control
mosquitoes to a host, including exhaled carbon dioxide, lactic programmes can make a major contribution to advancing human
acid, host odours, warmth and moisture. Blood-feeding can take and economic development. Aside from direct health benefits,
place inside human habitations (endophagy) or outdoors reductions in vector-borne diseases enable greater productivity
(exophagy), depending on the mosquito species. has implications and growth, reduce household poverty, increase equity and
for the selection and effectiveness of vector control women’s empowerment, and strengthen health systems [15].
interventions. Despite the clear evidence in broad support of vector control
efforts, the major vector-borne diseases combined still account
Female Anopheles mosquitoes blood feed predominantly at night, for around 17% of the estimated global burden of communicable
although some species may bite during the day in heavily shaded diseases, claiming more than 700 000 lives every year [16].
conditions, and some exhibit a peak in biting activity in the early Recognizing the great potential to enhance efforts in this area,
evening or early morning. The blood-feeding preferences WHO led the development of the Global vector control response
(zoophily/anthropophily, endophagy/exophagy) as well as the 2017–2030 [16], which is outlined in the subsequent section.
interplay between the peak biting time of Anopheles vectors and
the activity and sleeping patterns of the human hosts has Between 2000 and 2015, the infection prevalence of
important consequences for malaria transmission and the choice Plasmodium falciparum in endemic Africa was halved and the
of appropriate vector control interventions. incidence of clinical disease fell by 40% [17]. Malaria control
interventions averted an estimated 663 million (credible interval
After blood-feeding, female mosquitoes rest in order to digest (CI) 542–753 million) clinical cases in Africa, with ITNs making
the blood meal and mature their eggs. Female mosquitoes may the largest contribution (68% of cases averted). Indoor residual
rest indoors (endophily) or outdoors (exophily), and this depends spraying (IRS) contributed an estimated 13% (11–16%), with a
on innate species preferences as well as the availability of larger proportional contribution where intervention coverage
suitable resting sites in the local environment. The mosquitoes’ was high [17].
choice of post-feeding resting site also has major implications for
the selection of control interventions. Global vector control response 2017–2030
37 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The vision of WHO and the broader infectious diseases should be implemented early to prevent the spread and increase
community is a world free of human suffering from vector-borne in the intensity of resistance. The overarching concepts of such
diseases. In 2017, the World Health Assembly welcomed the resistance management strategies were outlined in the Global
Global vector control response 2017–2030 [16] (GVCR) and plan for insecticide resistance management in malaria vectors
adopted a resolution to promote an integrated approach to the (GPIRM) in 2012 [21].
control of vector-borne diseases. The approach builds on the
concept of integrated vector management (IVM), but with Guidance on monitoring of insecticide resistance, interpretation
renewed focus on improved human capacity, strengthened of test results and implications for decision-making are given in
infrastructure and systems, improved surveillance, and better the WHO Test procedures for monitoring insecticide resistance in
coordination and integrated action across sectors and diseases. malaria vector mosquitoes [22] and in the Framework for a national
Development programmes, including, for example, irrigated plan for monitoring and the management of insecticide resistance in
agriculture, hydroelectric dam construction, road building, forest malaria vectors [23]. When deciding whether adjustments to the
clearance, housing development and industrial expansion, all national malaria strategic plan are required in a given area, at
have the potential to influence vector-borne diseases, offering least the following must be considered for that locality:
the opportunity for intersectoral collaboration and the adoption
of strategies other than those based on insecticides. • current and past transmission levels;
• current and past interventions deployed, including the
The ultimate aim of the GVCR is to reduce the burden and threat coverage, usage and duration of efficacy;
of vector-borne diseases through effective, locally adapted, • the insecticide resistance profile of the main vector species
sustainable vector control in full alignment with Sustainable (including resistance intensity and resistance mechanisms);
Development Goal 3.3: to end epidemics of malaria by 2030. and
• other entomological information including vector species
Effective and sustainable vector control is achievable only with distribution, abundance and other bionomic data.
sufficient human resources, an enabling infrastructure and a
functional health system. As recommended under the GVCR,
The susceptibility of mosquitoes to insecticides and
national programmes should lead a vector control needs
determination of the species-specific presence, intensity and
assessment across the relevant sectors [18] to help appraise
mechanisms of resistance in vector populations can be used to
current capacity, define the requisite capacity to conduct
guide the selection of the most appropriate insecticidal products
proposed activities, identify opportunities for improved
to deploy. Generally, if mosquitoes are found to be resistant to
efficiency in vector control delivery, and guide resource
an insecticide, insecticides with a different mode of action
mobilization to implement the national strategic plan.
should be deployed. However, there are reports of mosquitoes
having differential susceptibility to insecticides within the same
Prevention, mitigation and management of insecticide
class, and questions have been raised about the level of cross-
resistance
resistance between pyrethroid products [21]. The Global Fund to
Widespread and increasing insecticide resistance poses a threat
Fight AIDS, Tuberculosis and Malaria recently commissioned a
to effective malaria vector control. Failure to mitigate and
review of the interpretation of insecticide resistance assays
manage insecticide resistance is likely to result in an increased
when selecting insecticidal products [24].The review aimed to
burden of disease, potentially reversing some of the substantial
answer the question: In areas where pyrethroid resistance exists,
gains made in controlling malaria over the last decade.
but mosquitoes of the same population differ in their
WHO maintains a global insecticide resistance database and an susceptibility to different pyrethroids, should programmes
online mapping tool that consolidate information on the status consider selecting one pyrethroid over another in order to
of the insecticide susceptibility of Anopheles mosquitoes in manage insecticide resistance? Based on a review of evidence
malaria-endemic countries. The latest data reveal that almost from molecular, laboratory and field data, the authors concluded
90% of the malaria-endemic countries reporting insecticide that differences between adult mosquito mortalities in
resistance have detected resistance of their vectors to at least pyrethroid insecticide resistance assays are not indicative of a
one insecticide class. Globally, resistance to pyrethroids is true or operationally relevant difference in the potential
widespread, having been detected in at least one malaria vector performance of pyrethroids currently in common use
in 68% of the sites for which data were available. Resistance to (deltamethrin, permethrin, α-cypermethrin and λ-cyhalothrin).
organochlorines was reported in 64% of the sites. Resistance to Consequently, switching between pyrethroid insecticides (to
carbamates and organophosphates was less prevalent, detected improve intervention efficacy) should not be used as a means of
in 34% and 28% of the sites that reported monitoring data, managing insecticide resistance. This finding supports WHO’s
respectively [4]. past and present position. Given that pyrethroid resistance in
mosquitoes is widespread, WHO encourages the development
To date, there is no evidence of operational failure of vector and continued evaluation of nets treated with alternative
control programmes as a direct result of increasing frequency of insecticides [25].
pyrethroid resistance [19][20]. Based on past experience,
however, it is likely that operational failure will eventually occur Key technical principles for addressing insecticide resistance are
if effective insecticide resistance management (IRM) strategies as follows:
are not designed and implemented. Ideally, such strategies
38 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• Insecticides should be deployed with care and deliberation mixture should have a similar residual life and remain
in order to reduce unnecessary selection pressure and bioavailable over time; in practice, this is difficult to achieve,
maximize impact on disease. National malaria programmes particularly for vector control products that are meant to
(NMPs) should consider whether they are using insecticides last for a number of years, such as long-lasting insecticidal
judiciously, carefully and with discrimination, and if there is nets (LLINs). An ITN product containing a pyrethroid and a
a clear epidemiological benefit. pyrrole insecticide and another containing a pyrethroid and
• Vector control programmes should avoid using a single class a juvenile hormone mimic have been developed and
of insecticide everywhere and over consecutive years. prequalified by WHO. Trials are ongoing to assess the
Whenever possible, vector control programmes should epidemiological impact of these products after which their
diversify from pyrethroids to preserve their effectiveness. public health value will be assessed in order to develop any
Although pyrethroids will continue to be used for ITNs in WHO recommendation. A mixture of a pyrethroid and a
the near term, they should not generally be deployed for neonicotinoid insecticide for IRS has been prequalified by
IRS in areas with pyrethroid ITNs, whether alone or WHO.
combined with insecticides from a different class.
• Rotations involve switching between insecticides with
• IRM principles and methods should be incorporated into all
different modes of action at pre-set time intervals,
vector control programmes, not as an option, but as a core
irrespective of resistance frequencies. The theory is that
component of programme design.
resistance frequencies will decline (or at least not increase)
• NMPs should engage with the agricultural sector to
during the period of non-deployment of insecticides with a
coordinate insecticide use, with the aim of avoiding use of
specific mode of action.
the same classes of insecticide for both crop protection and
• Mosaics involve the deployment of insecticides with
public health within the same geographical area.
different modes of action in neighbouring geographical
• Routine monitoring of insecticide resistance is essential to
areas. The optimal spatial scale (size of areas) for mosaics
inform the selection and deployment of insecticides.
has yet to be determined, and rotations are generally
• The additional costs of deploying new vector control tools
considered to be more practical and feasible.
as part of a comprehensive IRM response should be
• Combinations expose the vector population to two classes
balanced against the potential long-term public health
of insecticides with differing modes of action through the
impact. Where feasible, formal economic evaluation is
co-deployment of different interventions in the same place,
encouraged to investigate the likely incremental costs and
such as ITNs co-deployed with non-pyrethroid IRS (where
effectiveness of potential IRM approaches, relative to
both are at high coverage; see recommendation under
feasible alternatives, for a given context.
section 4.1.2).
Approaches
For malaria vector control, however, there is still little evidence
Historically, the most common way insecticides have been
of the success of these strategies and no consensus on the best
deployed to control malaria vectors has been through
IRM approach or approaches to apply in a given situation.
“sequential use”. In essence, this is when a single insecticide class
Success of a particular approach will likely depend on mosquito
is used continuously or repeatedly until resistance has rendered
genetics, behaviour and population dynamics, and the chemical
it less effective or ineffective, after which a switch is made to an
nature of the insecticides and their formulation. A 2013 review
insecticide with a different mode of action to which there is no
of experimental and modelling studies on insecticide, pesticide
(or less) resistance. In theory, this may allow for an eventual
and drug resistance concluded that mixtures generally lead to
switch back to the original insecticide class if resistance
the slowest evolution of resistance [26]. However, more recently,
decreases to the point that it is no longer detectable by means
an exploration of overlaps between agriculture and public health
of bioassays.
found that – owing to caveats and case specificity – there is only
The agricultural industry has had some success in managing weak evidence of one IRM approach being better than another,
resistance by using different insecticides over space and time. and that the standard practice of using insecticides until
Similar approaches have been proposed with the aim of resistance emerges before switching to an alternative (i.e.
preventing or delaying the spread and increase of resistance by sequential use) may be equally effective under certain
removing selection pressure or by killing resistant mosquitoes. circumstances. More data, both from research and programmatic
These strategies include mixtures of insecticides, mosaic operations, are needed to compare resistance management
spraying, rotations of insecticides and deployment of multiple approaches in the field [27] and to improve understanding of the
interventions in combination. biological mechanisms that are likely to favour different
approaches in different situations [28][29].
• Mixtures are co-formulations that combine two or more
insecticides with different modes of action. Effective Evidence-based planning
deployment of a mixture requires the presence of resistance To achieve optimal impact against malaria, control measures
to all insecticides in the mixture to be rare, so that any must be suitable for the geographic area (based on vector
individual mosquito that survives exposure to one bionomics) and, well targeted and deployed at sufficient
insecticide is highly likely to be killed by the other coverage. Without an evidence base or sufficient capacity to
insecticide or insecticides. Ideally, all insecticides in a deploy interventions appropriately, resources may be used
39 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
suboptimally. Given the heavy reliance on insecticidal forest areas or do not sleep in protected houses, or when local
interventions – primarily ITNs and IRS – the impacts on the mosquito vector species exhibit one or more behaviours that
environment and insecticide resistance of local vectors are key enable them to avoid vector control interventions, such as biting
considerations in vector control planning and implementation. outside early in the evening before people have retired indoors
The inappropriate deployment of insecticides both in agriculture and/or resting outdoors. The sources and risk of residual
and in public health programmes has the potential to result in transmission may, therefore, vary by location, time and the
avoidable insecticide contamination of the environment and/or existing components of the current malaria programme.
development of insecticide resistance of local vectors. Ideally,
IRM practices should be implemented as part of routine In some settings, supplementary interventions may be used in
operations, rather than waiting for resistance to spread or addition to ITNs or IRS to further reduce transmission.
increase and for control failure to be suspected or confirmed. A Recommendations on larviciding with chemical or biological
pragmatic approach must be taken that seeks to select insecticides and the use of house screening are outlined in a
appropriate vector control interventions based on the insecticide subsequent chapter. Supplementary interventions should be
resistance profile of the major malaria vectors in the target area. implemented in accordance with the principles outlined in
To outline how resistance will be monitored and managed, NMPs the Global vector control response 2017–2030 [16].
should develop and implement national plans in accordance with
Residual transmission can be difficult to measure, as is the
the WHO Framework for a national plan for monitoring and
specific impact of supplementary tools on this component of
management of insecticide resistance in malaria vectors [23].
ongoing transmission. Standardized methods for quantifying and
Detailed information on insecticide resistance monitoring
characterizing this component of transmission are required in
methods and on how to use the data to inform the selection of
order to evaluate the effectiveness of single or combined
appropriate interventions will be provided in the revised WHO
interventions in addressing this biological challenge to malaria
Test procedures of monitoring insecticide resistance in malaria
prevention, control and elimination.
vectors, anticipated to be published in 2022. Further information
on insecticide resistance monitoring and, more broadly, on There is an urgent need for greatly improved knowledge of the
entomological surveillance is included in the WHO Malaria bionomics of the mosquitoes responsible for maintaining local
surveillance, monitoring & evaluation: a reference manual, which transmission. New interventions and strategies should be
outlines priority data across different transmission settings [30]. evaluated against these vectors in order to effectively address
residual transmission. While this knowledge is being gained and
IRM plans should be revisited regularly to consider new interventions are being developed, NMPs must prioritize the
information, and to integrate new interventions once they have effective implementation of current interventions to reduce
been supported by WHO recommendations and prequalified. transmission to the lowest level possible. At the same time, they
should collaborate with academic or research institutions to
Vector control across different malaria transmission settings generate local evidence on the magnitude of the problem of
Access to effective vector control interventions will need to be residual transmission of malaria, including information on human
maintained in the majority of countries and locations where and vector behaviours, and the effectiveness of existing and
malaria control has been effective. This includes settings with novel interventions.
ongoing malaria transmission, as well as those in which
transmission has been interrupted but in which some level of Acceptability, participation and ethical considerations
receptivity and vulnerability remains. Malaria elimination is Community participation in the implementation of vector control
defined as the interruption of local transmission (reduction to interventions often takes the form of “instruction” or
zero incidence of indigenous cases) of a specified malaria “information”, with decisions about the need for interventions
parasite species in a defined geographical area as a result of being made at international and national levels. Taking into
deliberate intervention activities. Following elimination, account communities’ views on the recommended interventions
continued measures to prevent re-establishment of transmission may promote acceptance and adherence to the intervention.
are usually required [30]. Interventions are no longer required Increased levels of participation (e.g. consultation, inclusion and
once eradication has been achieved. Malaria eradication is shared decision-making) should be included in the development
defined as the permanent reduction to zero of the worldwide and deployment of vector control interventions – from inception
incidence of infection caused by all human malaria parasite through to the planning and implementation stages.
species as a result of deliberate activities.
WHO acknowledges that appropriate policy-making often
requires explicit consideration of ethical matters in addition to
Residual transmission
scientific evidence. However, the ethical issues relevant to
WHO acknowledges that malaria can persist despite high
vector-borne disease control and research have not received the
coverage of antimalarial interventions, including in areas with
analysis necessary to further improve public health programmes.
optimal access to and use of ITNs or with high IRS coverage [31].
Moreover, WHO Member States lack specific guidance in this
This persistence of malaria transmission following the
area. The Seventieth World Health Assembly [32] requested the
implementation in time and space of a widely effective malaria
Director-General “to review and provide technical guidance on
programme is referred to as residual transmission. Residual
the ethical aspects and issues associated with the
transmission occurs as a result of a combination of human and
implementation of new vector control approaches in order to
vector behaviours, for example, when people reside in or visit
develop mitigating strategies and solutions; and to undertake a
40 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
review of the ethical aspects and related issues associated with The systematic review of the cost and cost-effectiveness of
vector control implementation that include social determinants vector control interventions that was used to inform the current
of health, in order to develop mitigating strategies and solutions vector control guidelines was published in 2021, as part of a
to tackle health inequities.” A scoping meeting was convened by broader systematic review on the cost and cost-effectiveness of
WHO to identify the ethical issues associated with vector-borne malaria control interventions, drawing on evidence published
diseases [33]. Unique ethical issues associated with vector between 2005 and 2018 [35]. The body of evidence on vector
control that were identified include the ethics of coercive or control interventions was based on the use of ITNs/LLINs, IRS
mandated vector control, the deployment of insecticides (and and larval source management (LSM) mostly in sub-Saharan
growing vector resistance to insecticides), and research on and/ African countries. The review reported that, overall, WHO-
or deployment of new vector control technologies. Genetically recommended malaria interventions including vector control
modified mosquitoes are one such innovation that presents represent value for money; however, there was great variation in
potential challenges, including how to prevent their spread the costs of intervention delivery, reflecting not only differences
beyond the intended geographical target areas and limit in the actual resource use, but also the various types of costing
potential effects on the local fauna. In 2020 WHO published methodologies employed. The available cost and cost-
guidance on vector-borne disease and ethical effectiveness data focused largely on individual interventions
considerations [34]. Work is continuing the develop guidance in and less so on packages of interventions, which are
this area. recommended for effective malaria control. The authors
reported that, due to the heterogeneity of the study contexts
Equity, gender and human rights and the way data were presented, comparative analysis of the
WHO advocates for optimal coverage with recommended vector cost-effectiveness of interventions was not possible.
control interventions. As such, malaria vector control should be
implemented without discrimination on the basis of age, sex, The WHO Global Malaria Programme is working with partners
ethnicity, religion or other characteristics. In some cases, special to update the evidence review on the cost and cost-
effort is required to reach populations that are geographically effectiveness of the vector control interventions covered in the
isolated or adopt a nomadic lifestyle. Guidelines to support future Guideline development
deliberations, for example, by building and updating a database
Resource implications and prioritization for the cost and cost-effectiveness of vector control and other
In the Guidelines, resource implications and the cost- malaria interventions. It is also planned that systematic reviews
effectiveness of vector control interventions have been largely commissioned in the future will include a search of the literature
addressed by drawing on a recent systematic review of the cost on both the cost and cost-effectiveness of interventions under
and cost-effectiveness of vector control interventions [35] and consideration as well as those previously approved.
expert opinion within the GDG.
41 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
goal of providing full population coverage. protection to the community as a whole beyond the individual
protection provided by ITNs.
Insecticide-treated nets
For the ITN classes covered by WHO recommendations as Large-scale field trials [40][44] and transmission
interventions for use in protecting populations at risk of models [45][46] originally suggested that community coverage
malaria, including in areas where malaria has been eliminated (i.e. the proportion of human population using an ITN with
but the risk of reintroduction remains, WHO recommends effective insecticide treatments each night) of ≥ 50% is
products that have been prequalifed by WHO. WHO Member expected to result in some level of community-wide
States and their procurement partners are encouraged to draw protection. The WHO-commissioned review indicated that this
on the list of prequalified products to inform their choice of area-wide protection may start to occur at lower coverage
product(s). levels (Lines et al unpublished evidence). The review modelled
the short-term effect of increasing ITN coverage on the EIR
An ITN may repel, disable and/or impact the fecundity of (infectious bites per person per year) in an area with high
mosquitoes that come into contact with the insecticide on the malaria transmission and an insecticide-susceptible,
netting material in addition to providing a physical barrier, anthropophilic vector, assuming fixed human infectiousness. In
thereby protecting the individual user. In addition, some the coverage range of 15% to 85%, an additional 20% increase
studies have indicated that ITNs produce a “community effect”, in coverage of the human population at risk was shown to
which means that when enough ITNs are being used in a result in a reduction in malaria transmission intensity of
community, the survival of the mosquito population as a whole approximately 50% (these findings are taken from the report
is affected; this effect increases the protection against malaria submitted to WHO; findings may be revised if indicated by
for ITN users and extends protection to members of the peer review). Additional ITN coverage is always beneficial in
community who do not sleep under an terms of providing more protection to individuals – both users
ITN [36][37][38][39][40]. However, such a community effect and non-users of ITNs – and, conversely, any reduction in
has not been observed in all settings [41][42][43]. The WHO coverage may result in increased malaria transmission.
Global Malaria Programme commissioned a review to examine However, there may be diminishing marginal returns to
the evidence for a community effect and to investigate the increasing coverage at higher levels. In terms of absolute cases
biological mechanisms by which ITNs provide both personal- of malaria averted, a reduction in malaria transmission when
and community-level protection against malaria. The review increasing ITN coverage from 80% to 100% may not generate
also investigated what factors may determine the presence of the same impact as a 20% increase in coverage at lower levels
a community effect and moderate its intensity (Lines et al of coverage; the marginal costs required to increase coverage
unpublished evidence). at high levels (>80%) will also increase due to growing system
inefficiencies. At the country level, these diminishing returns
The review concluded that a community effect does occur in must be balanced against potential investments in other cost-
the majority of settings, and that its extent is driven by a effective malaria prevention and control activities by means of
number of contextual factors. These factors include vector a well-informed prioritization process.
behaviour (particularly the extent of anthropophily, i.e. the
propensity to feed on people, and endophagy, i.e. the Three main ITN classes are recognized by WHO as given
tendency of mosquitoes to blood-feed indoors); the relative below. With the March 2023 update to the guidelines, these
availability of human and non-human hosts in the locality; the classes are now formally:
level of ITN coverage and use in a community; the insecticide
used (its residual insecticidal activity and repellency); and the • ITNs designed to kill host-seeking insecticide-susceptible
resistance of the local malaria vectors, both physiological and mosquito populations that have demonstrated
behavioural, to the insecticide on the net. public health value compared to untreated nets and
whose entomological effects consist of killing and
The ITN coverage threshold for when the community effect reducing the blood-feeding of insecticide-susceptible
becomes apparent depends on a large number of contextual mosquito vectors. This intervention class covers
factors. Regardless of the context-dependent starting pyrethroid-only nets prequalified by WHO and
threshold, the extent of the community-level protection conventionally treated nets that rely on periodic re-
increases as ITN coverage and net use in a given community treatment with a WHO prequalified self-treatment kit.
increases. Because ITNs kill insecticide-susceptible mosquitoes Public health value has been demonstrated for products
that come into contact with the insecticide on the netting within this class and WHO recommends use of
material, more mosquitoes will be killed as ITN coverage pyrethroid-only LLINs prequalified by WHO for large-
increases. This killing effect reduces both mosquito population scale deployment.
density and mosquito longevity, resulting in fewer malaria • ITNs designed to kill host-seeking insecticide-resistant
vectors overall and a lower infectivity rate as fewer mosquitoes and for which a first-in-class product
mosquitoes will survive the time it takes for the malaria demonstrates public health value compared to the
parasite to develop in the mosquito. Consequently, the epidemiological impact of pyrethroid-only nets. This class
reduced density, age and proportion of the local mosquito includes nets that are treated with a pyrethroid insecticide
population that is infective offer an additional level of and a synergist such as piperonyl butoxide (PBO) and nets
42 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
treated with insecticides other than pyrethroid-based studies that have been carried out in the chronic phase of
formulations. Public health value has been demonstrated emergencies [48][49][50][51][52][53][54] (Messenger et al
for this class and WHO has issued recommendations for unpublished evidence). However, deployment of such
deployment of pyrethroid-PBO nets and for pyrethroid- interventions may be logistically challenging during the acute
chlorfenapyr nets in areas with pyrethroid-resistant phase of a humanitarian emergency. In the following sections,
mosquitoes. recommendations regarding the deployment of ITNs and
• ITNs designed to sterilize and/or reduce the fecundity of IRS are provided.
host-seeking insecticide-resistant mosquitoes for which a
first-in-class product demonstrates public health value Some vector control interventions and personal protection
compared to the epidemiological impact of pyrethroid- measures have been specifically designed for deployment in
only nets. Nets treated with pyrethroid + pyriproxyfen (an emergency situations. Such interventions include insecticide-
insect growth regulator), which fall into this class, are now treated plastic sheeting (ITPS), which can be used to construct
conditionally recommended for deployment instead of temporary shelters; insecticide-impregnated blankets or
pyrethroid-only LLINs. topsheets, which may be included in emergency relief kits
provided at the outset of an emergency; repellents; and
treating cattle with insecticides. For all of these interventions,
ITNs are most effective where the principal malaria vector(s) a limited number of studies have evaluated their efficacy in
mosquitoes bite predominantly at night after people have humanitarian emergencies [54] (Messenger et al unpublished
retired under their nets. ITNs can be used both indoors and evidence) and, as such, the evidence base on the effectiveness
outdoors, wherever they can be suitably hung (although of these interventions against malaria is currently insufficient
hanging nets in direct sunlight should be avoided, as sunlight to formulate recommendations.
can affect insecticidal activity).
As in more stable settings, the appropriateness and
Indoor residual spraying effectiveness of vector control in humanitarian emergencies
IRS is the application of a residual insecticide to potential will depend on:
malaria vector resting surfaces, such as internal walls, eaves
• the malaria infection risk;
and ceilings of houses or structures (including domestic animal
• the behaviour of the human population (e.g. mobility,
shelters), where such vectors are likely to come into contact
where they are sleeping or being exposed to vector
with the insecticide. IRS with a WHO-prequalified product is
mosquitoes); and
recommended for large-scale deployment in most malaria-
• the behaviours of the local vector population (e.g. indoor
endemic locations. IRS is most effective where the vector
resting, indoor biting, early evening or night biting).
population is susceptible to the insecticide(s) being applied,
where the majority of mosquitoes feed and rest indoors, and
where most structures are suitable for spraying. In deciding
whether to deploy IRS, programmes should consider whether In humanitarian emergencies, further consideration must be
achieving the target coverage of IRS is feasible. given to whether the delivery of vector control interventions is
feasible. This may depend on:
Humanitarian emergencies
The first priorities for malaria control in a humanitarian • the type of shelter available (e.g. ad hoc refuse materials,
emergency are prompt and effective diagnosis and plastic sheeting, tents, more permanent housing); and
treatment [47]. Deployment of ITNs and IRS have been shown • the available infrastructure, resources and human capacity
to provide protection against malaria in the limited number of to deliver vector control.
Pyrethroid-only long-lasting insecticidal nets (LLINs) should be deployed for the prevention and control of malaria in
children and adults living in areas with ongoing malaria transmission.
• WHO recommends ITNs that have been prequalified by WHO for deployment in protecting populations at risk of malaria.
• ITNs are most effective where the principal malaria vector(s) bite predominantly at night after people have retired under their
nets.
• ITNs can be used both indoors and outdoors, wherever they can be suitably hung (though hanging nets in direct sunlight
should be avoided, as sunlight can affect insecticidal activity).
Practical Info
The current WHO recommendation for ITNs applies only to those mosquito nets that have been prequalified by WHO and
43 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
that contain only an insecticide of the pyrethroid class (categorized as ‘pyrethroid-only LLINs’).
As with all insecticide-based interventions, the insecticide resistance profile of the vectors within the area of deployment
should be assessed. If pyrethroid-resistance is detected, pyrethroid-PBO ITN or pyrethroid-chlorfenapyr ITNs should be
considered for distribution, and pyrethroid-pyrproxyfen ITNs may be considered, instead of pyrethroid-only nets (see the
following recommendations on the other types of nets).
ITNs are generally acceptable to most communities. In many malaria-endemic countries, untreated nets were in use for many
years prior to the introduction of ITNs and, even where there is not a long history of their use, they have become familiar
tools for preventing mosquito bites. Individuals often appreciate the extra privacy afforded by a net, as well as its
effectiveness in controlling other nuisance insects. In very hot climates, ITNs may be less acceptable, as they are perceived
to reduce air flow, making it too hot to allow for a comfortable sleep. In areas where mosquito densities are low or where
malaria transmission is low, individuals and communities may perceive less benefit to using nets.
When deploying ITNs, coverage must be optimized such that both personal and community-level effects are maximized and
maintained in endemic settings. Post-distribution monitoring of nets is essential, reporting their durability, usage and
coverage. Evaluation of the impact on vectors, such as their abundance, EIR and behaviour, and insecticide resistance status
can be used to inform and guide future deployment.
Nets should be handled and disposed of appropriately to minimize risk to human and animal health and of environmental
contamination. WHO recommends that old nets are not burned in the open air but are buried, preferably in non-permeable
soil and away from water sources. Burning may lead to the release of dioxins, which are harmful to human health. The
insecticides used on nets are toxic to aquatic organisms and so should not be disposed of in water.
Evidence To Decision
No undesirable effects were identified in the systematic review. However, the panel noted that brand new nets recently
removed from packaging may cause slight, transitory irritation to skin, eyes, nose, etc. Some users complain that the nets
are too hot to sleep under, especially during the warmer seasons. As with any insecticide-based intervention, ITNs may
also play a role in insecticide resistance development in Anopheles vectors, and there is a risk of environmental
contamination with potential toxic effects on animals if nets are not handled or disposed of carefully (see section on
Practical Info).
The systematic review determined that, overall, the evidence that ITNs have an impact on malaria was high compared to
no nets and compared to untreated nets.
44 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
communication [BCC])
• Teachers/health facility staff, where appropriate, trained for distribution channel
• Entomologists for quality control (QC) assessments
• Environmental assessment support staff
• Shipping of ITNs may require large trucks for transport of containerized nets from
port of entry to centralized warehouses and onward to the district or other level.
• Vehicles to provide transport of ITNs and potentially distributors to the
Transport community (last mile) to enumerate persons/households, provide BCC and
distribute ITNs
• Vehicle maintenance costs
• Fuel
• ITNs
• Inventory management forms
• Recipient lists, distribution forms, including recipient sign-off sheets, daily
Supplies
distribution reports, inventory status reports, recipient status reports, and BCC
materials (e.g. flip charts, posters, banners, staff clothing)
• M&E data collection forms
• ITN quality/durability assessment materials – e.g. cone bioassay material
• Distribution supervisors
Governance/ programme • BCC supervision
management • M&E survey support for assessing coverage and use
• QC supervision
Justification
The systematic review [55] followed the original 2003 analysis, which included insecticide-treated curtains and ITNs
together and included two studies solely evaluating insecticide-treated curtains and one study evaluating both ITNs and
insecticide-treated curtains. There was no obvious heterogeneity that would lead to a subgroup analysis to examine whether
the effects were different, and the results from studies evaluating insecticide-treated curtains were consistent with the
results of those evaluating ITNs. The GDG drew on the analysis to make recommendations related to ITNs only.
The systematic review [55] reported high-certainty evidence that, compared to no nets, ITNs are effective at reducing the
rate of all-cause child mortality, the rate of uncomplicated episodes of P. falciparum, the incidence rate of severe malaria
episodes, and the prevalence of P. falciparum. ITNs may also reduce the prevalence of P. vivax, but here the evidence of an
45 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Compared to untreated nets, there was high certainty evidence that ITNs reduce the rate of uncomplicated episodes of P.
falciparum and reduce the prevalence of P. falciparum. There was moderate certainty evidence that ITNs also reduce all-cause
child mortality compared to untreated nets. The effects on the incidence of uncomplicated P. vivax episodes and P. vivax
prevalence were less clear.
The systematic review did not identify any undesirable effects of pyrethroid ITNs.
Research Needs
• Determine the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria infection), as well as
potential harms and/or unintended consequences of new types of nets and insecticides in areas where resistance to
pyrethroids is high.
• Determine the comparative effectiveness and durability of different pyrethroid-only net types.
• Determine the effectiveness of nets in situations of residual/outdoor transmission.
• Determine the impact of ITNs in transmission ‘hotspots’ and elimination settings.
Pyrethroid-PBO ITNs instead of pyrethroid-only LLINs can be deployed for the prevention and control of malaria in children
and adults in areas with ongoing malaria transmission where the principal malaria vector(s) exhibit pyrethroid resistance.
The conditionality of this recommendation is largely driven by the current higher unit cost of pyrethroid-PBO ITNs compared to
pyrethroid-only LLINs and therefore the uncertainty of their cost-effectiveness. Furthermore, as PBO is less wash-resistant than
pyrethroids, its bioavailability declines faster over the three-year estimated life of an ITN; therefore, the added impact of
pyrethroid-PBO ITNs over that of pyrethroid-only LLINs may decline over time. The evidence comes from two sites in eastern Africa
with pyrethroid resistance and not from other geographies where transmission levels and vector characteristics may vary. PBO acts
by inhibiting certain metabolic enzymes, primarily oxidases, and so are likely to provide greater protection than pyrethroid-only
LLINs where mosquitoes display mono-oxygenase-based insecticide resistance mechanisms.
In deciding whether pyrethroid-PBO ITNs may be appropriate in their context, malaria programmes should:
• consider the deployment of pyrethroid-PBO ITNs in areas where resistance to pyrethroids in local vectors has been detected;
• determine whether resources are adequate to cover the extra cost of pyrethroid-PBO ITNs, while ensuring that coverage of
populations at risk of malaria is not affected;
• note that WHO recommends that ITNs prequalified by WHO be selected for deployment.
Practical Info
Given that the evidence indicates that unwashed pyrethroid-PBO ITNs are more effective than pyrethroid-only LLINs in
areas with pyrethroid resistance up to 25 months post-deployment, the decision on whether to switch from pyrethroid-only
LLINs to pyrethroid-PBO ITNs, or another ITN product designed to provide enhanced efficacy in areas of pyrethroid
resistance, should be guided by resource availability. WHO recommends that pyrethroid-PBO ITNs be used where
pyrethroid resistance is confirmed using standard procedures [22]. Given that pyrethroid-PBO nets are designed to provide
improved impact against resistant mosquitoes in which pyrethroid resistance is, at least in part, conferred by a
monooxygenase-based resistance mechanism, determining the presence of such resistance mechanisms in local vector
populations will provide additional information to help target deployment.
In deciding whether to use potentially more expensive pyrethroid-PBO ITNs, malaria programmes should consider the
impact this switch may have on vector control coverage. Deployment of pyrethroid-PBO ITNs must only be considered in
situations where coverage with effective vector control (primarily ITNs or IRS) will not be reduced. The primary goal must be
to ensure continued access and use of ITNs or IRS at levels that ensure optimal coverage for all people at risk of malaria as
part of an intervention package. Post-distribution monitoring of nets to estimate coverage in terms of access to and use of
nets and other malaria interventions is recommended.
Pyrethroid-PBO ITNs should not be considered a tool that can alone effectively manage insecticide resistance in malaria
vectors. Despite the recent recommendation of other ITN classes and associate product, the development and evaluation of
46 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
ITNs treated with non-pyrethroid insecticides and other innovative vector control interventions for deployment across all
settings continues to remain a priority to provide alternatives for use in a comprehensive IRM strategy.
The systematic review reported that the washing of pyrethroid-PBO ITNs may result in lower mosquito mortality and higher
blood-feeding success than the washing of pyrethroid-only LLINs. The durability of pyrethroid-PBO ITNs compared to
pyrethroid-only LLINs has been questioned previously based on wash-resistance data. The added epidemiological and
entomological impact of pyrethroid-PBO ITNs depends on the bioavailability and retention of PBO on/in the net. If this is
reduced significantly over time and/or declines with washing, the greater impact of pyrethroid-PBO ITNs over pyrethroid-
only LLINs in terms of protection against malaria may be limited to less than three years. In addition, at present, it is
unknown how differences in the design/composition of pyrethroid-PBO ITNs affect their relative efficacy. A series of
experimental hut trials with entomological end-points using non-inferiority designs have recently been completed with as a
means to provide clarity in this respect [56]. As part of M&E activities, data collected by programmes on net durability would
provide information on the life span of pyrethroid-PBO ITNs under field conditions and hence on the period over which the
additional impact is maintained.
Programmes that decide to switch from pyrethroid-only LLINs to pyrethroid-PBO ITNs based on concerns regarding
continued effectiveness and/or insecticide resistance status of local vectors, should not revert back to the use of pyrethroid-
only LLINs thereafter. Instead, programmes should plan for continued deployment of pyrethroid-PBO ITNs in that
geographic area or develop plans for deployment of other equally or more effective new interventions once these are
covered by a WHO recommendation.
Evidence To Decision
The review also reported entomological outcomes, mosquito mortality and mosquito blood-feeding success derived
from experimental hut studies. In areas classified by the authors as having highly pyrethroid-resistant mosquitoes,
unwashed pyrethroid-PBO ITNs were found to result in higher mosquito mortality and lower blood-feeding success
compared to unwashed pyrethroid-only LLINs. Comparing washed pyrethroid-PBO ITNs to washed pyrethroid-only
LLINs, however, the review reported that it was unclear whether the washed pyrethroid-PBO ITNs had a greater effect
on mosquito mortality, although the washed pyrethroid-PBO ITNs did decrease the blood-feeding success of
mosquitoes.
In areas defined as having moderate, low (defined by the review team as 31–60% and 61–90% mosquito mortality,
respectively, in discriminating dose assays) or no pyrethroid insecticide resistance, the review did not identify any
studies with epidemiological outcomes. Regarding entomological outcomes, mosquito mortality was only shown to be
higher with unwashed pyrethroid-PBO ITNs compared to unwashed pyrethroid-only LLINs in those areas with moderate
insecticide resistance. Little or no difference was seen in terms of mosquito mortality or blood-feeding rates when
washed or unwashed pyrethroid-PBO ITNs were used in areas with low or no resistance compared to pyrethroid-only
LLINs.
Given that the systematic review was limited to two studies with malaria outcomes, a number of potential effect
modifiers could not be examined. However, as with pyrethroid-only LLINs, the GDG concluded that the extent of the
impact of pyrethroid-PBO ITNs is likely to vary in different settings and will depend on a number of factors, such as the
behaviour of the main malaria vectors and their level and mechanism(s) of insecticide resistance, the parasite prevalence
in that area, and the usage of nets within a community.
The systematic review did not report any harms or unintended consequences of the intervention. However, the GDG
noted that, compared to pyrethroid-only LLINs, pyrethroid-PBO ITNs may play an as yet unknown role in the
development of insecticide resistance in Anopheles mosquito vectors, such as increasing selection pressure for non-
oxygenase resistance mechanisms or perhaps increasing the intensity of oxygenase resistance. In the absence of
empirical evidence, this potential undesirable effect was judged to be small.
47 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The systematic review assessed that the overall certainty of evidence that pyrethroid-PBO ITNs have an impact on
malaria parasite prevalence was moderate.
Resources
Similar resources, other than the cost of the ITN itself, are needed for the deployment of the different ITN products that
are now available within the WHO recommended classes. (See table provided under 'Resources and other
considerations' for pyrethroid-only ITNs.)
Based on the available cost data, the GDG judged that there are currently additional costs associated with deploying
pyrethroid-PBO and other types of ITNs over pyrethroid-only LLINs. Due to the likely scale of ITN deployment, any
additional cost per net would amount to a considerable additional budget associated with a switch away from
pyrethroid-only LLINs, which would need to be met in order to maintain coverage. The GDG, however, remarked that
unit costs change over time and, as they do, a review will be needed to determine whether this cost discrepancy
remains. National programmes are encouraged to pay specific attention to the commodity cost, as this will also vary
depending on required quantities and lead-times and will be a key ingredient to the separately developed guidance on
ITN prioritization.
Apart from the higher cost of the net, the GDG identified no additional resource requirements associated with a switch
from pyrethroid-only LLINs to pyrethroid-PBO ITNs. Based on experience to date, pyrethroid-PBO ITNs require similar
resources to those identified for the distribution of pyrethroid-only LLINs (see table provided under “Resources and
other considerations” for pyrethroid-only LLINs). It would be necessary to assess the insecticide resistance status in the
principal vector(s) in the area where deployment is planned in order to determine whether pyrethroid resistance is
present and thus to justify such deployment. However, regular insecticide resistance testing by means of bioassays
should form part of routine programme monitoring operations and therefore should already be part of the budget.
Further information justifying the use of pyrethroid-PBO ITNs could be generated using standard WHO
procedures [22] to determine if a monooxygenase-based mechanism is at least partially involved in conferring pyrethroid
resistance.
The systematic review reported that cost-effectiveness analyses comparing pyrethroid-PBO ITNs and pyrethroid-only
LLINs are currently not available [59]. The GDG concluded that the cost-effectiveness of pyrethroid-PBO ITNs
compared to pyrethroid-only LLINs may vary. In areas of pyrethroid resistance, pyrethroid-PBO ITNs may have greater
impact on malaria than pyrethroid-only LLINs during the period for which the PBO is bioavailable. However, PBO is less
wash-resistant than pyrethroids and its bioavailability therefore declines faster over the three-year estimated life of an
ITN. The added impact of pyrethroid-PBO ITNs over that of pyrethroid-only LLINs may be lost or decline considerably
over time.
In addition to the issue of durability, the cost-effectiveness may also depend on a number of potential effect modifiers,
such as the malaria transmission level and vector characteristics in an area. Lastly, the GDG was concerned that, given
flatlined funding for malaria [4], the procurement of pyrethroid-PBO ITNs may negatively impact programmes’ ability to
maintain ITN coverage of at-risk populations. Due to the current moderately higher cost of this commodity, there is a
risk that existing net coverage could not be maintained if no additional funds were made available to cover the
additional expenditure required to purchase the same quantity of nets as previously deployed.
Equity
The impact on the equity of using pyrethroid-PBO ITNs instead of pyrethroid-only LLINs was judged to vary by the
GDG. If switching to more costly pyrethroid-PBO ITNs resulted in lower coverage of those at risk of contracting malaria
with preventive tools, equity would likely be reduced. However, if the switch resulted in no reduction in coverage and
48 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
those populations who were previously provided with pyrethroid-only LLINs were then protected against malaria by a
slightly more effective intervention, equity would likely increase.
Acceptability
No research was identified regarding the acceptability of pyrethroid-PBO ITNs. However, the GDG judged that such
nets would be equally acceptable to key stakeholders, given that they are by-and-large physically the same as and used
similarly to pyrethroid-only LLINs.
Feasibility
No research was identified regarding the feasibility of implementing pyrethroid-PBO ITNs. Nevertheless, the GDG
judged that distributing such nets would be equally feasible as for pyrethroid-only LLINs.
Justification
Pyrethroid-PBO ITNs combine pyrethroids and a synergist, which acts by inhibiting certain metabolic enzymes, primarily
oxidases, within the mosquito that would otherwise detoxify or sequester insecticides before they could reach their target
site in an insect. Therefore, compared to a pyrethroid-only LLIN, a pyrethroid-PBO ITNs should have an increased killing
effect on malaria vectors that express elevated oxidases, which is commonly associated with pyrethroid resistance.
The systematic review [59] identified and included two trials [57][58], both from eastern Africa, evaluating parasite
prevalence in areas where pyrethroid-PBO ITNs were deployed compared to pyrethroid-only LLINs. Both trials were
conducted in areas with highly pyrethroid-resistant mosquitoes, defined by the review team as mosquitoes demonstrating
<30% mortality in discriminating dose assays. Parasite prevalence was reduced by approximately 20% up to 25 months after
distribution. The Tanzanian trial has been extended further to establish whether this effect lasts the full duration of an LLIN's
intended 36-month life span, but results are not yet publicly available.
Although the two epidemiological trials included in the review were from areas where pyrethroid resistance was determined
to be high, the methods used by the authors to determine the level of resistance and the categorization of the different
bands of resistance intensity were not consistent with those recommended by WHO [22]. In many parts of Africa, as well as
other parts of the world, pyrethroid resistance is becoming more prevalent and is generally increasing in intensity in the
presence of continued selection pressure [4]. The panel therefore concluded that pyrethroid-PBO ITNs are likely to offer
greater protection against malaria than pyrethroid-only LLINs in most areas where pyrethroid resistance is detected and
mediated by elevated oxidases, regardless of resistance intensity.
When moving from the evidence provided to a decision on the strength of the recommendation, the GDG concluded that
the recommendation should be conditional rather than strong for this intervention. In the context of guideline development,
a conditional recommendation reflects the lower strength of a recommendation and one for which the GDG concludes that
the desirable effects of adhering to the recommendation probably outweigh the undesirable effects, but the panel is not
confident about these trade-offs. The conditionality of this recommendation was based on the fact that the available
evidence was only from African sites with pyrethroid resistance, rather than from other geographies; the moderate
additional benefit of deploying pyrethroid-PBO ITNs compared to pyrethroid-only LLINs; the overall moderate certainty of
the results; the higher unit cost of pyrethroid-PBO ITNs compared to pyrethroid-only LLINs; and the uncertainty of cost-
effectiveness.
Research Needs
WHO encourages additional high-quality research to generate further evidence on:
• the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria infection) and potential harms/
unintended consequences of pyrethroid-PBO ITNs in areas where the mechanisms of resistance in vector species are
not oxidase-based and in areas of lower malaria transmission intensity;
• contextual factors (e.g. acceptability, feasibility, resource use, cost-effectiveness, equity, values and preferences) related
to pyrethroid-PBO ITNs;
• the durability of pyrethroid-PBO ITNs.
49 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Pyrethroid-chlorfenapyr ITNs should be deployed instead of pyrethroid-only LLINs for prevention of malaria in adults and
children in areas with pyrethroid resistance.
Note: Recommendations on deployment of pyrethroid-chlorfenapyr nets were separated into two distinct recommendations for
better clarity, but share the same evidence to decision, justification, practical info and research needs. Please refer to the following
section.
Evidence To Decision
Pyrethroid-chlorfenapyr ITNs can be deployed instead of pyrethroid-PBO ITNs for prevention of malaria in adults and
children in areas with pyrethroid resistance.
The conditionality of the recommendation to deploy pyrethroid-chlorfenapyr ITNs instead of pyrethroid-PBO ITNs is based on the
GDG’s judgement that the balance of desirable and undesirable effects probably favours pyrethroid-chlorfenapyr ITNs over
pyrethroid-PBO ITNs. However, the evidence for this recommendation is from only one trial in Africa.
In deciding whether to deploy pyrethroid-chlorfenapyr ITNs instead of pyrethroid-only LLINs or pyrethroid-PBO ITNs, malaria
programmes should:
• determine whether resources are adequate to cover the extra costs compared to pyrethroid-only LLINs or pyrethroid-PBO
ITNs, while ensuring optimal coverage of populations at risk of malaria;
• generate additional information or conduct analyses with the aim of maximizing impact through targeted deployment (e.g.
stratification of malaria risk, assessment of the characteristics of local vectors, such as pyrethroid resistance mechanisms).
ITNs for prevention of malaria in adults and children in areas with pyrethroid resistance; and
• note that WHO recommends that ITNs prequalified by WHO be selected for deployment.
Practical Info
Given that pyrethroid-chlorfenapyr ITNs are designed to provide improved impact against insecticide-resistant mosquitoes,
pyrethroid resistance in potential target areas should be confirmed using standard procedures [22], as should the
susceptibility of local vectors to chlorfenapyr. In any case, pyrethroid-chlorfenapyr ITNs should not be considered a tool that
alone can effectively manage insecticide resistance in malaria vectors.
As with all malaria interventions, post-distribution monitoring of ITNs to estimate coverage in terms of access to and use of
ITNs is recommended. WHO also recommends that programmes conduct studies of ITN survival, which includes
assessments of ITN integrity, each time a campaign uses a new product such as pyrethroid-chlorfenapyr ITNs. Such studies
will provide information on the product’s life span under field conditions and thus enable estimation of the period over
which the additional impact against malaria may be maintained. The systematic review reported that, two years after
deployment, 34% of pyrethroid-chlorfenapyr ITNs were torn (defined as hole area ≥ 790 cm²) and therefore not fit for use,
compared to 28% of pyrethroid-only LLINs and 43% of pyrethroid-PBO ITNs.
50 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Evidence To Decision
The systematic review reported [Barker et al unpublished evidence] that one trial [61] recorded 90 (44.1%) adverse
events in the group assigned to the pyrethroid-only LLINs, 17 (8.5%) in the pyrethroid-chlorfenapyr ITN group and 17
(8.5%) in the pyrethroid-PBO ITN group. The authors also narratively reported that skin irritation was the most
commonly reported adverse event; however, no adverse event was assessed as serious. While five deaths were reported
in the cohort, three of these were from drowning, one was due to severe malaria and one to pneumonia; all of these
deaths were judged to be unrelated to the study interventions.
The review also reported data on ITN integrity from the United Republic of Tanzania [61]. The numbers (proportion) of
torn ITNs (defined as hole area ≥ 790 cm² and therefore not serviceable) were reported as 86 (28%) in the pyrethroid-
only LLIN group, 96 (34%) in the pyrethroid-chlorfenapyr ITN group and 81 (43%) in the pyrethroid-PBO ITN group.
The GDG noted that, compared to pyrethroid-only LLINs, pyrethroid-chlorfenapyr ITNs may exert an as yet unknown
selection pressure for the development of resistance to pyrrole insecticides and non-oxygenase resistance mechanisms
in Anopheles mosquito vectors.
Overall, the GDG judged that the extent of undesirable effects associated with pyrethroid-chlorfenapyr ITNs was small
compared to either pyrethroid-only LLINs or pyrethroid-PBO ITNs and that the overall balance of effects probably
favours pyrethroid-chlorfenapyr ITNs.
Based on the systematic review [Barker et al unpublished evidence], the GDG concluded that the overall certainty of
evidence that pyrethroid-chlorfenapyr ITNs have an impact against malaria was moderate.
Resources
Similar resources, other than commodity costs, would be needed for the deployment of pyrethroid-chlorfenapyr ITNs as
those listed for pyrethroid-only LLINs. (See table provided under “Resources and other considerations” for pyrethroid-
only LLINs.)
Based on the cost data reported by the study in the United Republic of Tanzania [61], pyrethroid-chlorfenapyr ITNs were
estimated to cost US$ 3.02 per ITN, while pyrethroid-only LLINs and pyrethroid-PBO ITNs were estimated to cost US$
2.07 and US$ 2.98 per ITN, respectively. Based on these data, the GDG judged that there are currently moderate
additional costs associated with deploying pyrethroid-chlorfenapyr ITNs instead of pyrethroid-only LLINs. Due to the
scale of existing ITN coverage, the moderate additional cost per ITN could amount to considerable additional costs
51 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
associated with a switch from pyrethroid-only LLINs to pyrethroid-chlorfenapyr ITNs, which would need to be met in
order to maintain the same population coverage.
The GDG, however, remarked that unit costs change over time and often decrease as new technologies are brought to
scale. As pyrethroid-chlorfenapyr ITNs are scaled up, further review will be needed to determine whether this cost
difference remains. National programmes are encouraged to pay specific attention to the commodity cost, as this will
also vary depending on required quantities and lead-times and will be a key ingredient to the separately developed
guidance on ITN prioritization.
Insecticide resistance status of the principal vector(s) in the area where deployment is planned should be assessed to
justify deployment of pyrethroid-chlorfenapyr nets. However, regular insecticide resistance testing by means of
bioassays [22] should already be part of routine monitoring operations and programme budgets.
The systematic review reported that the study conducted in the United Republic of Tanzania [61] carried out
cost-effectiveness analyses that compared pyrethroid-chlorfenapyr ITNs and pyrethroid-PBO ITNs to pyrethroid-only
LLINs over the two-year period of the trial. Pyrethroid-chlorfenapyr ITNs were estimated to avert 152 DALYs [SD 72]
per 10 000 total population, while pyrethroid-PBO ITNs averted 37 DALYs [SD 72] per 10 000 population. When
considering the costs of malaria diagnosis and treatment, pyrethroid-chlorfenapyr ITNs were reported to be less costly
(incremental cost US$ 2894 [SD 1129] per 10 000 population) than pyrethroid-PBO ITNs (US$ 4816 [SD 1360]) from all
perspectives. From societal and household perspectives, pyrethroid-chlorfenapyr ITNs would be more effective and less
costly than either pyrethroid-only LLINs or pyrethroid-PBO ITNs over a two-year period. The GDG concluded that the
cost-effectiveness would probably favour pyrethroid-chlorfenapyr ITNs over pyrethroid-only LLINs and pyrethroid-PBO
ITNs.
The GDG was concerned that, given flatlined funding for malaria [4], the procurement of pyrethroid-chlorfenapyr ITNs
may negatively impact the ability of programmes to maintain ITN coverage of at-risk populations. Due to the current
moderately higher cost of this commodity, there is a risk that programmes may not be able to maintain existing ITN
coverage or coverage of other malaria interventions if no additional funds to cover the higher costs are made available.
Some pragmatic prioritization guidance [3] has been provided with a view to supporting programmes in decision-making
around the deployment of new types of nets in resource-constrained environments.
Equity
The GDG judged that the impact on the equity of using pyrethroid-chlorfenapyr ITNs instead of pyrethroid-only LLINs
or pyrethroid-PBO ITNs is variable. If switching from pyrethroid-only LLINs to more costly pyrethroid-chlorfenapyr ITNs
would result in lower coverage of preventive interventions for those at risk of malaria, equity may be reduced. However,
if the switch resulted in no reduction in coverage (due to increased funding or price reduction) and those populations
who were previously provided with pyrethroid-only LLINs were then protected from malaria by a more effective
intervention, equity would likely increase.
Acceptability
No research was identified regarding the acceptability of pyrethroid-chlorfenapyr ITNs. However, the GDG judged that
such ITNs would be acceptable to key stakeholders, given that they are largely similar to pyrethroid-only LLINs and
pyrethroid-PBO ITNs in terms of their appearance, design and use, and given that they are currently available at a cost
similar to that of pyrethroid-PBO ITNs.
Feasibility
Although no research was identified regarding the feasibility of implementing pyrethroid-chlorfenapyr ITNs, the GDG
judged that deploying these ITNs would be as feasible as deploying pyrethroid-only LLINs or pyrethroid-PBO ITNs.
Justification
Pyrethroid-chlorfenapyr ITNs combine two active ingredients: a pyrethroid and a pyrrole insecticide. They are designed to
52 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
kill mosquitoes that are resistant to pyrethroids and, as such, fall into the second class of ITNs recognized by WHO. Pyrrole
insecticides such as chlorfenapyr disrupt adenosine 5'-triphosphate production in the mosquito’s mitochondria, thereby
reducing the target insects' ability to produce energy and leading to cell dysfunction and subsequent death. Pyrethroids,
meanwhile, target voltage-gated sodium channels associated with the nervous system of the insect, which results in
muscular paralysis and rapid death. Due to its different mode of action, chlorfenapyr is, therefore, unlikely to show any
cross-resistance to standard neurotoxic insecticides such as pyrethroids. Furthermore, death of the insect may occur 24–48
hours after exposure to chlorfenapyr, in contrast to pyrethroids, which result in a more rapid kill. The different entomological
mode and site of action of chlorfenapyr may reduce selection pressure for insecticide resistance. By including two active
ingredients in an ITN, the likelihood of the mosquitoes being resistant to both is greatly reduced. Therefore, compared to
pyrethroid-only LLINs or pyrethroid-PBO ITNs, pyrethroid-chlorfenapyr ITNs should have an increased killing effect against
pyrethroid-resistant malaria vectors and thus a greater impact against malaria.
The systematic review [Barker et al unpublished evidence] identified and included two trials [61][62] from eastern and
western Africa evaluating the impact of pyrethroid-chlorfenapyr ITNs on incidence of clinical malaria and prevalence of
malaria infection, compared to pyrethroid-only LLINs or pyrethroid-PBO ITNs. Both trials were conducted in areas with high
malaria transmission (malaria infection prevalence in children under 10 years of age recorded as 20–40%) and
pyrethroid-resistant mosquitoes. Compared to pyrethroid-only LLINs, incidence of clinical malaria (defined as malaria
symptoms, i.e. current fever with a temperature ≥ 37.5°C or fever in the past 48 hours, plus malaria parasitaemia) was
reduced by approximately 55% one year after deployment of pyrethroid-chlorfenapyr ITNs and by 40% two years post-
deployment. Prevalence of malaria infection (regardless of symptoms) was reduced by approximately 20% one year after
deployment and by approximately 45% two years post-deployment. Compared to pyrethroid-PBO ITNs, pyrethroid-
chlorfenapyr ITNs had little or no effect on incidence of clinical malaria one year after their deployment. However, after two
years, incidence was reduced by 35%. Prevalence of malaria infection was reduced by approximately 20% one year post-
deployment and by 30% two years post-deployment. The trials in Benin and the United Republic of Tanzania are continuing
and will investigate whether the protective effect lasts the full duration of an LLIN's intended 36-month life span. Results are
not available yet.
When moving from the evidence provided by the systematic review to a decision as to the strength of the recommendation,
the GDG concluded that there should be a strong recommendation to deploy pyrethroid-chlorfenapyr ITNs instead of
pyrethroid-only LLINs in areas where malaria vectors are resistant to pyrethroids. This was due to the large effect against
malaria and the high certainty that the benefits of deploying pyrethroid-chlorfenapyr ITNs instead of pyrethroid-only LLINs
would outweigh any harms. However, the panel concluded that the recommendation to deploy pyrethroid-chlorfenapyr ITNs
instead of pyrethroid-PBO ITNs in areas of insecticide resistance should be conditional. This was based on the fact that the
available evidence was from only one trial in the United Republic of Tanzania, where intensity of malaria transmission is high
and An. funestus is the primary malaria vector, which in turn limits generalizability of the findings to other geographies with
different anopheline vectors and eco-epidemiological characteristics. Furthermore, deploying pyrethroid-chlorfenapyr ITNs
was associated with a moderate additional benefit compared to pyrethroid-PBO ITNs two years after ITN deployment, but
with little or no difference in malaria outcomes one year after deployment.
Research Needs
WHO encourages additional high-quality research to generate further evidence on:
• the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria infection) and potential harms/
unintended consequences of pyrethroid-pyriproxyfen ITNs in areas with insecticide resistance traits in the local primary
vectors that differ from those of the available studies;
• contextual factors (e.g. acceptability, feasibility, resource use, cost-effectiveness, equity, values and preferences) related
to use of pyrethroid-chlorfenapyr ITNs; and
• the durability of pyrethroid-chlorfenapyr ITNs under field conditions.
53 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Pyrethroid-pyriproxyfen ITNs can be deployed instead of pyrethroid-only LLINs for prevention of malaria in adults and
children in areas with pyrethroid resistance.
The conditionality of the recommendation to deploy pyrethroid-pyriproxyfen ITNs instead of pyrethroid-only LLINs is based on the
GDG’s concerns that the available evidence indicates poor cost-effectiveness of pyrethroid-pyriproxyfen ITNs compared to
pyrethroid-only LLINs and that the extra resources currently required to purchase these ITNs may negatively impact on coverage
and equity.
In deciding whether pyrethroid-pyriproxyfen ITNs should be deployed instead of pyrethroid-only LLINs, malaria programmes should:
• determine whether resources are adequate to cover the extra cost compared to pyrethroid-only LLINs, while ensuring optimal
coverage of populations at risk of malaria;
• generate additional information or conduct analyses with the aim of maximizing impact through targeted deployment (e.g.
stratification of malaria risk, assessment of the characteristics of local vectors, such as pyrethroid resistance mechanisms); and
• note that WHO recommends that ITNs prequalified by WHO be selected for deployment.
Note: Recommendations on deployment of pyrethroid-pyriproxifen nets were separated into two distinct recommendations for
better clarity, but share the same evidence to decision, justification, practical info and research needs. Please refer to the following
section.
Evidence To Decision
Pyrethroid-pyriproxyfen ITNs are not recommended for deployment over pyrethroid-PBO ITNs for prevention of malaria in
adults and children in areas with pyrethroid resistance.
The conditionality of the recommendation against the deployment of pyrethroid-pyriproxyfen ITNs instead of pyrethroid-PBO ITNs
is based on the GDG’s judgement that the balance of effects favours pyrethroid-PBO ITNs over pyrethroid-pyriproxyfen ITNs and
that, based on current cost and efficacy data, pyrethroid-PBO ITNs are more cost-effective. The GDG acknowledged that evidence
to support this recommendation is derived from only a single trial in Africa.
Practical Info
Given that pyrethroid-pyriproxyfen ITNs are designed to provide improved impact against resistant mosquitoes, pyrethroid
resistance in potential target areas should be confirmed using standard procedures [22], as should susceptibility of the local
vectors to pyriproxyfen. In any case, pyrethroid-pyriproxyfen ITNs should not be considered a tool that alone can effectively
manage insecticide resistance in malaria vectors.
As with all malaria interventions, post-distribution monitoring of ITNs to estimate coverage in terms of access to and use of
ITNs is recommended. WHO also recommends that programmes conduct studies of ITN survival each time a campaign uses
a new product such as pyrethroid-pyriproxyfen ITNs, including assessment of ITN integrity. Such studies will provide
information on the life span of the product under field conditions and thus enable estimation of the period over which the
additional impact against malaria may be maintained. The systematic review reported that, two years after deployment, 39%
of pyrethroid-pyriproxyfen ITNs were torn (defined as having a total hole area ≥ 790 cm² and therefore assumed to be not fit
for use), compared to 28% of pyrethroid-only LLINs and 43% of pyrethroid-PBO ITNs.
54 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Evidence To Decision
The trial from the United Republic of Tanzania [61] included in the systematic review reported 90 (44.1%) adverse
events in the pyrethroid-only LLIN group, 80 (38.8%) in the pyrethroid-pyriproxyfen ITN group and 17 (8.5%) in the
pyrethroid-PBO ITN group. The authors also narratively reported that skin irritation was the most commonly reported
adverse event; however, no adverse event was assessed as serious. While five deaths were reported in the cohort, three
of these were from drowning, one was due to severe malaria and one was due to pneumonia; all deaths were judged to
be unrelated to the study interventions.
The review also reported data from the same trial [61] on ITN integrity. The numbers (proportion) of ITNs that were torn
(defined as hole area ≥ 790 cm²) were reported as 86 (28%) in the pyrethroid-only LLIN group, 109 (39%) in the
pyrethroid-pyriproxyfen ITN group and 81 (43%) in the pyrethroid-PBO ITN group.
Overall, the GDG judged the magnitude of undesirable effects associated with pyrethroid-pyriproxyfen ITNs to be small
compared to pyrethroid-only LLINs. However, compared to pyrethroid-PBO ITNs, the undesirable effects were judged to
be large. Overall, the GDG concluded that, compared to pyrethroid-only LLINs, the balance of effects probably favours
pyrethroid-pyriproxyfen ITNs, but when comparing pyrethroid-pyriproxyfen ITNs to pyrethroid-PBO ITNs, the balance
of effects was judged to favour the comparator, namely pyrethroid-PBO ITNs.
Based on the systematic review, the GDG concluded that the overall certainty of evidence that pyrethroid-pyriproxyfen
ITNs have an impact against malaria was moderate, compared to both pyrethroid-only LLINs and pyrethroid-PBO ITNs.
Resources
Apart from the higher commodity cost of pyrethroid-pyriproxyfen ITNs, similar resources would be needed for their
deployment as those listed for pyrethroid-only LLINs. (See table provided under “Resources and other considerations”
for pyrethroid-only LLINs.)
Based on cost data reported by the study in the United Republic of Tanzania [61], pyrethroid-pyriproxyfen ITNs were
estimated to cost US$ 3.68 per ITN, while pyrethroid-only LLINs and pyrethroid-PBO ITNs were estimated to cost US$
2.07 and US$ 2.98 per ITN, respectively. Based on these data, the GDG judged that there are currently moderate
additional costs associated with deploying pyrethroid-pyriproxyfen ITNs instead of pyrethroid-PBO ITNs. Based on the
likely scale of ITN deployment, this moderate additional cost per ITN could amount to considerable additional costs
associated with a switch to pyrethroid-pyriproxyfen ITNs, which would need to be met to maintain the same population
coverage. The GDG, however, remarked that unit costs change over time and often decrease as new technologies are
brought to scale. As pyrethroid-pyriproxyfen ITNs are scaled up, further review will be needed to determine whether
this cost difference remains. National programmes are encouraged to pay specific attention to the commodity cost, as
this will also vary depending on required quantities and lead-times and will be a key ingredient to the separately
developed guidance on ITN prioritization .
55 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
To justify the deployment of pyrethroid-pyriproxyfen nets, the insecticide resistance status of the principal vector(s) in
the area where deployment is planned should be assessed. However, regular insecticide resistance testing by means of
bioassays [22] should already be part of routine monitoring operations and programme budgets.
The systematic review reported that the study conducted in the United Republic of Tanzania [61] carried out
cost-effectiveness analyses comparing pyrethroid-pyriproxyfen ITNs and pyrethroid-PBO ITNs with pyrethroid-only
LLINs over the two-year period of the trial. Pyrethroid-pyriproxyfen ITNs were estimated to incur 9 DALYs [SD 71] per
10 000 total population, while pyrethroid-PBO ITNs averted 37 DALYs [SD 72] per 10 000 population. When
considering the costs of malaria diagnosis and treatment, pyrethroid-pyriproxyfen ITNs were reported to be the more
costly (incremental cost US$ 9621 [SD 1327] per 10 000 population), whereas pyrethroid-PBO ITNs were less costly
(US$ 4816 [SD 1360]) from all perspectives. The GDG concluded that the cost-effectiveness would probably favour
pyrethroid-only LLINs or pyrethroid-PBO ITNs over pyrethroid-pyriproxyfen ITNs.
The GDG was concerned that, given flatlined funding for malaria [4], the procurement of pyrethroid-pyriproxyfen ITNs
may negatively impact the ability of programmes to maintain ITN coverage of at-risk populations while not improving
impact. Due to the current moderately higher cost of this commodity, there is a risk that programmes may not be able to
maintain existing ITN coverage or coverage of other malaria interventions if no additional funds to cover the additional
costs are made available. Some pragmatic prioritization guidance [3] has been provided with a view to supporting
programmes in decision-making around the deployment of new types of nets in resource-constrained environments.
Equity
The GDG judged that the impact on the equity of using pyrethroid-pyriproxyfen ITNs instead of pyrethroid-only LLINs
or pyrethroid-PBO ITNs would vary. If switching from either of these types of nets to more costly pyrethroid-
pyriproxyfen ITNs resulted in lower coverage of preventive interventions for those at risk of malaria, equity may be
reduced. However, if the switch resulted in no reduction in coverage (due to increased funding or a price reduction) and
those populations who were previously provided with potentially less effective pyrethroid-only LLINs were then
protected from malaria by a potentially slightly more effective intervention, equity may increase.
Acceptability
No research was identified regarding the acceptability of pyrethroid-pyriproxyfen ITNs. However, the GDG judged that
such ITNs would be acceptable to key stakeholders, given that they are largely similar to pyrethroid-only LLINs and
pyrethroid-PBO ITNs in terms of their appearance, design and use.
Feasibility
Although no research was identified regarding the feasibility of implementing pyrethroid-pyriproxyfen ITNs, the GDG
judged that deploying such ITNs would be as feasible as deploying pyrethroid-only LLINs or pyrethroid-PBO ITNs.
Justification
Pyrethroid-pyriproxyfen ITNs combine a pyrethroid insecticide and an insect growth regulator (IGR). The two ingredients
have different entomological effects. The pyrethroid insecticide rapidly kills mosquitoes by targeting voltage-gated sodium
channels associated with the nervous system of the insect. The IGR is a hormone mimic that does not directly kill insects,
but disrupts their growth and reproduction. Mosquitoes that are not killed by the pyrethroid may be sterilized and/or have
their fecundity reduced, thereby preventing multiplication of the insecticide-resistant mosquitoes. Pyriproxyfen has also
shown some impact on a mosquito’s life span. Pyrethroid-pyriproxyfen ITNs, therefore, fall into the third class of ITNs
recognized by WHO, which consists of ITNs primarily designed to sterilize and/or reduce the fecundity of insecticide-
resistant mosquitoes. It is unlikely that mosquitoes exposed to ITNs that combine a pyrethroid and an IGR will be resistant to
both active ingredients due to their different modes of action and limited to no selection pressure exerted so far for
pyriproxyfen resistance. As such, pyrethroid-pyriproxyfen ITNs could have a greater impact against malaria than pyrethroid-
only LLINs in areas with pyrethroid-resistant malaria vectors.
The systematic review [Barker et al unpublished evidence] identified and included three trials [61][62][63] from western and
56 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
eastern Africa, evaluating the impact of pyrethroid-pyriproxyfen ITNs on incidence of clinical malaria and prevalence of
malaria infection, compared to either pyrethroid-only LLINs or pyrethroid-PBO ITNs. All trials were conducted in areas of
high malaria transmission (malaria infection prevalence in children under 10 years of age recorded by the trials as 20–40%
and as 50–70% in children under 5) and pyrethroid-resistant mosquitoes. Compared to pyrethroid-only LLINs, incidence of
clinical malaria (defined as malaria symptoms, i.e. current fever of temperature ≥ 37·5°C or fever in the past 48 hours, plus
malaria parasitaemia) decreased by approximately 20% one year after deployment of pyrethroid-pyriproxyfen ITNs and by
15% two years post-deployment. Prevalence of malaria infection (regardless of symptoms) was reduced by approximately
30% one year post-deployment and by approximately 20% two years post-deployment. Compared with pyrethroid PBO
ITNs, the use of pyrethroid-pyriproxyfen ITNs, the use of pyrethroid-pyriproxyfen ITNs was associated with a two-fold
higher incidence of clinical malaria one year after ITN deployment, with a slightly increased or no effect on incidence two
years post-deployment. There was no effect on prevalence of malaria infection one or two years post-deployment. The trial
in Benin is continuing and will investigate whether the impact against malaria varies for the full duration (36-month life span)
of an LLIN. Results are not available yet.
The GDG concluded on a conditional recommendation to deploy pyrethroid-pyriproxyfen ITNs instead of pyrethroid-only
LLINs in areas where malaria vectors are resistant to pyrethroids. The recommendation for deployment was based on the
moderate effect against malaria and the GDG’s judgement that the benefits probably outweighed any harms of deploying
pyrethroid-pyriproxyfen ITNs instead of pyrethroid-only LLINs. The conditionality, however, was stipulated based on the
panel conclusion that pyrethroid-pyriproxyfen ITNs were less cost-effective than pyrethroid-only LLINs and, due to the
higher unit cost of pyrethroid-pyriproxyfen ITNs, extra resources would be required to replace pyrethroid-only LLINs with
these dual active ingredient ITNs. Unless additional resources are provided, a switch to pyrethroid-pyriproxyfen ITNs would
result in reduced coverage of populations at risk of malaria, thereby negatively affecting coverage and equity.
The panel conditionally recommended against the deployment of pyrethroid-pyriproxyfen ITNs instead of pyrethroid-PBO
ITNs in areas of insecticide resistance. This decision was based on the lack of evidence of pyrethroid-pyriproxyfen ITNs
having a greater impact against malaria compared to pyrethroid-PBO ITNs; the balance of effects favours pyrethroid-PBO
ITNs over pyrethroid-pyriproxyfen ITNs. Based on these results and the current unit costs of pyrethroid-pyriproxyfen ITNs,
pyrethroid-PBO ITNs are currently more cost-effective. Extra resources would be required while there would be no benefit
of deploying pyrethroid-pyriproxyfen ITNs instead of pyrethroid-PBO ITNs, and, in the absence of additional resources, this
would result in reduced coverage of malaria interventions for populations at risk of malaria, thereby negatively affecting
equity. The GDG also acknowledged that the available evidence on the efficacy of pyrethroid-pyriproxyfen ITNs compared
to pyrethroid-PBO ITNs was from only one trial conducted in the United Republic of Tanzania, where malaria transmission is
high and An. funestus is the primary malaria vector, which in turn limits generalizability of the findings to other geographies
with different anopheline vectors and eco-epidemiological characteristics.
Research Needs
WHO encourages additional high-quality research to generate further evidence on:
• the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria infection) and potential harms/
unintended consequences of pyrethroid-pyriproxyfen ITNs in areas with insecticide resistance traits in the local primary
vectors that differ from those of the available studies;
• contextual factors (e.g. acceptability, feasibility, resource use, cost-effectiveness, equity, values and preferences) related
to use of pyrethroid-pyriproxyfen ITNs;
• the durability of pyrethroid-pyriproxyfen ITNs over the expected life span of an LLIN (three years).
Insecticide-treated nets (ITNs) should be deployed for the prevention and control of malaria in children and adults in areas
with ongoing malaria transmission affected by a humanitarian emergency.
This recommendation is limited to classes of ITNs currently recommended by WHO. As with ITNs deployed in more stable settings,
WHO recommends that ITNs that are prequalified by WHO be selected for use in humanitarian emergencies.
When considering deployment of ITNs in humanitarian emergencies, the infrastructure, access, logistical capacity and resources
available must be taken into account, as these may influence the feasibility and cost of procuring and deploying nets.
57 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Practical Info
In deciding whether to deploy ITNs in emergency settings, consideration must be given to whether ITNs are appropriate for
that setting, taking into account vector characteristics, human behaviour and available infrastructure. ITNs are most effective
where the principal malaria vector(s) bite predominantly at night after people have retired under their nets and where the
mosquitoes are susceptible to the insecticides used to treat the nets. Data will need to be collected to assess whether these
criteria are met. There may be more limited capacity to gather such data in humanitarian emergencies than in more stable
settings. In addition to assessing whether ITNs are appropriate, consideration of the feasibility of deploying nets in a
particular emergency setting is important. Depending on the infrastructure, access, logistical capacity and resources
available, procuring and distributing nets may be more challenging than in more stable settings. Instability in such settings
may challenge long-term planning and so result in shorter lead times and consequently higher costs. It is also important to
determine whether the shelters or housing structures in such settings are suitable for hanging a net. In some situations, the
structure may have nowhere to hang a net or it may be too small to adequately accommodate a net.
Other considerations for the deployment, monitoring and evaluation of nets apply equally to emergency and non-emergency
settings. Please consult the practical information under the WHO recommendations for the different ITN classes. However,
as for collecting data to assess whether nets are suitable in an area, the feasibility and capacity to regularly collect
information for M&E in emergency settings must be assessed.
Evidence To Decision
The systematic review did not report any unintended consequences of the intervention. However, the GDG noted that
the potential undesirable effects identified for the use of ITNs in stable settings are also likely to apply in humanitarian
emergencies. The GDG also noted that if nets are deployed in settings where the population is accommodated in tents
or small houses (structures that are commonly shelters in emergency settings), uptake and use may be limited because
the restricted space may not allow the net to be hung easily and the net may encroach on the space required for other
household activities. The GDG judged these potential undesirable effects to be minimal.
Although the studies included in the systematic review were limited to the use of pyrethroid-only LLINs, the likely
benefits extend to other types of ITNs that are recommended by WHO for large-scale deployment in more stable
settings (e.g. pyrethroid-PBO nets). The GDG judged the balance of benefits and harms to favour the use of ITNs that
have been recommended for use in more stable settings to prevent and control malaria in humanitarian emergency
settings.
The systematic review assessed that the overall certainty of the evidence that pyrethroid-only LLINs ha ve s an impact
on malaria in humanitarian emergency settings was high.
58 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Resources
Based on cost data published in 2021 [35], the median economic cost of ITNs was US$ 1.39 per person protected per
year, drawing on data from non-emergency settings. The GDG noted that the cost of deploying nets in humanitarian
emergency settings may be higher than in stable settings for a number of reasons. First, the cost of transporting nets
may increase, particularly for locations that are difficult to access. Second, in some emergency settings, there may be a
need to establish human capacity for net delivery, which could incur further cost. Finally, given the nature of emergency
settings, the necessity for immediate deployment of interventions may require shorter lead times for procurement,
resulting in higher costs of the commodity. The GDG judged that deploying ITNs would therefore involve moderate
costs and cost more than deploying ITNs in stable settings.
A review of the cost and cost-effectiveness of malaria control interventions [35] in more stable settings reported that
the cost-effectiveness of ITNs compared to no ITNs was US$ 5.85 per episode averted, US$ 1281.97 per death
averted, and US$ 44.51 per disability-adjusted life year (DALY) averted. The GDG noted that the cost-effectiveness of
deploying pyrethroid-only LLINs may depend largely on the setting: the cost-effectiveness may vary with the
infrastructure in the setting and available capacity, as well as the malaria transmission level in the area of deployment.
The GDG judged that, while there may be some upfront costs to deliver nets in such settings, given the associated
benefits to protecting such vulnerable populations, deploying pyrethroid-only LLINs would be cost-effective compared
to no nets.
Equity
Providing ITNs to populations in areas with ongoing malaria transmission affected by humanitarian emergencies was
judged by the GDG to result in increased equity, as populations in these settings are at increased risk of malaria
infection.
Acceptability
No research was identified regarding the acceptability of pyrethroid-only LLINs in emergency settings. Nevertheless, the
GDG judged that ITNs would be acceptable to key stakeholders, given that they are generally well accepted in more
stable settings. The acceptability may improve further over time as users see the benefit to protecting themselves from
malaria.
Feasibility
No research was identified regarding the feasibility of implementing pyrethroid-only LLINs in humanitarian emergency
settings. The GDG judged that distributing ITNs would be feasible, but consideration would need to be given to
whether:
• the sleeping structures in the setting are amenable to having nets installed;
• nets can be procured in time and within the given budget;
• there is sufficient human capacity to deliver nets in the emergency setting; and
• there are sufficient resources available to cover potential extra costs to access the population, particularly hard-to-
reach populations and those affected by conflict.
Justification
The systematic review [54] (Messenger et al unpublished findings) compared pyrethroid-only LLINs to no nets in terms of
malaria outcomes in areas affected by humanitarian emergencies. The review concluded that deploying pyrethroid-only
LLINs was associated with reductions in P. falciparum parasite incidence, P. falciparum parasite prevalence and P. vivax
parasite incidence compared to no nets. It was unclear whether pyrethroid-only LLINs reduced P. vivax parasite prevalence in
these settings. The included studies were all from emergencies in the chronic phase in Asia – in the Republic of Union of
Myanmar, on the Myanmar–Thailand border, and in the Islamic Republic of Pakistan. Deploying nets in the acute stage of an
emergency may differ from deploying nets once some infrastructure has been established, due to numerous logistical
challenges. Humanitarian emergencies in other parts of the world may differ in terms of the available capacity, infrastructure,
community behaviour and acceptance.
59 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Given that the systematic review only identified and included four trials, a number of potential effect modifiers could not be
examined. However, as for pyrethroid-only LLINs deployed in more stable settings, the impact of nets may vary depending
on, for example, the behaviour of the mosquito species, the level and mechanism(s) of insecticide resistance, parasite
prevalence, and net usage by the population.
While the review included studies that only examined the impact of pyrethroid-only LLINs, other ITNs recommended by
WHO in more stable settings are likely to have a similar balance of benefits and harms to those deployed in humanitarian
emergencies. Important considerations regarding resource needs, acceptability and feasibility when deploying pyrethroid-
only LLINs in emergency settings should largely apply to other WHO-recommended ITNs. Based on the review findings and
these considerations, the GDG judged that the desirable effects of deploying WHO-recommended ITNs, not just pyrethroid-
only LLINs, in humanitarian emergencies compared to no nets would outweigh the undesirable effects. Based on the high
certainty of the findings from emergency settings and the feasibility, acceptability and cost-effectiveness of ITNs in more
stable settings, the panel felt that the recommendation should be classified as strong.
Research Needs
WHO encourages funding of high-quality research to generate further evidence on:
• the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria infection) and potential harms/
unintended consequences of ITNs in the acute phase of humanitarian emergencies (where logistics and priorities may
differ); and
• contextual factors (i.e. acceptability, feasibility, resource use, cost-effectiveness, equity, values and preferences) related
to products from the different ITN classes covered by a WHO recommendation deployed in humanitarian emergencies.
Achieving and maintaining optimal coverage with ITNs for malaria prevention and control (2019)
To achieve and maintain optimal ITN coverage, countries should apply mass free net distribution through campaigns,
combined with other locally appropriate delivery mechanisms such as continuous distribution using antenatal care (ANC)
clinics and the Expanded Programme on Immunization (EPI).
Recipients of ITNs should be advised (through appropriate communication strategies) to continue using their nets beyond
the three-year expected lifespan, irrespective of the condition and age of the net, until a replacement net is available.
Practical Info
To achieve and maintain optimal ITN coverage, countries should apply a combination of mass free net distribution through
campaigns and continuous distribution through multiple channels, in particular through ANC clinics and the EPI. Mass
campaigns are the only proven cost-effective way to rapidly achieve high and equitable coverage. Complementary
continuous distribution channels are also required because coverage gaps can start to appear almost immediately post-
campaign due to net deterioration, loss of nets, and population growth.
Mass campaigns should distribute one ITN for every two persons at risk of malaria. However, for procurement purposes, the
calculation to determine the number of ITNs required needs to be adjusted at the population level, since many households
have an odd number of members. Therefore, a ratio of one ITN for every 1.8 persons in the target population should be used
to estimate ITN requirements, unless data to inform a different quantification ratio are available. In places where the most
recent population census is more than five years old, countries can consider including a buffer (e.g. adding 10% after the 1.8
ratio has been applied) or using data from previous ITN campaigns to justify an alternative buffer amount. Campaigns should
also normally be repeated every three years, unless available empirical evidence justifies the use of a longer or shorter
interval between campaigns. In addition to these data-driven decisions, a shorter distribution interval may be justified during
humanitarian emergencies, as the resulting increase in population movement may leave populations uncovered by vector
control, potentially increasing their risk of infection as and the risk of epidemics.
Continuous distribution through ANC and EPI channels should remain functional before, during and after mass distribution
campaigns. In determining the optimal mix of ITN delivery mechanisms to ensure optimal coverage and maximized
efficiency, consideration should be given to the required number of nets, the cost per net distributed and coverage over
time. For example, during mass distribution campaign years, other delivery schemes may need to be altered to avoid-over
supply of ITNs.
60 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
“Top-up” campaigns (i.e. ITN distributions that take into account existing nets in households and provide each household
only with the additional number of nets needed to bring it up to the target number) are not recommended. Substantial field
experience has shown that accurate quantification for such campaigns is generally not feasible and the cost of accounting
for existing nets outweighs the benefits.
There should be a single national ITN plan and policy that includes both continuous and campaign distribution strategies.
This should be developed and implemented under the leadership of the NMP, based on an analysis of local opportunities
and constraints, and identification of a combination of distribution channels with which to achieve optimal coverage and
minimize gaps. This unified plan should include a comprehensive net quantification and gap analysis for all public sector ITN
distribution channels. As much as possible, the plan should include major ITN contributions by the private sector.
• ANC, EPI and other child health clinics: These should be considered high-priority continuous ITN distribution channels
in countries where these services are used by a large proportion of the population at risk of malaria, as occurs in much
of sub-Saharan Africa.
• Schools, faith- and community-based networks, and agricultural and food-security support schemes: These can also be
explored as channels for ITN distribution in countries where such approaches are feasible and equitable. Investigating
the potential use of these distribution channels in complex emergencies is particularly important.
• Occupation-related distribution channels: In some settings, particularly in Asia, the risk of malaria may be strongly
associated with specific occupations (e.g. plantation and farm workers and their families, miners, soldiers and forest
workers). In these settings, opportunities for distribution through channels such as private sector employers, workplace
programmes and farmers’ organizations may be explored.
• Private or commercial sector channels: These can be important channels for supplementing free ITN distribution
through public sector channels. Access to ITNs can also be expanded by facilitating the exchange of vouchers or
coupons provided through public sector channels for a free or subsidized ITN at participating retail outlets. ITN
products distributed through the private sector should be regulated by the national registrar of pesticides in order to
ensure that product quality is in line with WHO recommendations.
The procurement of ITNs with attributes that are more costly (e.g. nets of conical shape) is not recommended for countries
in sub-Saharan Africa, unless nationally representative data clearly show that the use of ITNs with particular attributes
increases significantly among populations at risk of malaria. To build an evidence base to support the purchase of more
costly nets, investigation into the population's preferences and whether adhering to those preferences translates into
increased use of ITNs may also be warranted, particularly in situations where standard nets are unlikely to suit the lifestyle
of specific population groups at risk of malaria, such as may be the case for nomadic populations.
The life spans of ITNs can vary widely among individual nets used within a single household or community, as well as among
nets used in different settings. This makes it difficult to plan the rate or frequency at which replacement nets need to be
procured and delivered. All malaria programmes that have undertaken medium- to large-scale ITN distributions should
conduct ITN durability monitoring in line with available guidance to inform appropriate replacement intervals. Where there is
evidence that ITNs are not being adequately cared for or used, programmes should design and implement BCC activities
aimed at improving these behaviours.
In countries where untreated nets are widely available, NMPs should promote access to ITNs. Strategies for treating
untreated nets can also be considered, for example, by supporting access to insecticide treatment kits.
As NMPs implement different mixes of distribution methods in different geographic areas, there will be a need to accurately
track ITN coverage at subnational levels. Subnational responses should be triggered if coverage falls below programmatic
targets. Tracking should differentiate among the contributions of various delivery channels to overall ITN coverage.
Countries should generate data on defined standard indicators of coverage and access rates in order to ascertain whether
optimal coverage has been achieved and maintained. The data should also inform changes in implementation in order to
improve performance and progress towards the achievement of programmatic targets. Currently, the three basic survey
indicators are: i) the proportion of households with at least one ITN; ii) the proportion of the population with access to an
ITN within their household; and iii) the proportion of the population reporting having slept under an ITN the previous night
(by age [<5 years; 5–14 years; 15+ years], gender and access to ITN).
61 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Justification
In December 2017, WHO published updated recommendations on Achieving and maintaining universal coverage with LLINs for
malaria control [64]. These recommendations were developed and revised based on expert opinion through broad
consultation, including multiple rounds of reviews by the Malaria Policy Advisory Group (MPAG). Under the section on
“practical information”, these recommendations have been summarized and slightly revised to clarify that these
recommendations are not specific to LLINs, but apply to ITNs in general.
Old ITNs should only be collected where there is assurance that: i) communities are not left without nets, i.e. new ITNs are
distributed to replace old ones; and ii) there is a suitable and sustainable plan in place for safe disposal of the collected
material.
If ITNs and their packaging (bags and baling materials) are collected, the best option for disposal is high-temperature
incineration. They should not be burned in the open air. In the absence of appropriate facilities, they should be buried away
from water sources and preferably in non-permeable soil.
Recipients of ITNs should be advised (through appropriate communication strategies) not to dispose of their nets in any
water body, as the residual insecticide on the net can be toxic to aquatic organisms (especially fish).
Practical Info
It is important to determine whether the environmental benefits outweigh the costs when identifying the best disposal
option for old ITNs and their packaging. For malaria programmes in most endemic countries, there are limited options for
dealing with ITN collection. Recycling is not currently a practical option in most malaria-endemic countries (with some
exceptions for countries with a well-developed plastics industry). High-temperature incineration is likely to be logistically
difficult and expensive in most settings. In practice, when malaria programmes have retained or collected packaging material
in the process of distributing ITNs, it has mostly been burned in the open air. This method of disposal may lead to the release
of dioxins, which are harmful to human health.
If such plastic material (with packaging an issue at the point of distribution and old ITNs an intermittent issue at household
level when the net is no longer in use) is left in the community, it is likely to be re-used in a variety of ways. While the
insecticide exposure entailed by this kind of re-use has yet to be fully studied, the expected negative health and
environmental impacts of leaving the waste in the community are considered to be less than amassing it in one location and/
or burning it in the open air.
Since the material from nets represents only a small proportion of total plastic consumption, it will often be more efficient
for old ITNs to be dealt with as part of larger and more general solid-waste programmes. National environment management
authorities have an obligation to consider and plan for what happens to old ITNs and packaging materials in the environment
in collaboration with other relevant partners.
Justification
Currently, ITNs and the vast majority of their packaging (bags and baling materials) are made of non-biodegradable
plastics [65].The large-scale deployment of ITNs has given rise to questions as to the most appropriate and cost-effective
way to deal with the resulting plastic waste, particularly given that most endemic countries do not currently have the
resources to manage ITN collection and waste disposal programmes.
A pilot study was conducted to examine patterns of ITN usage and disposal in three African countries (the Republic of
Kenya, the Republic of Madagascar and United Republic of Tanzania). Findings of this pilot study, along with other
background information were used to generate recommendations through the WHO Vector Control Technical Expert Group
(VCTEG) and MPAG on best practices with respect to managing waste.
The following are the main findings from the pilot study and other background material:
• ITNs entering domestic use in Africa each year contribute approximately 100 000 tonnes of plastic and represent a per
capita rate of plastic consumption of 200g per year. This is substantial in absolute terms; however, it constitutes only
approximately 1% to 5% of the total plastic consumption in Africa and thus is small compared to other sources of plastic
62 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
IRS should be deployed for the prevention and control of malaria in children and adults living in areas with ongoing malaria
transmission.
WHO recommends that WHO-prequalified insecticidal products be selected for IRS use and that these be selected based on the
insecticide susceptibility of the local malaria vector(s). IRS is considered an appropriate intervention where:
Practical Info
IRS is considered an appropriate intervention where:
• the majority of the vector population tends to feed and rest indoors;
• people mainly sleep indoors at night;
• the malaria transmission pattern is such that the population can be protected by one or two rounds of IRS per year;
• the majority of structures are suitable for spraying.
When selecting insecticides to be used for IRS, it is important to investigate the resistance profile of the local vectors in
order to select insecticides to which the vectors are susceptible.
Insecticide formulations currently used for IRS fall into five major insecticide classes with three modes of action, based on
their primary target site in the vector. WHO-prequalified products have been assessed for their safety, quality and
entomological efficacy, which includes evaluation of their mortality effect on mosquitoes when applied to a range of interior
surfaces of dwellings found in malaria-endemic areas.
63 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Acetylcholinesterase inhibitors
• Neonicotinoids: clothianidin
IRS products using four of these insecticide classes have been prequalified by WHO; as of August 2020, there were no
organochlorine IRS formulations prequalified, including DDT. This means that no DDT product has been assessed by WHO
for its efficacy, safety and quality for vector control, and no inspection of manufacturing sites has been conducted. Unlike
the other four classes covered by WHO’s recommendation for IRS, DDT has been classified as a persistent organic pollutant.
As such, its production and use are strictly restricted by an international agreement known as the Stockholm Convention on
Persistent Organic Pollutants [66]. The Convention’s objective is to protect both human health and the environment from
persistent organic pollutants. When the Stockholm Convention was established in 2004, it provided an exemption for the
production and use of DDT for disease vector control, mainly because of the absence of equally effective and efficient
alternatives at the time. The recent expansion of products available for IRS and overall expansion of vector control
interventions has provided additional options.
WHO actively supports the promotion of chemical safety and, together with the United Nations Environment Programme,
shares a common commitment to the global goal of reducing and eventually eliminating the use of DDT, while minimizing
the burden of vector-borne diseases. DDT use for malaria vector control has declined over the years and WHO supports
continuation of this trend.
In some areas, the use of DDT may be warranted. The decision to use DDT for malaria vector control needs to be based on a
detailed analysis that considers all other potential options for vector control and provides clear reasoning for choosing DDT
over the other options. WHO considers DDT to be a last resort, not a first choice. If DDT is selected, it should be used under
strict control measures and only for the intended purpose. Its use requires that the conditions set by the Stockholm
Convention be met. Effective use and safe storage of DDT rely on compliance with well-established and well-enforced rules
and regulations in accordance with national guidelines and following WHO technical guidance provided in the WHO
Operational manual for IRS for malaria transmission, control and elimination [67]. Where DDT is deployed, it is essential for
adequate resources and technical support to be in place to ensure the sound management of this persistent organic
pollutant.
Countries that are using DDT for malaria vector control need to regularly (at least once every two years) reassess whether
there is a justified continued need for DDT. The outcome of such assessment should be reported to the WHO Global
Malaria Programme and to the Secretariat of the Stockholm Convention as part of the formal reporting process [66].
When selecting products and formulations, residual efficacy needs to continue for at least three months after the application
of the insecticide to the substrate (usually cement, mud or wood) [68]. Insecticides are available in various formulations to
increase their longevity on different surfaces.
Community acceptance of IRS is critical to the programme’s success, particularly as it requires householders to grant
permission for spray teams to enter their house. It also involves disruption to the household, requiring householders to
remove personal items from their house prior to spraying. Furthermore, some insecticide formulations leave unsightly
residue on sprayed surfaces. Repeated, frequent spraying of houses over extended periods can lead to refusal by
householders. Reduced acceptance has been an impediment to effective IRS implementation in various parts of the
world [69]. It is therefore important to develop information, education and communication (IEC) strategies to keep the
community informed and to ensure full support and cooperation.
In areas with ongoing malaria transmission, optimal coverage of IRS should be maintained. Implementation of the
intervention should take place prior to the onset of the peak transmission season. Following application of the insecticide(s),
64 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The WHO Operational manual for IRS for malaria transmission, control and elimination [67] aims to assist malaria programme
managers, entomologists and public health officers in designing, implementing and sustaining high-quality IRS programmes.
Evidence To Decision
The systematic review also compared IRS to pyrethroid-only ITNs in areas of intense and unstable malaria transmission.
It concluded that in areas of intense malaria transmission, IRS may reduce malaria incidence compared to ITNs (RR: 0.88;
95% CI: 0.78–0.98; one study; low-certainty evidence), but there may be little or no difference between IRS and ITNs in
terms of parasite prevalence (RR: 1.06; 95% CI: 0.91–1.22; one study; very low-certainty evidence). Comparing IRS with
ITNs in areas of unstable transmission, the review reported that IRS may be associated with increased malaria incidence
(RR: 1.48; 95% CI: 1.37–1.60; one study; low-certainty evidence) and parasite prevalence (RR: 1.70; 95% CI: 1.18–2.44;
one study; low-certainty evidence) compared to ITNs.
No undesirable effects were identified in the systematic review. However, IRS may play an as yet undetermined role in
insecticide resistance development in Anopheles vectors.
The systematic review assessed that the overall certainty of the evidence that IRS has an impact on malaria was low.
65 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• PPE
• Spray pump repair parts
• Insecticide and packaging (including return/clean packaging)
• Soap/bathing materials
Supplies
• Inventory management forms
• Documentation paperwork/forms or electronic devices
• Entomological supplies for wall cone bioassays and maintenance of adult
mosquitoes
• M&E data collection forms
Justification
When carried out correctly, IRS has historically been shown to be a powerful intervention to reduce adult mosquito vector
density and longevity and, therefore, to reduce malaria transmission. However, despite its long tradition and the large body
of associated operational experience, few randomized controlled trials (RCTs) have been conducted on IRS. Therefore, the
availability of data suitable for use in the meta-analysis was limited [70] and the certainty of evidence reported by the
systematic review was low. The GDG considered that despite the low certainty of the evidence, a strong recommendation
for the intervention is warranted based on the fact that a number of implementation trials and programmatic data have
demonstrated impact against malaria. The GDG considered that this body of evidence, when viewed as a whole, provides
higher certainty evidence (compared to the evidence from the systematic review) of the effectiveness of IRS as a malaria
prevention and control intervention. The GDG judged that, based on the systematic review comparing IRS and ITNs, ITNs
are an equally effective alternative intervention in areas where local vectors are susceptible to the insecticides being
used [70].
66 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
An updated systematic review of data on IRS interventions from recent studies, RCTs and other designs is being undertaken
to further support this recommendation or modify it as appropriate.
Research Needs
• Generate further evidence on the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria
infection) and potential harms and/or unintended consequences of IRS.
• Determine the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria infection) and potential
harms/unintended consequences of IRS in urbanized areas with changing housing designs.
• Determine the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria infection) and potential
harms/unintended consequences of IRS using new insecticides in areas where mosquitoes are resistant to currently
deployed insecticides.
• Determine the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria infection) of IRS in areas
with different mosquito behaviours (such as in areas with outdoor transmission).
• Given the relatively high cost of implementing IRS, especially in the context of growing insecticide resistance and when
delivering IRS in more remote areas, there is a need to investigate new approaches to delivering IRS to increase the
cost-effectiveness of this intervention.
IRS can be deployed for the prevention and control of malaria in children and adults in areas with ongoing malaria
transmission affected by a humanitarian emergency.
The conditionality of this recommendation is largely driven by the very low certainty of the evidence that IRS reduces malaria in
such settings and due to concerns around feasibility and cost.
When deciding whether IRS may be appropriate for prevention and control of malaria in humanitarian emergency settings,
programmes should consider:
• whether the structures are suitable for spraying. Some shelters provided in emergency settings may not be suitable for
application of insecticides, such as open-sided structures and those built from materials that affect the residual nature of the
insecticides;
• whether the target coverage of IRS can be feasibly achieved in the setting;
• whether there are sufficient resources to cover the relatively high costs associated with an IRS programme. In such settings,
transport of commodities to hard-to-reach areas, coupled with the need to quickly procure items and establish human
capacity to deliver the intervention, is likely to incur higher costs than when deploying IRS in more stable settings.
As with the deployment of IRS in more stable settings, WHO recommends that WHO-prequalified insecticides be selected for IRS
use in humanitarian emergencies. It is important to ensure that the vector population is susceptible to the insecticide selected for
spraying.
Practical Info
In deciding whether to deploy IRS in emergency settings, as in more stable settings, consideration must be given to whether
IRS is a suitable intervention for that setting, taking into account vector characteristics, human behaviour and available
infrastructure. IRS is considered an appropriate intervention where the majority of the vector population feeds and rests
indoors; the vectors are susceptible to the insecticide that is being deployed; people mainly sleep indoors at night; the
majority of structures are suitable for spraying; and where high enough coverage can be achieved to provide community-
level protection. Data will need to be collected to assess whether these criteria are met. Data on vector composition,
density, behaviour and insecticide susceptibility prior to deploying IRS not only provide information as to whether IRS is
suitable in that setting, but also provide baseline information against which changes can be detected and monitored.
Combined with data on coverage, this information can be used to gauge the effectiveness and efficiency of IRS. However,
there may be more limited capacity to regularly gather such data in humanitarian emergencies than in more stable settings.
Data are also required on the structures present in humanitarian emergencies to assess whether they are amenable to IRS.
Open-sided structures or those with surfaces constructed from materials that impact the residual nature of the spray may
not be suitable.
Initiating any IRS programme requires a well-defined management system to be established with dedicated human, logistical,
transport and financial resources. Programmes and implementing partners should consider whether the logistical needs
67 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
(acquisition of commodities and equipment, recruitment of personnel and transport) can be met in emergency situations
with the available resources within the given timeframe. Timeliness is a key factor in obtaining the maximum benefits from
IRS; the spray should be applied over the shortest period of time just prior to the onset of the transmission season. As with
ITNs, instability in humanitarian emergencies may reduce the options for long-term planning, resulting in shorter lead times
for establishing a programme and acquiring supplies and equipment than in more stable settings. If commodities and
personnel have to be sourced at short notice, procurement costs may be higher. Costs may also increase if more expensive
means of transport are required for deployment in more remote, less accessible areas or those affected by conflict.
As with more stable settings, ensuring optimal coverage to provide community-level protection is critical. To support this
community acceptance of IRS is essential. Given that in some humanitarian emergencies, the local language may differ to
that of the affected population, consideration should be given to whether messaging needs to be adapted.
Evidence To Decision
The GDG judged that the extent of the desirable effects of IRS compared to no IRS is likely to vary depending on a
number of factors. Many of these factors also apply to more stable settings: IRS works best when the majority of
vectors rest indoors and are susceptible to the insecticides used; where people sleep indoors; where the population is
not nomadic; and where the structures are sprayable and not too scattered. The suitability of structures for spraying is
an important factor to consider in emergency settings. Tents are often used to provide emergency shelter and not all
tent material will allow the application of the insecticide by spraying; in some areas, structures are open-sided. It may
be that IRS is more appropriate in the chronic phase of an emergency than in the acute phase due to the type of shelter,
infrastructure and human capacity likely to have been established by this later stage.
The systematic review did not report any unintended consequences of the intervention. However, the GDG noted that
undesirable effects may be similar to those that may arise when deploying IRS in non-emergency settings (see “Evidence
to decision” section of the recommendation for IRS). These undesirable effects were judged by the GDG to be minimal.
The GDG judged the balance of benefits and harms to probably favour the use of IRS against malaria compared to no
IRS in humanitarian emergency settings.
The systematic review assessed the overall certainty of evidence that IRS has an impact on malaria in humanitarian
emergency settings to be very low.
Resources
The resources needed for IRS in humanitarian emergencies are, at a minimum, the same as those needed for delivery of
68 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
IRS in more stable settings (see “Resources and other considerations” table, section 4.1.1), but the overall cost is likely to
be higher due to the various logistical issues noted below. Based on cost data published in 2021 [35] the median
economic cost per person protected per year was estimated to be US$ 5.70 in stable settings. As in stable settings,
establishing an IRS programme in an area for the first time requires a great amount of resources. In emergency settings,
increased costs are assumed to be associated with transporting commodities and personnel to areas where access is
limited by geography or conflict, the fact that shorter lead times for procurement generally result in higher cost of
goods, and the need to quickly establish capacity (recruitment and training of personnel, establishment of operation
sites, i.e. stores, soak pits, and wash areas) to protect the at-risk population and avoid a potential malaria epidemic. The
GDG therefore judged that deploying IRS in such settings would likely involve high costs.
Data from a review of the cost and cost-effectiveness of malaria control interventions deployed in stable
settings [35] reported that the cost-effectiveness of IRS compared to no IRS was US$ 840.44 per death averted and
US$ 25.16 per DALY averted. The GDG noted that the cost-effectiveness of deploying IRS is likely to vary depending on
the malaria transmission level in the area of deployment and other contextual factors. However, the GDG judged that
IRS is likely to be cost-effective compared to no IRS, given the benefits of protecting vulnerable populations from
malaria in such settings.
Equity
Providing IRS to populations in areas with ongoing malaria transmission affected by humanitarian emergencies was
judged by the GDG to result in increased equity by providing the most vulnerable with an effective malaria prevention
intervention
Acceptability
No research was identified regarding the acceptability of IRS in emergency settings. Despite the lack of evidence, the
GDG judged that IRS is likely to be acceptable to key stakeholders, given that IRS is generally accepted in more stable
settings.
Feasibility
No evidence was included in the systematic review and no studies were identified by the GDG regarding the feasibility
of implementing IRS in humanitarian emergency settings.
The GDG judged that the feasibility of IRS would vary, likely depending on whether:
• the structures in such settings are amenable to being sprayed; open-sided structures and certain surface materials
would not be suitable for spraying;
• commodities can be acquired and skilled personnel recruited with the resources available within the given
timeframe;
• access to the population is feasible, which may involve higher costs than in more stable settings.
The GDG noted that IRS may be more feasible in the chronic phase of a humanitarian emergency, when shelter, general
infrastructure and human resources are better established than in the acute stages. In the acute phase of an emergency,
there may be other competing demands on resources and overall capacity.
Justification
The systematic review [54] (Messenger et al unpublished evidence) included four studies conducted in the Islamic Republic of
Pakistan and The Republic of the Sudan that compared IRS versus no IRS on malaria outcomes in areas affected by
humanitarian emergencies. The review included only one observational study showing that P. falciparum was reduced, but
the certainty of evidence was considered to be very low. One RCT showed no effect of IRS on P. falciparum parasite
prevalence (low-certainty evidence). IRS was reported to reduce both P. vivax parasite incidence and prevalence based on
two observational studies, but the certainty of evidence was assessed to be very low. All studies were conducted during the
69 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
chronic phase of the emergency. Deploying IRS in the acute stage of an emergency may differ from employing IRS once
some infrastructure has been established, due to numerous logistical challenges.
Given that the systematic review only identified and included four studies, a number of potential effect modifiers could not
be examined, and the generalizability of the findings was limited. Humanitarian emergencies in other parts of the world may
differ in terms of available capacity, infrastructure, community behaviour and acceptance. As for many vector control
interventions, the impact of IRS may vary in different settings depending on a number of factors, such as the behaviour of
the mosquito species, the level and mechanism(s) of insecticide resistance in vectors, parasite prevalence, and coverage of
IRS in the population. As with deploying IRS in more stable settings, IRS will only be effective where vectors rest primarily
indoors and mosquitoes are susceptible to the insecticide being deployed.
The review findings provided little evidence of an impact on malaria outcomes in humanitarian emergencies. Given the
effectiveness of IRS programmes in reducing malaria burden in more stable settings, however, the GDG judged that the
desirable effects of deploying IRS compared to no IRS in humanitarian emergencies would likely outweigh the undesirable
effects. Given the low certainty of the evidence, the panel felt that the recommendation should be classified as conditional.
Considerations of feasibility and the cost and cost-effectiveness of implementing IRS in such settings were viewed by the
GDG as important. In humanitarian emergencies, the shelters provided may not be amenable to spraying and there may be
higher costs associated with deploying IRS in such settings than in more stable ones.
Research Needs
WHO encourages funding of high-quality research to generate further evidence on:
• the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria infection) and potential harms/
unintended consequences of IRS in the acute phase of humanitarian emergencies (where logistics and priorities may
differ);
• contextual factors (i.e. acceptability, feasibility, resource use, cost-effectiveness, equity, values and preferences) related
to IRS deployed in humanitarian emergencies.
Prioritize optimal coverage with either ITNs or IRS over combination (2019)
The co-deployment of ITNs and IRS is not recommended for prevention and control of malaria in children and adults in areas
with ongoing malaria transmission. Priority should be given to delivering either ITNs or IRS at optimal coverage and to a high
standard, rather than introducing the second intervention as a means to compensate for deficiencies in the implementation
of the first intervention.
In settings where optimal ITN coverage, as specified in the strategic plan, has been achieved and where ITNs remain effective,
additionally implementing IRS may have limited utility in reducing malaria morbidity and mortality. Given the resource constraints
across malaria endemic countries, it is recommended that effort be focused on good-quality implementation of either ITNs or IRS,
rather than deploying both in the same area. However, the combination of these interventions may be considered for resistance
prevention, mitigation or management should sufficient resources be available.
Practical Info
Given the resource constraints across malaria-endemic countries, the deployment of a second vector control intervention on
top of optimal coverage with an existing one should only be considered as part of a broader prioritization analysis aimed at
achieving maximum impact with the available resources. In many settings, a switch from ITNs to IRS or vice versa, rather
than their combination, is likely to be the only financially feasible option. Deployment of either intervention needs to ensure
optimal coverage of populations at risk of malaria and ensure they are delivered to a high standard. Further guidance on best
practices for ensuring high-quality deployment of interventions is provided in the WHO Indoor residual spraying: An
operational manual for IRS for malaria transmission, control and elimination [67] and in the Alliance for Malaria Prevention toolkit.
70 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Evidence To Decision
The certainty of evidence identified in the systematic review showing no benefit to adding IRS in situations where ITNs
are already being used was graded as moderate.
Justification
The systematic review published in 2019 [74] on the deployment of IRS in combination with ITNs (specifically pyrethroid-
only LLINs) provided evidence that, in settings where there is optimal coverage with ITNs and where these remain effective,
IRS may have limited utility in reducing malaria morbidity and mortality. WHO guidance was developed accordingly to
emphasize the need for good-quality implementation of either ITNs or IRS, rather than deploying both in the same area [75].
However, the co-deployment of these interventions may be considered for resistance prevention, mitigation or management
should sufficient resources be available
Insecticide resistance threatens the effectiveness of insecticidal interventions and hence is a key consideration in
determining which vector control interventions to select to ensure maximum impact. One approach to the prevention,
mitigation and management of vector insecticide resistance is the co-deployment (or combination) of interventions with
different insecticides (see Section 4.1 on “Prevention, mitigation and management of insecticide resistance”). Therefore,
WHO guidance developed based on the systematic review [74] differentiates between the effect of combined interventions
on malaria morbidity and mortality versus the utility of this approach in a resistance management strategy [75].
A summary of the conclusions (with minor updates for clarity) used to develop the above recommendations is as follows:
• In settings with high ITN coverage where ITNs remain effective, IRS may have limited utility in reducing malaria
morbidity and mortality. However, IRS may be implemented as part of an IRM strategy in areas where ITNs are in
use [21].
• Malaria control and elimination programmes should prioritize the delivery of ITNs or IRS at optimal coverage and to a
high standard, rather than introducing the second intervention as a means to compensate for deficiencies in the
implementation of the first intervention.
• If ITNs and IRS are to be deployed together in the same geographical location, IRS should be conducted with a non-
pyrethroid insecticide.
• Evidence is needed to determine the effectiveness of combining IRS and ITNs in malaria transmission foci, including in
low transmission settings. Evidence is also needed from different eco-epidemiological settings outside of Africa.
• All programmes in any transmission setting that decide to prioritize the combined deployment of ITNs and IRS over
other potential use of their financial resources should include a rigorous programme of M&E (e.g. a stepped wedge
introduction of the combination) in order to confirm whether the additional inputs are having the desired impact.
Countries that are already using both interventions should similarly undertake an evaluation of the effectiveness of the
combination versus either ITNs or IRS alone.
• The approach of co-deploying interventions for resistance management was developed largely based on experience
with agricultural pest management, and the evidence base from public health remains weak.
71 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Research Needs
• Further evidence is needed on the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria
infection) and potential harms and/or unintended consequences of co-deploying non-pyrethroid IRS with ITNs vs ITNs
only in areas with insecticide-resistant mosquito populations.
• Determine whether there are comparative benefits (incidence of malaria [infection or clinical] and/or prevalence of
malaria infection), as well as potential harms/unintended consequences of combining non-pyrethroid IRS with ITNs vs
IRS only in areas with insecticide-resistant mosquito populations.
• Determine the acceptability of co-deploying IRS and ITNs among householders and communities.
• Evaluate new tools for monitoring the quality of IRS and ITN interventions.
Access to effective vector control using ITNs or IRS at optimal coverage levels should be ensured for all populations at risk
of malaria in most epidemiological and ecological settings.
Practical Info
Financial considerations such as cost and cost-effectiveness are major drivers of decision-making, and the selection of
malaria vector control interventions and determination of their coverage should thus be embedded in a prioritization process
that considers the cost and effectiveness of all available malaria interventions and aims at achieving maximum impact with
the available resources. Evaluations of the relative cost and cost-effectiveness of ITNs and IRS are ongoing to inform revision
of the guidelines.
Justification
ITNs can provide both personal and community-level protection when nets are deployed at the community rather than
individual level, with the aim of providing sufficient nets to cover all household inhabitants. Similarly, IRS will have a greater
effect on mosquito populations and therefore transmission if deployed at high coverage. It is therefore important to
maximize access to ITNs or IRS in communities that are at risk of malaria. This will involve quantification of needs to enable
access for all household inhabitants when placing procurement orders and putting in place appropriate delivery
structures. For malaria vector control interventions recommended for large-scale deployment, namely ITNs and IRS, optimal
coverage refers to providing populations at risk of malaria with access to ITNs coupled with health promotion to maximize
use and ensuring timely replacement; or providing these populations with regular application of IRS. Either intervention
should be deployed at a level that provides the best value for money while reflecting programmatic realities. In practice, this
often means quantifying commodities to provide full access by the population at risk, while realizing that this will not result
in 100% coverage or 100% access due to various system inefficiencies. Being cognizant of such constraints, decision-making
should then consider other alternatives as part of the intervention package, ranging from chemoprevention to
supplementary vector control, instead of pursuing the idealistic goal of providing full population coverage.
In terms of the relative effectiveness of IRS compared to pyrethroid-only ITNs, a systematic review published in
2010 [70] reported low-certainty evidence that, in areas of intense malaria transmission, IRS may be associated with lower
malaria incidence, but no effect was evident for parasite prevalence. In areas of unstable transmission, ITNs may be
associated with lower malaria incidence and prevalence; however, the certainty of evidence was determined to be very low.
The panel therefore could not provide a definitive conclusion on the comparative effectiveness of these interventions. WHO
currently views these two interventions as being equally effective ways of delivering an insecticide. The actual effectiveness
in reducing the burden of malaria is dependent on the insecticide(s) used on the ITN or applied by IRS. Decisions on whether
to deploy IRS or ITNs need to be informed by a number of factors, such as data on insecticide resistance, past and present
experience of using interventions (including feasibility of deployment and acceptability and use by end-users), vector
behaviours and the current options available within the context. Given these various considerations, the wide range of
different contexts and the lack of correlation between insecticide resistance data assessed using bioassays and the actual
effectiveness of an insecticidal intervention in controlling vectors, no general recommendation to guide the selection of ITNs
over IRS can be made.
72 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
In areas with ongoing local malaria transmission (irrespective of both the pre-intervention and current level of transmission),
vector control interventions should not be scaled back. Ensuring access to effective malaria vector control at optimal levels
for all inhabitants of such areas should be pursued and maintained.
Practical Info
Access to effective vector control interventions will need to be maintained in the majority of countries and locations where
malaria control has been effective. This includes settings with ongoing malaria transmission, as well as those in which
transmission has been interrupted but some level of receptivity and importation risk remains. Malaria elimination is defined
as the interruption of local transmission (reduction to zero incidence of indigenous cases) of a specified malaria parasite
species in a defined geographical area as a result of deliberate intervention activities. Following elimination, continued
measures to prevent re-establishment of transmission are usually required [30]. Interventions are no longer required once
eradication has been achieved. Malaria eradication is defined as the permanent reduction to zero of the worldwide incidence
of infection caused by all human malaria parasite species as a result of deliberate activities.
There is a critical need for all countries with ongoing malaria transmission, and in particular those approaching elimination, to
build and maintain strong capacity in disease and entomological surveillance and health systems. The capacity to detect and
respond to possible resurgences with appropriate vector control relies on having the necessary entomological information
(i.e. susceptibility status of vectors to insecticides, as well as their biting and resting preferences). Such capacity is also
required for the detailed assessment of malariogenic potential, which is a pre-condition for determining whether vector
control can be scaled back (or focalized).
If areas where transmission has been interrupted are identified, the decision to scale back vector control should be based on
a detailed analysis that includes assessment of the receptivity and importation risk of the area, as well as an assessment of
the active disease surveillance system, and capacity for case management and vector control response.
Justification
A comprehensive review of historical evidence and mathematical simulation modelling undertaken for WHO in 2015
indicated that the scale-back of malaria vector control was associated with a high probability of malaria resurgence, including
for most scenarios in areas where malaria transmission was very low or had been interrupted [76]. Both the historical review
and the simulation modelling clearly indicated that the risk of resurgence was significantly greater at higher EIRs and case
importation rates, and lower coverage of active case detection and case management.
Once transmission has been reduced to very low levels approaching elimination, ensuring optimal access to vector control
for at-risk populations remains a priority, even though the size and demographics of the at-risk populations may change as
malaria transmission is reduced.
As malaria incidence falls and elimination is approached, increasing heterogeneity in transmission will result in foci with
ongoing transmission in which vector control may need to be optimized and enhanced. Such foci may be the result of
particularly high vectorial capacity, lapsed prevention and treatment services, changes in parasites that make the current
strategies less effective, or reintroduction of malaria parasites by the movement of infected people or infected
mosquitoes. Monitoring the coverage, quality and impact of vector control interventions is essential to maintain the
effectiveness of control. Guidance on entomological surveillance across the continuum from control to elimination is
provided elsewhere [30].
Once elimination has been achieved, vector control may need to be continued by targeting defined at-risk populations to
prevent reintroduction or re-establishment of local transmission.
It is acknowledged that malaria transmission can persist following the implementation of a widely effective malaria
programme. The sources and risks of residual transmission may vary by location, time and the existing components of the
current malaria programme. This variation is potentially due to a combination of both mosquito and human behaviours, such
as when people live in or visit forest areas or do not sleep in protected houses, or when local mosquito vector species bite
and/or rest outdoors and thereby avoid contact with IRS or ITNs/LLINs.
Once elimination has been achieved, optimal vector control coverage should be maintained in receptive areas where there is
73 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
74 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Larviciding (2019)
Insecticides can be regularly applied to water bodies (larviciding) for the prevention and control of malaria in children and
adults as a supplementary intervention to ITNs or IRS in areas with ongoing malaria transmission where aquatic habitats are
few, fixed and findable.
The conditionality of this recommendation is due to the low certainty of evidence, the impact being limited to non-extensive
habitats, and concerns about feasibility.
• Larviciding only reduces vector density and so does not have the same potential for health impact as ITNs and IRS; ITNs
provide protection from biting vectors and both ITNs and IRS reduce adult longevity.
• Larviciding should not be seen as a substitute for ITNs or IRS or a means to fill a coverage gap in areas with significant malaria
risk; rather, larviciding represents a potential supplementary strategy for malaria control.
• Feasibility and cost-effectiveness should be taken into account; larviciding will generally be most cost-effective in areas where
larval habitats are few, fixed and findable, and likely less feasible in areas where the aquatic habitats are abundant, scattered
and variable.
The following settings are potentially the most suitable for larviciding as a supplementary measure implemented alongside ITNs or
IRS:
• urban areas: where breeding sites are relatively few, fixed and findable in relation to houses (which are targeted for ITNs or
IRS);
• arid regions: where larval habitats may be few and fixed throughout much of the year.
Practical Info
Larviciding is most likely to be cost-effective in urban areas where the appropriate conditions are more likely to be present.
Larviciding is not generally recommended in rural settings, unless there are particular circumstances limiting the larval
habitats and specific evidence confirming that such measures can reduce malaria incidence in the local setting. Determining
whether or not specific habitats have immature Anopheles larvae and are suitable for larviciding is essential and should be
based on expert technical opinion and knowledge.
WHO's 2013 Operational manual on larval source management [81] concluded that ITNs and IRS remain the backbone of
malaria vector control, but LSM represents an additional (supplementary) strategy for malaria control in Africa. Larviciding
will generally be most effective in areas where larval habitats are few, fixed and findable, and likely less feasible in areas
where the aquatic habitats are abundant, scattered and variable. Determination of whether or not specific habitats are
suitable for larviciding should be based on assessment by an entomologist. The WHO operational manual focuses on sub-
Saharan Africa, but the principles espoused are likely to hold for other geographic regions that fit the same criteria. The
following settings are potentially the most suitable for larviciding as a supplementary measure implemented alongside ITNs
or IRS:
• urban areas: where breeding sites are relatively few, fixed and findable in relation to houses (which are targeted for ITNs
or IRS);
• arid regions: where larval habitats may be few and fixed throughout much of the year.
Larviciding is likely to be more acceptable in communities that have a good understanding of the lifecycle of mosquitoes and
the link with the transmission of malaria or other diseases. Community members may have concerns about larvicides being
applied to drinking water or other domestic water sources. A well-designed community sensitization programme is required
to ensure that communities fully understand the intervention and that any concerns about health and safety aspects are
75 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
addressed.
Evidence To Decision
The systematic review [82] reported that larviciding for non-extensive larval habitats less than 1km2 may have an effect
in reducing malaria incidence (rate ratio: 0.24; one trial; low-certainty evidence) and parasite prevalence (risk ratio: 0.79;
95% CI: 0.71–0.89; two studies; low-certainty evidence) compared to no larviciding. However, it is not known whether
larviciding has an effect on malaria incidence (OR: 1.97; 95% CI: 1.39–2.81; one study; very low-certainty evidence) or
parasite prevalence (OR: 1.49; 95% CI: 0.45–4.93; one study; very low-certainty evidence) compared to no larviciding in
large-scale aquatic habitats.
No undesirable effects were identified in the systematic review. However, larviciding may affect non-target fauna;
communities may not accept its application to sources of drinking water or water used for other domestic purposes.
For larval habitats less than 1km2, the systematic review assessed that the overall certainty of evidence that larviciding
has an impact on malaria was low. In larger habitats, the certainty of evidence was judged to be very low.
• Larvicide
Supplies • PPE
• Entomological supplies for larval monitoring and rearing/maintenance of adult
mosquitoes
76 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• Computer/communication equipment
Justification
Larviciding is deployed for malaria control in several countries, including the Federal Republic of Somalia and the Republic
of The Republic of the Sudan. However, the systematic review on larviciding conducted in 2019 [82] assessed that the
certainty of evidence of impact on malaria incidence or parasite prevalence was moderate or low in non-extensive habitats.
Since larviciding only reduces vector density, it does not have the same potential for health impact as ITNs and IRS – both of
which reduce vector longevity (a key determinant of transmission intensity) and provide protection from biting vectors. As a
result, larviciding should never be seen as a substitute for ITNs or IRS in areas with significant malaria risk.
Research Needs
• Further evidence is needed on the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria
infection) and potential harms/unintended consequences of larviciding.
• Evaluate new technologies for identifying aquatic habitats.
No recommendation can be made because the evidence on the effectiveness of a specific larval habitat modification and/or
larval habitat manipulation intervention for the prevention and control of malaria was deemed to be insufficient.
Practical Info
Although the available evidence that met the inclusion criteria for the systematic review was considered insufficient to
develop specific recommendations, national programmes may decide to use environmental management (habitat
modification and/or manipulation) to avoid the creation, and reduce the availability of, larval habitats, where deemed
appropriate based on expert guidance and local knowledge. If such strategies are employed, the selection of the specific
intervention(s) should be highly contextual, i.e. it should take into account the specific environment, the types of
interventions relevant to that environment, the resources needed and their availability, the feasibility of the intervention(s),
acceptability by local stakeholders and potential impact on equity. The selection should also take into account previous
experience either gained locally or from other areas of similar ecological and epidemiological characteristics where such
intervention(s) have been implemented. Additionally, the selection of the comparator should consider other interventions
that are known to be cost-effective, for example, larviciding. Where the decision is taken to invest resources into larval
habitat modification and/or larval habitat manipulation, the intervention(s) should be designed and conducted with the
explicit aim of generating data to demonstrate effective malaria control, preferably supported with environmental and
entomological data as secondary end-points.
When assessing the impact of environmental management against malaria, it is important that the testing of the
77 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
intervention(s) under investigation be conducted specifically for the purpose of preventing or controlling malaria by reducing
the availability and productivity of larval habitats. For example, dams are generally constructed for water management,
irrigation or power production purposes, not for malaria control. In fact, in some cases, their construction may result in
increased larval production due to the creation of standing water bodies. The controlled release of water from the
impoundment of a dam, however, is considered an example of habitat manipulation – a recurrent activity that potentially
controls mosquito larvae by increasing the flow rate of downstream water with the aim of preventing mosquito development
and so controlling malaria transmission. This is one example of the multitude of interventions that fall under the broad
category of larval habitat modification and/or manipulation. To be able to generate evidence on the efficacy of larval habitat
modification and/or manipulation in preventing malaria, and to facilitate the interpretation of the evidence once generated,
it is important to well define the interventions that are being evaluated and, importantly, compare how the water conditions
of larval habitats at the intervention and control sites are affected. For example, if the intervention aimed to increase the
water flow to downstream areas, the evaluation should include an assessment of whether this was achieved, the extent to
which this impacted the development of the immature and adult stages of the mosquito, and, ultimately, whether there was
an epidemiological impact against malaria in the intervention arms compared to control areas. This information will then
support the evolution of WHO guidance in this area and, ultimately, guide the choice and implementation of efficacious
interventions.
Evidence To Decision
The systematic review (Martello et al unpublished evidence) identified two studies that investigated the impact of habitat
manipulation by controlling the release of water from flood gates of dams or spillways (overflow channels) across
streams to flush downstream areas with water against malaria. It is unknown whether larval habitat manipulation has an
effect on malaria parasite prevalence compared to no larval habitat manipulation (relative risk: 0.01; 95% CI: 0.0–0.16;
one study; very low-certainty evidence). It is unknown whether larval habitat manipulation combined with IRS has an
effect on malaria clinical incidence compared to IRS alone (odds ratios or relative risks could not be calculated because
the numbers of participants in each arm or at follow-up were not reported; one study; very low-certainty evidence).
Justification
The systematic review (Martello et al unpublished evidence) to inform WHO recommendations in this area identified only two
controlled before-after studies meeting the inclusion criteria with epidemiological outcomes that investigated the impact of
larval habitat manipulation alone. No studies investigating the impact of larval habitat modification on malaria outcomes
were identified. Two other identified studies combined habitat manipulation with larviciding and so the effect of the two
could not be separated. One study was conducted in an urban area of the Republic of the Philippines in 1960 and the other
in a forested area of the Republic of India in 2008 where annual IRS was also conducted. The studies provided low- or very
low-certainty evidence that the controlled release of water from flood gates of dams to discharge excess water or using
78 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
spillways (overflow channels) across streams to automatically flush downstream areas with water (continually or
intermittently) reduced clinical malaria incidence or parasite prevalence. The evidence was downgraded due to the lack of
appropriate randomization or poor statistical reporting. The studies examined very specific interventions, each studied in a
single site, which the GDG judged would limit their generalizability. The systematic review reported a number of other
studies with only entomological outcomes investigating a wide range of highly heterogeneous interventions falling under the
broad term of larval habitat manipulation and/or modification, some of which may only be appropriate in specific ecologies.
Given the broad range of interventions and settings in which larval habitat manipulation and/or modification may be applied,
the GDG judged that the potential impact, feasibility, acceptability and resource needs for each intervention are likely to be
highly variable.
Although it is acknowledged that there is a wealth of historical research on environmental management of malaria, the
literature did not meet the eligibility criteria to be included in this systematic review. Therefore, there remains a continued
need to robustly demonstrate the epidemiological impact of environmental management (habitat modification and/or
manipulation) on malaria incidence and prevalence through further well-designed intervention studies.
Research Needs
The GDG encourages funding of high-quality research on the impact of habitat manipulation and/or modification on malaria
transmission to inform the development of specific WHO recommendations in this area. A number of evidence gaps and
associated requirements were identified:
• Determine the impact (incidence of clinical malaria and/or prevalence of malaria infection) and potential harms/
unintended consequences of the different interventions.
• Epidemiological evidence is required on the efficacy against malaria of the same intervention implemented in different
settings (where vector species may differ).
• Detailed descriptions are needed of the interventions deployed, as well as larval habitat types and vector species
targeted. The impact of the intervention on the water conditions of the larval habitats should be assessed, i.e.
properties of the habitat that the intervention aims to modify such as water flow, volume, sunlight penetration, salinity
or other physical conditions.
• Evidence is needed on contextual factors, (i.e. acceptability, feasibility, resource use, cost-effectiveness, equity, values
and preferences) related to larval habitat modification and/or manipulation is needed.
No recommendation can be made because no evidence on the effectiveness of larvivorous fish for the prevention and
control of malaria was identified.
Evidence To Decision
The GDG recognized that there are specific settings in which the intervention is currently implemented, and in these
specific settings programme staff consider it to be effective.
79 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Justification
The systematic review conducted in 2017 on the use of larvivorous fish [83] did not identify any studies demonstrating
impact on malaria and so there is insufficient evidence to support a recommendation. The GDG recognized that there are
specific settings in which the intervention is currently implemented, and in these specific settings programme staff consider
it to be effective. In some of the settings where larvivorous fish are being deployed, programmatic evidence exists; however,
this was not determined appropriate for inclusion in the systematic review due to unsuitable study design or other concerns.
The GDG acknowledged that there may be data at the country/programme level that it is not aware of.
Research Needs
• Determine the impact (incidence of malaria (infection or clinical) and/or prevalence of malaria infection) and potential
harms/unintended consequences of the use of larvivorous fish.
The deployment of topical repellents in areas with ongoing malaria transmission is not recommended if the aim is to prevent
and control malaria at the community level.
The panel recommended against the implementation of topical repellents with the aim of controlling malaria at the community
level, given the lack of evidence of a significant impact. To achieve community-level impact, it is likely that a high level of individual
compliance would be needed. Further work is required to separate out the potential protective effects at the individual and/or
community level and therefore fully assess the potential public health value of topical repellents.
Evidence To Decision
Based on expert opinion and in line with current WHO recommendations, topical repellents may still be useful in
providing personal protection against malaria.
The systematic review assessed that the overall certainty of the evidence that topical repellents have an impact on
malaria at the community level was very low.
80 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Justification
The RCTs included in the systematic review conducted in 2018 [77] provided low certainty evidence of a possible effect of
topical repellents on malaria parasitaemia (P. falciparum and P. vivax). The evidence is insufficiently robust to determine
whether topical repellents have an effect on clinical malaria.
Research Needs
• Determine the impact (incidence of malaria (infection or clinical) and/or prevalence of malaria infection) and potential
harms/unintended consequences of topical repellents for individuals in specific settings and target populations.
Deployment of insecticide-treated clothing is not recommended for the prevention and control of malaria at the community
level in areas with ongoing malaria transmission; however, insecticide-treated clothing may be beneficial as an intervention
to provide personal protection against malaria in specific population groups.
The GDG recommended against the deployment of insecticide-treated clothing due to the lack of evidence of an impact in the
general population. In the absence of ITNs, there is some evidence that insecticide-treated clothing may reduce the risk of malaria
infection in specific populations such as refugees and military personnel.
Evidence To Decision
The systematic review assessed that the overall certainty of the evidence that insecticide-treated clothing in specific
populations has an impact on malaria was low.
Justification
The systematic review carried out in 2018 [77] provided low-certainty evidence that insecticide-treated clothing may have
protective efficacy against P. falciparum and P. vivax cases, at least in certain specific populations (refugees, military personnel
and others engaged in occupations that place them at high risk) and where ITNs are not in use. There was no evidence
81 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Research Needs
• Determine the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria infection) and potential
harms/unintended consequences of insecticide-treated clothing in the general population.
• Identify approaches to enhance acceptability/desirability and increase uptake and adherence.
• Develop formulations that improve the durability of insecticidal efficacy.
No recommendation can be made because the evidence on the effectiveness of spatial/airborne repellents for the
prevention and control of malaria was deemed to be insufficient.
Evidence To Decision
Justification
The systematic review published in 2018 [77] concluded that there is very low-certainty evidence that spatial or airborne
repellents may have protective efficacy against malaria parasitaemia. Therefore, no recommendation on the use of spatial/
airborne repellents in the prevention and control of malaria can be made until more studies assessing malaria
epidemiological outcomes have been conducted.
Research Needs
• Determine the impact (incidence of malaria [infection or clinical)] and/or prevalence of malaria infection) and potential
harms/unintended consequences of spatial/airborne repellents.
• Develop spatial repellent insecticide formulations that provide a long-lasting effect.
Space spraying is not recommended for the prevention and control of malaria in children and adults in areas with ongoing
malaria transmission; IRS or ITNs should be prioritized instead.
The panel recommended against the deployment of space spraying to control malaria, given the lack of evidence of impact against
malaria. Due to the short-lived nature of the insecticides used, space spraying is generally costly and wasteful of resources.
82 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Evidence To Decision
The panel judged that any anticipated desirable effect of space spraying is likely to be small, as the insecticide
formulations used are short-lived. Anopheles mosquitoes are generally considered to be less susceptible to space
spraying than Culex or Aedes.
The systematic review assessed that the overall certainty of the evidence that space spraying has an impact on malaria
was very low.
Justification
Only observational study was identified by the systematic review and the certainty of the evidence was graded as very
low [84]. The lack of data from RCTs, other trial designs or quasi-experimental studies has therefore hampered a
comprehensive assessment of this intervention and the review concluded that it is unknown whether space spraying causes
a reduction in the incidence of malaria. The anticipated desirable effects of space spraying are likely to be small, as the
insecticide formulations used are short-lived. Anopheles mosquitoes are generally considered to be less susceptible to space
spraying than Culex or Aedes. Space spraying is frequently applied when cases are at their peak, which is followed by a
decline in cases, whether or not control measures are applied. Nevertheless, space spraying is often deployed in response to
outbreaks of mosquito-borne disease. Due to the high visibility of this intervention, the decision to use this approach is
usually made to demonstrate that the authorities are taking action in response to the outbreak. This practice should be
strongly discouraged given the limited evidence of the intervention’s effectiveness, the high cost and the potential wastage
of resources. The GDG therefore felt it necessary to develop a clear recommendation against space spraying for malaria
control.
Research Needs
• Determine the impact (incidence of malaria (infection or clinical) and/or prevalence of malaria infection) and potential
harms/unintended consequences of space spraying, particularly in emergency situations.
83 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Screening of residential houses can be used for the prevention and control of malaria in children and adults in areas with
ongoing malaria transmission.
The GDG determined that a conditional recommendation should be given for house screening because of the low- to moderate-
certainty evidence of an impact against malaria. Furthermore, programmes would need to consider a number of local contextual
factors when considering screening of residential houses as a public health strategy, such as:
Programmes should note that this recommendation addresses the use of screening of windows, ceilings, doors and/or eave spaces,
and does not cover other ways of blocking entry points into houses.
Practical Info
If house screening is being considered as a means to prevent malaria, it is important to identify who the end-user will be and
how the intervention will be implemented, i.e. whether screening of houses will be a tool that the programme promotes for
individuals or communities to implement at their own cost, or whether it will be undertaken as a programmatic initiative.
Depending on the approach, the resources needed, feasibility, uptake and impact on equity may vary and would need to be
considered.
Screening of houses may be done post-construction or could be a standard feature for new homes. Intersectoral
collaboration, for example, between health, housing and environmental sectors, is crucial in the implementation of house
screening. It is also important to consider what standards and criteria, if any, need to be set for screening materials and
designs, as they are for buildings.
Screening of residential houses should be part of an IVM approach as promoted under the GVCR [16]. Deployment of
interventions recommended for large-scale deployment (such as ITNs or IRS) should be maintained, and communities should
be encouraged to continue using ITNs regularly or allow their houses to be sprayed, even if screening has been installed.
In settings where national or local government authorities are not able to provide screening of residential houses as a public
health strategy (e.g. due to feasibility/resource challenges), they should promote its use in affected communities.
If house screening is deployed or adopted by communities to prevent malaria, post-distribution monitoring of the
intervention is needed to assess material durability, usage and coverage. This information should guide how regularly
screens require replacement or repair and provide information on the sustainability of the intervention.
Evidence To Decision
The systematic review noted from a pooled analysis of the two studies that individuals living in screened houses
(covered eaves, windows and doors) were 16% less likely to sleep under a mosquito net (risk ratio: 0.84; 95% CI:
0.65–1.09; two trials, 203 participants). However, the results from the two studies were discrepant: In the Federal
Democratic Republic of Ethiopia, the study [86] found no difference in ITN use in screened or unscreened homes, while
84 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
the study [87] in the Republic of the Gambia found that reported use of ITNs was lower in houses with screened ceilings
(26%, 70/272) than in control houses (35%, 57/162; p=0.04). In the Gambian study, the number of mosquitoes in the
house were reduced, which could have resulted in fewer participants feeling the need to use a net to prevent biting.
None of the other pre-specified outcomes (all-cause mortality; other disease incidence; adverse effects; unintended
effects other than bed net usage) were reported in the included studies.
Based on the evidence presented in the review, the GDG judged that in some settings there may be potential
undesirable effects associated with house screening; however, all of the potential effects identified by the GDG were
judged to be small:
• Inhabitants of screened houses may stop or reduce their use of other effective interventions such as
ITNs, especially if house screening is perceived to greatly reduce mosquito entry and/or be sufficient alone to
protect against malaria. The decline or discontinuation in the use of interventions is likely not limited to those
deployed with house screening; if any intervention that is deployed in conjunction with another is perceived to be
sufficiently effective alone, use of the co-deployed intervention may decline.
• Screening of available entry points for mosquitoes into the house may result in reduced airflow and ventilation, and
increased indoor temperatures compared to unscreened openings. While the GDG remarked that, as a result,
occupants may open doors and windows (thereby negating the benefit of screening and, in turn, increasing the risk
of mosquito exposure), in Côte d’Ivoire this was not the case. Households with screened openings did not differ
from those with no screening in terms of opening and closing windows [88]. Reduced airflow and ventilation has
been shown to result in increased respiratory problems and infections [89] and increased indoor air pollution, which
negatively affects human health [90][91][92]. However, if household inhabitants routinely close entry points at
night, such as windows, screening these openings would allow for increased airflow and ventilation compared to
when they are closed, thereby reducing indoor temperatures as shown in the Republic of the Gambia [93][94].
The systematic review assessed that the overall certainty of the evidence that house screening has an impact on malaria
was low.
Resources
Resources needed for the screening of houses may depend on whether the intervention is deployed by the programme
or implemented by the community. The table below, compiled by the GDG, lists resources that should be considered.
Note that this table does not include resource needs for product selection or assessment of impact of the intervention.
85 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• Adequate construction material for screening (including but not limited to wood/
Supplies screen, fasteners)
• BCC materials (e.g. flip charts, posters, banners, staff clothing)
• M&E data collection forms
• Construction tools/equipment
Equipment
• Computer/communication equipment
• Construction/installation supervisors
Governance/ programme
• BCC supervision
management
• M&E survey support for coverage
Equity
National programmes considering the adoption of screening of residential houses as a public health strategy should
assess how the implementation of a screening programme would affect health equity in the community. Depending on
how the intervention is deployed, the effect on equity may vary. For example, if individuals are encouraged to screen
houses themselves, equity may be reduced. If the intervention is deployed at the programme level, it may be increased.
The impact on equity may also depend on house structure and conditions, as some features may not allow for screening.
Acceptability
The studies included in the systematic review used in-depth interviews and focus group discussions to assess
community acceptance of the intervention. In both studies, participants reported that the intervention reduced the
number of indoor mosquitoes and house flies. Most participants in both trials chose to have screening after the duration
of the trial. Additionally, participants in the study from the Republic of the Gambia reported a reduction in entry of other
animals, such as bats, cockroaches, earwigs, geckos, mice, rats, snakes, and toads. In both trials, participants expressed
concern that screening would be damaged by domestic animals and children, or that it would become dirty. In the
Ethiopian study, some participants reported that they made further efforts to reduce mosquito entry after screening
installation, such as filling in wall openings with mud.
Feasibility
National programmes considering the adoption of screening of residential houses as a public health strategy should
86 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
assess:
• whether the structure and condition of the residential houses in the community allow for the installation of
screening and are accessible;
• whether adequate resources are available, particularly if houses require screening to be made bespoke and if there
is a need to renovate some houses to enable screening;
• the level of community buy-in (acceptability and/or willingness to implement the intervention);
• the feasibility of implementation if it is on a large scale, including the impact on resource use and potential changes
in cost-effectiveness of the programme, and also taking into account the values, preferences and cultural norms of
the main stakeholders; and
• how the intervention will be delivered and maintained.
Justification
The systematic review [85] identified only two eligible published studies assessing the impact of housing modifications on
malaria epidemiological outcomes conducted in the Federal Democratic Republic of Ethiopia and the Republic of the
Gambia . Both studies investigated the impact of house screening (screening of windows, ceilings, doors and/or eaves) with
untreated materials against malaria. The authors concluded that screening may reduce clinical malaria incidence, parasite
prevalence, prevalence of anaemia and EIR. In the trials included in the systematic review, research teams deployed
screening at the community level and, as a result, there is currently no evidence as to the benefits and harms of individuals
or communities deploying screens themselves. The review identified several studies that were yet to be published on the
efficacy of insecticide-treated screening, eave tubes or other forms of housing modifications, but the data were not available
at the time for inclusion in the review.
Given that only two trials were included in the review, a number of potential effect modifiers could not be examined, and the
generalizability of the findings was limited. The panel concluded that untreated screening of residential houses may prevent
malaria and reduce malaria transmission, and that these desirable effects would outweigh the undesirable effects. However,
in translating this evidence into a recommendation strength, the GDG concluded that the recommendation should be
conditional due to the low- to moderate-certainty evidence and based on a number of contextual factors. The panel judged
that policy-makers considering house screening should assess the feasibility, acceptability, impact on equity and resources
needed for screening houses in their contexts in order to determine whether such an intervention would be appropriate for
their setting.
Research Needs
WHO encourages funding of high-quality research on the impact of interventions under the broad category of “housing
modifications” to further inform the development of specific WHO recommendations. Results from four trials awaiting
publication are likely to enrich the current evidence base on housing modifications for preventing malaria and controlling
malaria transmission. Publication of these studies is strongly encouraged.
• Further evidence is needed on the impact (incidence of malaria [infection or clinical] and/or prevalence of malaria
infection) and potential harms/unintended consequences of house screening, as well as other housing modification
interventions deployed alone or in combination.
• Epidemiological evidence is required on the efficacy against malaria of the same intervention implemented in different
settings (where vector species may differ).
• Evidence is needed on contextual factors (i.e. acceptability, feasibility, resource use, cost-effectiveness, equity, values
and preferences) related to house screening, as well as other housing modification interventions.
• Determine the resource needs, costs and cost-effectiveness of various deployment options for house screening (at the
programme, community and individual level).
• Develop deployment mechanisms and foster community buy-in for house screening and other housing modification
interventions.
87 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
intervention. If the initial two studies generate contradictory and potential harms/unintended
or inconsistent results or suffer from design limitations that consequences of pyrethroid-PBO
preclude comprehensive assessment of an intervention’s nets from areas where the
potential public health value, further trials with mechanisms of resistance in vector
epidemiological endpoints may be required. As such, WHO species are not oxidase-based and
encourages the use of appropriate study designs, including the from areas of lower malaria
generation of baseline data and appropriate follow-up times transmission intensity.
that consider the characteristics of the intervention and its
intended deployment, expected durability/residual efficacy and Further evidence is needed on the
replacement intervals, and the epidemiology (e.g. pathogen durability of pyrethroid-PBO nets.
transmission intensity) of the selected study site. WHO
Determine the impact (incidence of
encourages studies to be conducted for durations that
malaria [infection or clinical] and/
maximize the likelihood that the study objectives and targeted
ITNs in or prevalence of malaria infection)
statistical power will be robustly achieved so as to strengthen
humanitarian and potential harms/unintended
the evidence used to inform deliberations by a GDG regarding
emergencies consequences of ITNs in the acute
a potential WHO recommendation. Detailed descriptions of
phase of humanitarian
the setting, interventions deployed, and vector species
emergencies (where logistics and
targeted are required. Investigators are encouraged to share
priorities may differ).
their study design and methodology with WHO prior to
commencing the study in order to enable the VCAG to validate Further evidence is needed on the
whether the data generated are likely to provide quality impact (incidence of malaria
evidence to inform the development of a WHO [infection or clinical] and/or
recommendation. High research standards should be employed prevalence of malaria infection)
in conducting, analysing and reporting studies, ensuring that and potential harms/unintended
studies are adequately powered, and appropriate consequences of IRS.
randomization methods and statistical analyses are used.
WHO requires studies to be conducted in compliance with Determine the impact (incidence of
international ethical standards and good clinical and laboratory malaria [infection or clinical] and/
practices. Further information on evaluation standards for or prevalence of malaria infection)
vector control interventions can be found in Norms, standards and potential harms/unintended
and processes underpinning WHO recommendations on vector consequences of IRS in urbanized
control [95]. areas with changing housing
designs.
88 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
89 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
90 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
the intermittent preventive treatment of malaria in pregnancy To support decision making, each chemoprevention
(IPTp), perennial malaria chemoprevention (PMC), previously recommendation is accompanied by a summary of available
known as intermittent preventive treatment in infants (IPTi), research evidence, an explanation of how this was used to
seasonal malaria chemoprevention (SMC), intermittent inform the recommendation and practical information regarding
preventive treatment in school aged children (IPTsc), post- key considerations for implementation.
discharge malaria chemoprevention (PDMC) and mass drug
administration (MDA) for malaria burden and transmission Protection for travellers to malaria-endemic areas
reduction, and mass relapse prevention. Each of these The primary target for these guidelines is people living in
recommendations reflects the biological plausibility that a endemic areas and no formal recommendations regarding
treatment course of an effective antimalarial will clear any preventive chemotherapy are currently included for non-immune
existing, and prevent new, malaria infections. This underlying people travelling to malaria endemic regions.
principle can inform the adaptation of recommendations to
People growing up in endemic countries will increasingly be non-
maximise impact in different settings.
immune as malaria control improves. However, epidemiological
The updated chemoprevention recommendations reflect the changes will be heterogeneous and future guidelines will need to
paradigm shift, outlined in the introduction, to provide greater consider the use of chemoprophylaxis among people growing up
flexibility to NMPs to adapt control strategies to suit their in areas without malaria (e.g. some urban settings) who then
settings. Standard processes have been used to develop travel within their own country to places where malaria is
evidence-based recommendations which are not unduly endemic (e.g. many rural settings). The potential of
restrictive. We no longer specify strict age groups, transmission chemoprophylaxis for people at risk of occupational exposure to
intensity thresholds, numbers of doses or cycles, or specific malaria (e.g. farmers, miners) also warrants consideration.
drugs. The effectiveness of a chemoprevention programme will Readers interested in the use of antimalarial agents to prevent
be influenced by a host of contextual and other factors (e.g. malaria in people travelling from non-endemic settings to areas
intensity of malaria transmission, extent of seasonal variation in of malaria transmission are directed to the WHO International
transmission, the age group targeted by the chemoprevention travel and health guidance [2].
programme, the preventive efficacy of the drugs used, the
In summary, travellers should start chemoprophylaxis before
frequency of dosing, duration of protection of each treatment
entering an endemic area, to assess tolerability and, for slowly
course, availability of drugs, coverage achieved, adherence to the
eliminated drugs, to build up therapeutic concentrations. Malaria
recommended regimen) and by the mix of interventions being
may be prevented by taking drugs that inhibit liver-stage (pre-
deployed in each setting. NMPs are therefore encouraged to
erythrocytic) development (causal prophylaxis) or drugs that kill
consider local data to determine how best to tailor
asexual blood stages (suppressive prophylaxis). Causal
chemoprevention strategies to local needs and determine which
prophylactics (atovaquone + proguanil) can be stopped soon
age groups should be prioritized where, for how long, how
after leaving an endemic area, whereas suppressive
frequently, and with which drugs. Subnational tailoring is
prophylactics must be taken for at least 4 weeks after leaving
increasingly needed, for example to recognize the variation in
the area in order to eliminate asexual parasites emerging from
duration of the transmission season even within a country,
the liver weeks after exposure.
meaning that 3, 4, 5 or more cycles of SMC may be warranted in
different subnational areas.
91 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
In malaria-endemic areas, pregnant women of all gravidities should be given antimalarial medicine at predetermined
intervals to reduce disease burden in pregnancy and adverse pregnancy and birth outcomes.
• Sulfadoxine-pyrimethamine (SP) has been widely used for malaria chemoprevention during pregnancy and remains effective in
improving key pregnancy outcomes.
• IPTp-SP should start as early as possible in the second trimester and not before week 13 of pregnancy.
• Doses should be given at least one month apart, with the objective of ensuring that at least three doses are received.
• Antenatal care (ANC) contacts remain an important platform for delivering IPTp. Where inequities in ANC service and reach
exist, other delivery methods (such as the use of community health workers) may be explored, ensuring that ANC attendance is
maintained and underlying inequities in ANC delivery are addressed.
• IPTp is generally highly cost-effective, widely accepted, feasible for delivery and justified by a large body of evidence generated
over several decades.
Practical Info
Antimalarial medicine
WHO recommends that the medicines used for IPTp be different from those used as first-line malaria treatment. SP has
been widely used for chemoprevention during pregnancy and has been shown to be efficacious, safe, well tolerated,
available and inexpensive. A drug regimen that can be administered as a directly observed single dose, such as SP, is
preferable to a multi-day regimen.
The Guideline Development Group did not formally consider alternative drug regimens to SP for IPTp, or their associated
costs. However, recent studies of dihydroartemisinin-piperaquine (DHAP) in areas of high SP resistance have shown that,
although superior to SP in reducing malaria during pregnancy, the use of DHAP did not translate into better pregnancy
outcomes; SP was associated with better fetal growth, resulting in higher mean birthweights in all gravidae (Gutman et al
unpublished evidence (a)).
Transmission
In areas of moderate to high P. falciparum transmission, IPTp-SP should be given to all pregnant women. Whether there
continues to be a role for IPTp in areas where malaria transmission has fallen to low levels is uncertain. There is evidence
that even in areas with PfPR2-10 < 3%, IPTp-SP reduces maternal anaemia and may reduce low birthweight, as well as
maternal and placental infection (Gutman et al unpublished evidence (a)). Some of these effects may not be due to the effects
of IPTp-SP on malaria. There is currently insufficient data to define the level of transmission below which IPTp-SP may cease
to be cost-effective. Challenges of IPTp reintroduction after withdrawal caution against discontinuing IPTp-SP following a
recent reduction in malaria transmission.
Pregnancy
IPTp improves a wide range of outcomes in women in their first and second pregnancies, including maternal and placental
infection, maternal anaemia and low birthweight (Gutman et al unpublished evidence (a)). There is now evidence that IPTp also
reduces maternal infection in third or subsequent pregnancies, but there are currently too few trials to evaluate effects on
other outcomes in these women (Gutman et al unpublished evidence (a)). Administering IPTp to all pregnant women regardless
of number of pregnancies facilitates ease of IPTp implementation for health workers.
Dosage
IPTp-SP should ideally be administered as directly observed therapy (DOT) with three tablets of SP (each tablet containing
500 mg/25 mg SP), for the total required dosage of 1500 mg/75 mg SP.
Schedule
IPTp-SP should not be given before week 13 of pregnancy due to an increased risk of fetal malformation. IPTp-SP should
start in the second trimester and doses should be given at each scheduled ANC contact until the time of delivery, provided
that doses are at least one month apart. At least three doses of IPTp-SP should be received during pregnancy.
Delivery
ANC contacts remain an important platform for delivering IPTp, and so inequities in ANC service and reach should be
addressed. Research on alternative approaches to IPTp delivery (e.g. through community health workers) may identify
92 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
opportunities to increase coverage, while ensuring that ANC attendance is maintained. This may be useful for supporting
IPTp delivery while measures to address ANC inequities are implemented. Consideration should be given to contextual
factors such as the values and preferences of end-users, costs, coverage and sustainability of alternative delivery platforms.
Drug resistance
IPTp-SP appears to select for antifolate resistance mutations associated with low to moderate increases in drug resistance.
However, there is no convincing evidence of selection favouring key mutations, such as dhpsA581G, which is associated
with the loss of IPTp-SP efficacy (Plowe unpublished evidence). There is also insufficient evidence to withhold IPTp-SP in
areas where the prevalence of dhpsA581G exceeds a threshold of 10% (Plowe unpublished evidence). Although the ability of
IPTp-SP to clear existing infections and prevent new ones is compromised in areas of high to very high resistance, the
intervention still reduces low birthweight and maternal anaemia. Consequently, IPTp-SP should continue to be used in areas
of high SP resistance until more effective alternatives for malaria chemoprevention are found.
Contraindications
IPTp is not recommended for pregnant women before week 13 of pregnancy, or those with severe acute illness, or who are
unable to take oral medication, or women who during the last 30 days received a dose of any of the drugs being used for
IPTp, or those allergic to any of the components of SP. IPTp-SP should not be given to individuals receiving a sulfa-based
medicine as treatment or prophylaxis, including co-trimoxazole (trimethoprim–sulfamethoxazole) for HIV. High doses of folic
acid (daily dose ≥ 5 mg) have been shown to counteract the efficacy of SP as an antimalarial, and only low-dose formulations
(i.e. 0.4 mg daily) should be co-administered with SP.
Other considerations
Information about IPTp should be fully accessible to pregnant women. As with all health interventions, consent should be
obtained from the pregnant woman prior to administering IPTp.
Evidence To Decision
• Anaemia: IPTp-SP may reduce maternal anaemia (risk ratio: 0.90; 95% CI: 0.87–0.93; low-certainty evidence) and
increase maternal haemoglobin (mean difference: 0.19 g/dL higher; 95% CI: 0.15–0.22 g/dL higher; low-certainty
evidence) for each dose of SP in all gravidae. The effect is lower but remains significant in the highest SP resistance
areas1 (relative risk reduction: 8.2%; 95% CI: 3–13%). IPTp-SP also reduced maternal anaemia in areas with
PfPR2-10 < 3% (risk ratio: 0.91; 95% CI: 0.85–0.97).
• Placental and maternal malaria infection at delivery: IPTp-SP probably reduces placental infection (risk ratio: 0.78;
95% CI: 0.74–0.84; moderate-certainty evidence) and maternal malaria infection at delivery (risk ratio: 0.80; 95%
CI: 0.75–0.85; moderate-certainty evidence) for each dose of SP in all gravidae, compared to no IPTp-SP. Overall,
IPTp-SP was associated with a 20% reduction (95% CI: 16–24%) in placental or maternal malaria at delivery
compared to no IPTp-SP. The effect was greater in first and second pregnancies (24%; 95% CI: 19–29%) than in
third or subsequent pregnancies (17%; 95% CI: 13–20%). There was a trend towards reduced efficacy with
increased resistance, with a relative risk of 28% (95% CI: 20–36%) in the lowest resistance stratum and 22% (95%
CI: 14–29%), 8% (95% CI: 0–7%) and -5% (95% CI: -16–5%) in the moderate, high and very high resistance strata,
respectively. The effect of IPTp-SP in areas with PfPR2-10 < 3% was variable (risk ratio for maternal malaria: 0.73;
95% CI: 0.53–1.01; and for placental malaria: 0.89; 95% CI: 0.68–1.15).
• Adverse events: IPTp-SP had a pooled prevalence of serious adverse events of 3.84% (95% CI: 2.20–5.88%) and a
pooled prevalence of adverse events of 14.3% (95% CI: 4.9–27.5%). In two trials comparing IPTp-SP to placebo or
case management, the pooled risk ratio showed that IPTp-SP may reduce maternal adverse events (risk ratio: 0.56;
95% CI: 0.30–1.01; moderate-certainty evidence). Skin reactions were rarely reported, with a pooled prevalence of
0.4% (95% CI: 0.2–0.7%) among all women who took IPTp-SP and with no significant increase in the two trials
comparing IPTp-SP to placebo or case management (pooled risk ratio: 1.24; 95% CI: 0.34–4.58).
• Maternal death: The effect of IPTp-SP on maternal death is poorly documented. It is possible that IPTp-SP results in
little to no difference in maternal death (risk ratio: 1.17; 95% CI: 0.49–2.80; low-certainty evidence).
None of the studies in the systematic review reported on malaria infection, severe malaria, or maternal hospitalization.
93 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• Birthweight: IPTp-SP probably reduces low birthweight for each dose of SP compared to no IPTp-SP (risk ratio:
0.75; 95% CI: 0.71–0.78; low-certainty evidence) for all gravidae. The point estimate is slightly higher in first and
second pregnancies (26%; 95% CI: 21–31%) than in third or subsequent pregnancies (21%; 95% CI:
16–26%). Compared to no IPTp-SP, each dose of IPTp-SP probably increases mean birthweight for babies born to
women of all gravidae (mean difference: 57 g higher; 95% CI: 44–69 g; moderate-certainty evidence). IPTp-SP was
associated with a mean increase in birthweight of 67 g (95% CI: 50–85 g) in babies born to women in their first and
second pregnancies and 43 g (95% CI: 26–60 g) in third or subsequent pregnancies. The relative risk reduction in
low birthweight decreased with increasing SP resistance, remaining significant in high-resistance areas (relative risk
reduction: 23%; 95% CI: 16–29%), but becoming non-significant in the highest SP resistance areas (relative risk
reduction: 16%; 95% CI: -4–32%). Mean difference in birthweight was 65 g (95% CI: 44–87 g), 66 g (95% CI: 45–88
g) and 46 g (95% CI: 27–66 g) in the lowest, middle and high SP resistance areas, respectively. There was a non-
significant mean difference of 11 g (95% CI: -9–32 g) in the highest resistance areas.
• Adverse pregnancy outcomes: Each dose of IPTp-SP may reduce preterm delivery compared to no IPTp-SP (risk
ratio: 0.76; 95% CI: 0.71–0.81; very low-certainty evidence). However, the evaluation of preterm delivery and
number of SP doses is complicated because prematurity inherently reduces the opportunity to receive more SP
doses. It is uncertain whether IPTp-SP reduces stillbirths and spontaneous abortions compared to no IPTp-SP (risk
ratio: 0.68; 95% CI: 0.59–0.78; very low-certainty evidence).
None of the studies in the systematic review reported on malaria infection, anaemia, severe malaria, hospital
admissions, or death.
More information on the evidence can be found in the systematic review (Gutman et al unpublished evidence (a)).
1 Resistance was defined as low (Ala437Gly < 75% in Central/West Africa or Lys540Glu < 40% in Eastern/Southern Africa), medium (Ala437Gly ≥ 75%
in Central/West Africa or Lys540Glu 40–60% and AlaA581Gly < 5% in Eastern/Southern Africa), high (Lys540Glu ≥ 60 & Ala581Gly < 5% in Eastern/
Southern Africa) and very high (Lys540Glu ≥ 60% and dhps Ala581Gly ≥ 5% in Eastern/Southern Africa).
The certainty of evidence across the outcomes ranged from very low to moderate, with a number of the outcomes
deemed important by the GDG classed as moderate-certainty evidence. The GDG noted sustained impact of IPTp-SP
across all transmission and resistance settings. Consequently, the overall certainty of evidence for the outcomes of
interest was considered moderate by the GDG. This reflects the large number of observational studies contributing
useful information to these updated guidelines, building on the initial more robust data from randomized controlled
trials.
More information on the certainty of evidence assessments can be found in the ‘research evidence’ tab associated with
this recommendation online or in the annex of the pdf version.
• consultation with civil society, which indicated that chemoprevention to prevent malaria disease in pregnant
women was seen as a priority in endemic areas (CS4ME unpublished evidence);
• a synthesis of contextual factors from studies of IPTp-SP, although these lacked data on how IPTp-SP was valued
(Rodriguez et al unpublished evidence).
The GDG vote on values and preferences was equally split between “probably no important uncertainty or variability”
and “possibly important uncertainty or variability” in how the outcomes of IPTp are valued across contexts. The vote was
repeated and remained split. Those who voted for the latter felt that IPTp may be valued differently depending on the
transmission and resistance context. The consensus of the GDG was not to say that values and preferences vary but
rather to highlight the two positions.
94 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
More information can be found in the civil society consultation report (CS4ME unpublished evidence).
Resources
An individually randomized, placebo-controlled trial in a moderately intense transmission setting in Mozambique found
IPTp-SP to be a highly cost-effective intervention [96]. Based on data from 2007, the financial cost of delivering two
doses of IPTp-SP through ANC was about US$ 435.79 per 1000 pregnant women. Delivering two doses of IPTp-SP to
1000 pregnant women resulted in a total health system cost saving of US$ 422.74, 43% of which was attributed to
reduced hospital admissions. Consequently, the net intervention cost was US$ 13.17 per 1000 pregnant women. IPTp-
SP led to substantial household cost savings for women seen in the outpatient department (US$ 33.89 in direct costs;
95% CI: 6.10–77.20; and US$ 83.79 in indirect costs; 95% CI: 29.60–148.30). However, it did not lead to statistically
significant household cost savings for women who required admission for malaria (US$ 8.20 in direct costs; 95% CI:
-42.80–55.80; and US$ 11.44 in indirect costs; 95% CI: -20.50–42.70). Delivering IPTp-SP to 1000 pregnant women
was expected to avert 18.9 (95% CI: 4.4–33.8) neonatal deaths, or 555.2 (95% CI: 129.0–992.0) disability-adjusted life
years (DALYs). This study determined threshold values of some variables beyond which IPTp-SP was no longer cost-
effective. These were when ANC attendance is lower than 37.5%, the protective efficacy of IPTp-SP against maternal
infection is lower than 15%, maternal clinical malaria incidence is lower than 0.15 person-year at risk, or the maternal
case fatality ratio is lower than 0.15%.
Based on the data from Mozambique, the intervention costs of delivering two doses of IPTp-SP were US$ 41.46 per
DALY averted versus US$ 7.28 per DALY averted for three doses [96][97]. The cost of one dose of IPTp-SP was reported
to be between US$ 0.63 and US$ 0.79 [97][98].
The GDG considered that there were negligible costs and savings associated with implementing IPTp-SP and the
certainty of the evidence on the resources required was moderate. The GDG determined that IPTp is probably cost-
effective compared to no intervention.
More information on the evidence can be found in the summary of contextual factors report (Rodriguez et al unpublished
evidence).
Equity
Age, marital status, religion, and living in a rural area were found to influence the uptake of IPTp-SP in 13 studies.
Women under 20 years old were generally the least likely to receive three doses of IPTp-SP, with those between 25 and
34 most likely to receive IPTp-SP. Socioeconomic considerations including education level, employment status and
wealth index affected uptake of IPTp. Higher uptake was associated with being married and higher education, and some
studies found a strong association between employment status and IPTp-SP uptake. Many studies reported that women
in the “middle” to “richest” wealth index had higher uptake of IPTp-SP compared to those in the “poorest” to “poorer”
wealth categories, including receipt of at least three doses of IPTp-SP. Rural residence was inconsistently associated with
improved IPTp-SP uptake. Studies conducted in Burkina Faso, Côte d’Ivoire, and Sierra Leone reported that women who
lived in rural areas were more likely to take the recommended doses of IPTp-SP, while studies in Ghana, Malawi and
Nigeria reported that urban residence was associated with higher IPTp-SP uptake compared to rural residence. Living
more than 5 km from a health facility was also associated with poorer uptake of IPTp-SP.
The GDG considered that the health equity of IPTp varies depending on contextual factors, especially those influencing
access to ANC services. IPTp programmes that address inequities will likely improve coverage of IPTp and improve
pregnancy outcomes.
More information on the evidence can be found in the summary of contextual factors report (Rodriguez et al unpublished
evidence).
Acceptability
IPTp has been widely accepted by pregnant women. Greater knowledge about IPTp has been shown to increase
acceptance and uptake of the intervention. ANC attendance is a main driver influencing patient acceptance of IPTp-SP.
95 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Numerous studies have reported increased uptake of IPTp-SP with early initiation of education and counselling sessions
at ANC, specifically during the first trimester, as well as frequent ANC contacts. In general, women who were concerned
about the side effects of SP were less likely to take the recommended number of doses of IPTp-SP.
More information on the evidence can be found in the summary of contextual factors report (Rodriguez et al unpublished
evidence).
Feasibility
Limited knowledge and training of staff on the prevention and management of malaria in pregnancy, including
indications for IPTp-SP, contribute to poor uptake. Some health care workers expressed concerns over the lack of
ongoing training to update their knowledge, although this was country- and site-dependent. Other issues that impaired
the delivery of IPTp included stockouts of SP, under-prescribing of SP (< three doses), and inadequate staffing. DOT was
generally, but not always, associated with improved uptake of IPTp-SP. Utilization of DOT was variable, with between
5% and 67% of pregnant women reporting taking IPTp-SP under DOT [99][100][101][102].
The GDG considered IPTp implementation to be feasible, given that it is delivered through ANC.
More information on the evidence can be found in the summary of contextual factors report (Rodriguez et al unpublished
evidence).
Justification
This recommendation was developed using the Grading of Recommendations Assessment, Development and Evaluation
(GRADE) framework [103].
Sources of information
Recommendation development was informed by a systematic review (Gutman et al unpublished evidence (a)) and a report
summarizing evidence from published studies on contextual factors related to IPTp implementation (Rodriguez et al
unpublished evidence), including cost-effectiveness, feasibility, equity, values and acceptability. These sources of information
were supplemented by a cross-cutting review on chemoprevention and drug resistance (Plowe unpublished evidence), a civil
society consultation report on chemoprevention (CS4ME unpublished evidence) and contributions from the GDG
membership, which included former and current national malaria programme representatives. The GDG was supported by a
Steering Group, which included representatives from the WHO Departments for Sexual and Reproductive Health and
Research and Child Health and Development.
The systematic review addressed the GDG’s PICO (population, intervention, comparison, outcome) question regarding
whether women of all gravidities should be given SP as malaria chemoprevention to reduce disease burden in pregnancy
and/or adverse pregnancy and birth outcomes. In particular, the systematic review assessed the potential modifying effects
of gravidity, malaria transmission intensity, and SP resistance on the effectiveness of IPTp-SP.
The main outcomes of interest considered by the GDG in the systematic review were maternal anaemia and low birthweight.
Other outcomes of interest included maternal clinical malaria, placental infection, malaria infection, severe malaria, adverse
events, hospitalization, and death; and fetal/infant adverse pregnancy outcomes (spontaneous abortion, stillbirth or preterm
delivery), malaria infection, anaemia, severe malaria, hospital admissions, and death. Overall, 102 studies and 105 276
participants contributed to the systematic review. This included seven trials comparing IPTp-SP to placebo or passive case
detection, 12 trials or cohorts following women who received IPTp-SP, and 83 observational studies. The studies covered all
gravidae. All the included studies were conducted in sub-Saharan Africa, with more studies situated in Central and West
Africa (59.3%) than in Eastern and Southern Africa (40.7%). Given that IPTp is an intervention that has proven to be
effective, for ethical reasons, no new placebo-controlled trials have been conducted since the last update to the IPTp
recommendation. This review therefore included a large number of observational studies.
Summary of judgements
The Evidence-to-Decision framework captures the evidence from the systematic review considered by the GDG. The GDG
determined that the balance between desirable and undesirable effects favoured IPTp; negligible costs and savings were
associated with IPTp implementation delivered through ANC contacts; the certainty of the evidence on required resources
was moderate; and IPTp was probably cost-effective, probably acceptable to key stakeholders, and feasible to implement.
96 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The GDG concluded that a strong recommendation should be made for IPTp based on its moderate beneficial effects, small
undesirable effects, and moderate-certainty evidence.
Implementation
Please refer to the WHO policy brief for the implementation of intermittent preventive treatment of malaria in pregnancy using
sulfadoxine-pyrimethamine (IPTp-SP) [100] and the WHO recommendations on antenatal care for a positive pregnancy
experience [101]. A manual for subnational tailoring of malaria interventions is under development and expected for
publication in 2022.
Evaluation
The safety and impact of IPTp programmes should be routinely monitored. The effect of IPTp may be evaluated using routine
data on hospital deliveries, clinic and/or community health worker data.
The WHO chemoprevention efficacy study (CPES) protocol should be used to monitor the efficacy of medicines used for
chemoprevention. Although the potential effect of chemoprevention on the spread of drug resistance may be monitored by
the analysis of molecular markers associated with treatment outcomes, the correlation between molecular markers and the
efficacy of antimalarials for chemoprevention is unclear and results should be interpreted with caution. Given that SP
continues to have positive outcomes for mother and baby even in areas of very high SP resistance, national malaria
programmes may want to continue IPTp-SP programmes, despite worsening efficacy on malaria-specific outcomes.
Research Needs
Several evidence gaps were identified regarding IPTp. None should prevent adoption and implementation of IPTp.
Nevertheless, impact could potentially be enhanced by determining:
• the effectiveness of alternative drug regimens for IPTp, including SP + diydroartemisinin-piperaquine (DHAP);
• the non-malarial effect of SP on pregnancy outcomes2 ;
• the effectiveness of alternative approaches to IPTp delivery (e.g. community-based approaches) to improve uptake and
address inequities in coverage compared to comparable investment in ANC services.
Data on the safety and effectiveness of alternatives to SP for IPTp will be reviewed by WHO when the relevant meta-
analyses are available.
2 Despite a near complete loss of its antimalarial effects in areas of very high SP resistance, SP continues to positively impact fetal growth and maternal
anaemia. This may be mediated through a non-malarial pathway. This is consistent with the results of an individual patient data meta-analysis, including
data from six trials comparing IPTp with DHAP vs IPTp with SP. These data showed that IPTp with DHAP was much more effective than SP in reducing
malaria in areas of high SP resistance. However, this did not translate into better pregnancy outcomes, primarily because SP was associated with better fetal
growth and thus higher mean birthweights in all gravidae. This may reflect the broad-spectrum antimicrobial properties of sulfadoxine, a long-acting
sulfonamide, and the associated reduced risk of persistent bacterial infections, and/or its influence on the maternal gut microbiome, and/or its ability to
reduce inflammation (Gutman et al unpublished evidence (a)).
97 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
In areas of moderate to high perennial malaria transmission, children belonging to age groups at high risk of severe malaria
can be given antimalarial medicines at predefined intervals to reduce disease burden.
• Perennial malaria chemoprevention (PMC) schedules should be informed by the age pattern of severe malaria admissions, the
duration of protection of the selected drug, and the feasibility and affordability of delivering each additional PMC course (see
“Practical info”).
• Sulfadoxine-pyrimethamine (SP) has been widely used for chemoprevention in Africa, including for PMC. Artemisinin-based
combination therapies (ACTs) have been effective when used for PMC, but evidence is limited on their safety, efficacy,
adherence to multi-day regimens, and cost-effectiveness in the context of PMC.
• Previously, PMC was recommended in infants (<12 months of age) as intermittent preventive treatment in infants (IPTi). Since
the initial recommendation, new data have documented the value of malaria chemoprevention in children aged 12 to 24
months.
• The Expanded Programme on Immunization (EPI) platform remains important for delivering PMC. Other methods of delivery
can be explored to optimize access to PMC and integration with other health interventions.
• Moderate to high perennial malaria transmission settings are defined as areas with P. falciparum parasite prevalence greater
than 10% or an annual parasite incidence greater than 250 per 1000 [30]. These thresholds are indicative and should not be
regarded as absolutes for determining applicability of the PMC recommendation.
Practical Info
Antimalarial medicine
WHO recommends that medicines used for PMC be different from those used as first-line malaria treatment. SP has been
widely used for chemoprevention in Africa and has been shown to be efficacious, safe, well tolerated, available and
inexpensive. SP was evaluated in 10 trials for PMC, artesunate-amodiaquine (AS+AQ) in one trial, DHAP in one trial, and
sulfadoxine-pyrimethamine + artesunate (SP+AS) in one trial [106]. All regimens were found to be effective in reducing
clinical malaria. Although ACTs have been effective when used for PMC, evidence is limited on their safety (including
potential cumulative toxicity), efficacy, adherence to multi-day regimens, and cost-effectiveness in the context of PMC in
young children. A drug regimen that can be administered as a directly observed single dose, such as SP, is preferable to
multi-day regimens.
Age group
The target age group should be identified using local data on the age distribution of malaria admissions and severe disease.
Previously, PMC was recommended in infants (<12 months of age) as IPTi based on evidence generated in this age group
and an appreciation of the disease burden they bear. Since the initial recommendation, additional data have documented the
value of malaria chemoprevention in children aged 12 to 24 months. Three studies evaluated PMC doses in children aged 12
to 15 months [107][108][109], and one study evaluated monthly doses in children up to 24 months [110]. Evidence from
seasonal malaria chemoprevention (SMC) programmes, where the age of the target population overlaps with that of PMC,
also shows that the impact of chemoprevention on disease burden can be sustained beyond infancy with additional doses.
However, there is limited information on the safety and efficacy of malaria chemoprevention in children >15 months of age
in perennial transmission settings.
Dosage
Children in age groups at increased risk of severe disease should be given a complete course of antimalarials, at their
recommended treatment dose, as PMC. The drug dosage should be determined by the child’s weight wherever possible,
with dosing according to age only in situations where the child’s weight is unknown.
Frequency
The PMC schedule should be informed by the length of protective efficacy of the selected drug, as well as the feasibility of
delivering each additional PMC course. SP doses should be given at least one month apart. Eight trials have evaluated a
range of 3–6 doses of SP for PMC in the first year of life. Four trials have evaluated 1–12 doses of SP for PMC in the second
year of life. The safety and impact of PMC programmes should be routinely monitored.
Delivery
The EPI platform remains important for delivering PMC, especially in the first year of life, and it may be possible to make use
of the EPI or other routine health visits, or establish new contacts to reach children over 1 year of age. Research on
98 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
alternative approaches for PMC delivery beyond the EPI schedules may be warranted. Consideration should be given to
contextual factors such as values and preferences of end-users, costs, coverage and sustainability of alternative delivery
platforms.
Drug resistance
The impact of drug resistance on the protection provided by PMC with SP is currently unclear. The duration of protection of
SP has been shown to be 42 days in settings without parasite resistance mutations. This was reduced to 21 days in a setting
where 89% of parasites carried the quintuple mutation [111]. In settings with a Pfdhps540 mutation frequency of up to 50%,
3–4 doses of PMC with SP reduced clinical malaria by 30% over the first year of life [111]. However, in the setting where the
Pfdhps540 mutation frequency was 89%, no overall protective effect of PMC was observed [111]. The efficacy of SP for
treatment is affected by the frequency of mutation-carrying parasites, but there is little evidence that the frequency of
molecular markers predicts the efficacy of PMC.
Contraindications
PMC is not recommended for individuals receiving other forms of malaria chemoprevention (e.g. SMC or MDA). Although
PMC and SMC could, in principle, be delivered to different age groups in the same geographical area, for example where
there is perennial malaria transmission with seasonal peaks, there is no operational experience of the co-delivery of these
strategies. There is currently no experience of co-administration of PMC with the RTS,S/AS01 malaria vaccine.
PMC is not recommended in children with severe acute illness or those who are unable to take oral medication, children who
during the last 30 days received a dose of any of the drugs being used for PMC, or those allergic to any of the drugs being
used for PMC. PMC with SP should not be given to individuals receiving a sulfa-based medication as treatment or
prophylaxis, including co-trimoxazole (trimethoprim–sulfamethoxazole).
Other considerations
Information about PMC should be fully accessible to caregivers and key stakeholders, such as government officials and
religious leaders. As with all health interventions, consent should be obtained from the caregiver on behalf of the child prior
to administration of PMC.
Evidence To Decision
99 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• Adverse events: In one study, the frequency of gastrointestinal symptoms was higher in children who received
PMC with SP compared to placebo (risk ratio: 2.25; 95% CI: 1.51–3.35) [107].
• Potential drug–vaccine interactions and blood transfusions were outcomes not covered by the systematic review.
However, a study done in a subset of children enrolled in five randomized controlled trials in Ghana, Kenya,
Mozambique and the United Republic of Tanzania found that PMC with SP did not affect the serological response
to EPI vaccines [110].
More information on the evidence can be found in the systematic review [106].
The overall certainty of the evidence for the outcomes of interest was considered moderate by the GDG. Although the
certainty of evidence, summarized under “Benefits and harms”, ranged from low to high, the priority outcomes of clinical
malaria and anaemia were assessed as moderate-certainty evidence, while severe malaria was considered low-certainty
evidence.
More information on the certainty of evidence assessments can be found in the ‘research evidence’ tab associated with
this recommendation online or in the annex of the pdf version.
• consultation with civil society, which indicated that chemoprevention to prevent malaria disease in children under 5
years was seen as a priority in endemic areas (CS4ME unpublished evidence);
• a synthesis of contextual factors from trials and pilots of PMC, predominantly in sub-Saharan Africa, which showed
that PMC is generally widely accepted by caregivers (Steinhardt unpublished evidence (a)).
The GDG determined that there was probably no important uncertainty or variability in how the outcomes of PMC are
valued across contexts.
More information can be found in the summary of contextual factors report (Steinhardt unpublished evidence (a)) and civil
society consultation report (CS4ME unpublished evidence).
Resources
PMC is generally considered cost-effective or highly cost-effective due to its use of the EPI delivery platform to deliver
the inexpensive drug SP. The cost per dose delivered in nearly all studies was less than $0.25 for PMC with SP, but more
expensive with alternative drugs. PMC becomes less cost-effective in settings with a lower malaria burden, as there is
less potential to avert disease, and with the use of more expensive medicines. The GDG considered the overall costs of
implementing PMC with SP in children to be moderate and judged that PMC is probably cost-effective compared to no
intervention.
More information on the evidence can be found in the evidence profile associated with this recommendation.
Equity
Little information on equity of PMC is available. One study found no association between wealth quintile and coverage
of PMC [112].
The GDG considered that PMC probably increases health equity when delivered using the EPI platform, since access to
100 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
More information on the evidence can be found in the summary of contextual factors report (Steinhardt unpublished
evidence (a)).
Acceptability
PMC has been widely accepted by caregivers, especially when delivered alongside vaccinations using the EPI platform.
EPI has also been generally well accepted and perceived as beneficial. Despite some health workers not liking the
process of administering PMC and some complaints that it increased workload, most had positive perceptions of PMC,
with some suggesting that it improved EPI attendance.
The GDG considered that PMC was probably acceptable to key stakeholders.
More information on the evidence can be found in the summary of contextual factors report (Steinhardt unpublished
evidence (a)).
Feasibility
Despite logistical challenges such as access to clean water, crushing the tablets, and occasional drug shortages, PMC
implementation appears feasible when it is delivered through the EPI platform. One time-and-motion study in the
United Republic of Tanzania found that the median time used for PMC implementation was 12.4 minutes (ranging from
1.6 minutes to 28.9 minutes) per nurse per vaccination session [113].
More information on the evidence can be found in the summary of contextual factors report (Steinhardt unpublished
evidence (a)).
Justification
This recommendation was developed using the Grading of Recommendations Assessment, Development and Evaluation
(GRADE) framework [103].
Sources of information
Recommendation development was informed by a systematic review [106], independently evaluated using the AMSTAR-2
Checklist [114] (Steinhardt et al unpublished evidence (b)), and a report summarizing evidence from published studies on
contextual factors related to PMC implementation (Steinhardt unpublished evidence (a)), including cost-effectiveness,
feasibility, equity, values and acceptability. These sources of information were supplemented by a cross-cutting review on
chemoprevention drug resistance (Plowe unpublished evidence), a civil society consultation report on chemoprevention
(CS4ME unpublished evidence) and contributions from the GDG membership, which included former and current national
malaria programme representatives.
The systematic review addressed the GDG’s PICO (population, intervention, comparison, outcome) question regarding
whether children living in settings with perennial malaria transmission should be given antimalarial medicines as
chemoprevention to reduce disease burden. The main outcomes of interest were the impact of PMC on confirmed clinical
malaria, severe malaria, and anaemia. Other outcomes of interest included: hospital admissions (all-cause and malaria-
specific); all-cause mortality; adverse events; drug–vaccine interactions; parasite prevalence; and blood transfusions. Twelve
trials were included in the review, three of which were cluster-randomized controlled trials. All the trials were conducted in
sub-Saharan Africa: Gabon, Ghana, Kenya, Mali, Mozambique, Uganda, and the United Republic of Tanzania. SP was
evaluated in 10 trials, amodiaquine in one trial, AS-AQ in one trial, DHAP in one trial, and SP+AS in one trial1. The
systematic review included trials that compared PMC with no intervention in young children (aged eight weeks to 24
months), with length of follow-up ranging from nine to 36 months of age, and most studies delivering 3–4 doses of
antimalarial. The AMSTAR-2 Checklist assessment concluded that the systematic review was well conducted and covered
most of the outcomes identified by the GDG in the PICO question (Steinhardt et al unpublished evidence (b)). Three outcomes
of interest to the GDG were not covered by the systematic review, namely malaria-specific hospital admissions, blood
transfusions, and potential drug–vaccine interactions.
101 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Summary of judgements
The Evidence-to-Decision table captures the evidence from the systematic review considered by the GDG. The GDG
determined that the balance between desirable and undesirable effects favoured PMC; moderate costs were associated with
PMC implementation delivered through EPI; PMC was considered probably cost-effective, but the use of alternative delivery
strategies to EPI may affect the cost-effectiveness of PMC, as might the use of more expensive antimalarials; and PMC was
probably acceptable to key stakeholders and feasible to implement. The GDG concluded that a conditional recommendation
should be made for PMC based on its moderate beneficial effect and moderate certainty of evidence.
Implementation
Please refer to the Intermittent preventive treatment for infants using sulfadoxine-pyrimethamine (IPTi-SP) for malaria control in
Africa: implementation field guide [112].
Evaluation
The effect of introducing PMC may be evaluated using routine hospital, clinic and/or community health worker data.
The WHO chemoprevention efficacy study (CPES) protocol should be used to monitor the efficacy of medicines when used
for chemoprevention. Although the potential effect of chemoprevention on the spread of drug resistance may be monitored
by the analysis of molecular markers associated with treatment outcomes, the correlation between molecular markers and
the efficacy of antimalarials for chemoprevention is unclear and results should be interpreted with caution.
Research Needs
Several evidence gaps were identified regarding PMC. None should prevent adoption and implementation of PMC.
Nevertheless, impact could potentially be enhanced by determining:
• the efficacy of PMC with SP, and alternative PMC regimens, within 28 days of administration;
• updated costs and cost-effectiveness of PMC delivered through the EPI, including in settings with low coverage of
routine childhood immunization;
• the effectiveness of different SP dosing schedules for PMC in children aged eight weeks up to 24 months;
• the effect of administering PMC to children >24 months old;
• the safety, efficacy and cost-effectiveness of alternative combination drugs for PMC (e.g. sulfadoxine-pyrimethamine
plus amodiaquine [SP+AQ]);
• the costs of and coverage achieved by alternative approaches to delivering PMC;
• the effectiveness of PMC in different antimalarial drug resistance contexts.
In areas of seasonal malaria transmission, children belonging to age groups at high risk of severe malaria should be given
antimalarial medicines during peak malaria transmission seasons to reduce disease burden.
• Eligibility for seasonal malaria chemoprevention (SMC) is defined by the seasonality of malaria transmission and age groups at
risk of severe malaria. Thresholds for assessing these criteria change over time and location. Malaria programmes should
assess the suitability of SMC based on the local malaria epidemiology and available funding (see “Practical info”). The added
value of a seasonally targeted intervention is likely to be greatest where transmission is intensely seasonal.
• Monthly cycles of sulfadoxine-pyrimethamine plus amodiaquine (SP+AQ) have been widely used for SMC in African children
under 5 years old and have been shown to be efficacious, safe, well tolerated, available and inexpensive (Thwing et al
unpublished evidence).
102 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Practical Info
Antimalarial medicine
WHO recommends a combination medicine for SMC that is different from that used for first-line malaria treatment. The
component medicines should have closely matched pharmacology, such that no component is present in the absence of
other components for more than a minimal amount of time in order to reduce the risk of new infections encountering only a
single drug. SP+AQ has been evaluated in 12 studies of SMC and has been widely used for SMC in Africa. SP+AQ has been
shown to be efficacious, safe, well tolerated, available and inexpensive (Thwing et al unpublished evidence). The prevalence of
molecular markers of resistance to SP+AQ was low in the general population before and two years after SMC
implementation in seven countries in west and central Africa (Bhattarai et al unpublished evidence). Safety and efficacy have
been evaluated for several other drug combinations, but the lack of widescale implementation means that fewer data are
available on the potential risks of cumulative toxicity and impact on drug resistance.
Age group
Most research studies have evaluated SMC in children aged 3–59 months. SMC given to children <5 years old reduced the
risk of clinical malaria by almost three quarters (risk ratio: 0.27; 95% CI: 0.25–0.29) during the transmission season (Thwing
et al unpublished evidence). SMC has also been shown to reduce the incidence of clinical malaria in children <10 years old.
Studies conducted in one country comparing the effect of SMC among children <5 years old with that in children 5–9 years
old found no difference in the effect size for malaria incidence or prevalence, severe malaria, or anaemia (Thwing et al
unpublished evidence). The age group targeted for SMC should be informed by the local age pattern of severe malaria
admissions. The cost-effectiveness of SMC will become less favourable as programmes expand to age groups at lower risk of
severe disease and areas of lower malaria transmission [116].
Dosage
Children in age groups at increased risk of severe disease should be given a complete course of antimalarials, at their
recommended treatment dose, as SMC. The drug dosage should be determined by the child’s weight wherever possible, with
dosing according to age only in situations where the child’s weight is unknown.
Frequency
The number of cycles should be informed by the duration of the high-transmission season, based on the local malaria
epidemiology, and the length of preventive efficacy of the selected drug combination. SMC should be used to protect
children during the entire high-transmission season. Current evidence supports monthly administration of SMC for 3–4
cycles in shorter transmission settings, and up to six cycles have been evaluated in settings with longer transmission seasons
(Thwing et al unpublished evidence).
Delivery
SMC can be provided through door-to-door or fixed-point delivery. A study in Mali found that door-to-door delivery
achieved significantly higher coverage than fixed-point delivery (76.1% versus 62.2%, p = 0.0028) [117]. Further studies in
Mali and Gambia have supported that door-to-door delivery can achieve high coverage [118][119]. Studies found similar
SMC coverage in children given directly observed treatment compared to non-directly observed treatment [117][118].
Drug resistance
While some prospective trials and ecological studies of SMC with SP+AQ in West Africa have reported increased prevalence
of the dhfr/dhps quadruple and quintuple mutants, other studies have found no evidence of selection. No evidence has been
reported of SMC being followed by increased prevalence of the higher level resistance mutations that most severely impair
curative SP efficacy, nor does SMC appear to select for parasites carrying mutations associated with diminished AQ
susceptibility (Plowe unpublished evidence).
Contraindications
SMC is not recommended for individuals receiving other forms of malaria chemoprevention (e.g. MDA or PMC). Although
PMC and SMC could, in principle, be delivered to different age groups in the same geographical area (e.g. where there is
perennial malaria transmission with seasonal peaks), there is no operational experience of the co-delivery of these
strategies.
SMC is not recommended for children with severe acute illness or those who are unable to take oral medication, children
who during the last 30 days received a dose of any of the drugs being used for SMC, or children with an allergy to any of the
drugs being used for SMC. Children should not be given SMC including SP if they are receiving a sulfa-based medication as
treatment or prophylaxis, including co-trimoxazole (trimethoprim–sulfamethoxazole).
Other considerations
Information about SMC should be fully accessible to caregivers and key stakeholders, such as government officials and
103 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
religious leaders. As with all health interventions, consent should be obtained from the caregiver on behalf of the child prior
to administration of SMC.
Evidence To Decision
More information can be found in the systematic review (Thwing et al unpublished evidence).
Notes
Results from non-randomized studies were consistent with those from randomized studies across all reported outcomes (incidence of confirmed
clinical malaria; prevalence of infection at end of transmission season; prevalence of moderate anaemia; incidence of severe malaria; hospitalization;
and all-cause mortality, all for children <5 years), except for prevalence of moderate anaemia, where no effect was observed. Adverse events were not
reported.
There was little evidence of an effect on all-cause mortality. It is plausible that a reduction in severe malaria could translate into an impact on mortality.
This was observed in one of the studies that was excluded from the systematic review as it did not use a controlled design [120]. However, the
evidence is hard to ascertain due to potential risk of bias from the study designs (trials with clinical malaria as the main outcome are likely to minimize
mortality) and systems for reporting deaths in the studies. Implementation of SMC was associated with reductions in malaria deaths in hospitals by
42.4% (95% CI: 5.9–40.9) in Burkina Faso and by 56.6% (95% CI: 28.9–73.5) in Gambia [120].
The overall certainty of the evidence for the outcomes of interest was considered to be moderate. The certainty of
evidence, as summarized under “Benefits and harms”, ranged from low to high. The priority outcome of confirmed
clinical malaria was assessed as moderate-certainty evidence.
More information on the certainty of evidence assessments can be found in the ‘research evidence’ tab associated with
this recommendation online or in the annex of the pdf version.
104 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• consultation with civil society, which indicated that chemoprevention to prevent malaria disease in children under 5
years was seen as a priority in endemic areas (CS4ME unpublished evidence);
• a synthesis of contextual factors from trials and pilots of SMC (Bhattarai et al unpublished evidence), but no research
was identified that described values and preferences related to SMC.
The GDG determined that there was probably no important uncertainty or variability in how the main outcomes of SMC
are valued.
More information can be found in the summary of contextual factors report (Bhattarai et al unpublished evidence) and the
civil society consultation report (CS4ME unpublished evidence).
Resources
The GDG considered the overall costs of implementing SMC to be moderate. Important cost drivers of SMC are the
drug used and the mode of delivery (e.g. door-to-door vs fixed-point). SMC is considered a cost-effective addition to
standard care, with the estimated average total economic cost per malaria case averted ranging from US$ 2.91 to US
$67.77, depending, in part, on the choice of drug (Bhattarai et al unpublished evidence). Expanding SMC to children in age
groups beyond those at highest risk of severe disease, areas of lower malaria transmission, and the use of more
expensive antimalarials will likely influence the cost-effectiveness of SMC.
More information can be found in the summary of contextual factors report for SMC (Bhattarai et al unpublished
evidence).
Equity
The GDG considered that SMC is likely to enhance equitable service delivery based on similar coverage of the
intervention across wealth quintiles in all countries where it is being implemented (Bhattarai et al unpublished evidence).
There was generally no significant difference in SMC coverage by age or gender.
More information can be found in the summary of contextual factors report for SMC (Bhattarai et al unpublished
evidence).
Acceptability
SMC acceptability was generally high, with overall refusal rates <1% in five countries (Bhattarai et al unpublished
evidence). Consequently, the GDG considered SMC to be acceptable to key stakeholders.
More information can be found in the summary of contextual factors report for SMC (Bhattarai et al unpublished
evidence).
Feasibility
SMC delivery approaches and coverage vary across countries. For example, in Mali, SMC coverage was significantly
higher in children who received SMC using door-to-door delivery compared to fixed-point delivery (76.1% versus 62.2%,
p = 0.0028), while in Gambia, SMC delivery through village health workers achieved a substantially higher coverage level
than delivery by reproductive and child health teams (74% versus 48%, a difference of 27%; 95% CI: 16%–38%) [117].
Overall, the GDG considered SMC implementation to be feasible.
More information can be found in the summary of contextual factors report for SMC (Bhattarai et al unpublished
evidence).
105 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Justification
This recommendation was developed using the Grading of Recommendations Assessment, Development and Evaluation
(GRADE) framework [103].
Sources of information
WHO commissioned a systematic review to inform this guidance on SMC (Thwing et al unpublished evidence), and a separate
report summarizing evidence from published studies on contextual factors related to SMC implementation (Bhattarai et al
unpublished evidence), including cost-effectiveness, feasibility, equity, values and acceptability. These sources of information
were supplemented by a cross-cutting review on chemoprevention drug resistance (Plowe unpublished evidence), a civil
society consultation report on chemoprevention (CS4ME unpublished evidence) and contributions from the GDG
membership, which included former and current national malaria programme representatives.
The objectives of the systematic review were to assess the effect of SMC with antimalarial drugs on malaria disease burden
among children living in places with seasonal malaria transmission, with a specific focus on the age of the children (3–59
months vs 60–120 months of age), the number of treatment cycles during a season (3–4 cycles vs 5–6 cycles), and the drug
regimen; and to summarize contextual information regarding acceptability, feasibility, equity, safety, drug resistance, cost and
cost-effectiveness. The primary outcome of interest was incidence of confirmed clinical malaria. Other outcomes included:
parasite prevalence; incidence of infection; anaemia prevalence; blood transfusions; hospital admissions; severe malaria; all-
cause mortality; adverse reactions; and school attendance. Seventeen studies met the criteria for inclusion (12 randomized
and five non-randomized studies) and were included in the review. All studies were conducted in sub-Saharan Africa,
including in Burkina Faso, Gambia, Ghana, Mali, Niger, Nigeria and Senegal. Twelve studies used SP+AQ, three studies used
AS-AQ, one study used SP+AS, and one study used AL. Trials administering three to four cycles were usually located in the
sites with shorter transmission seasons, whereas studies administering five to six cycles were in areas with longer
transmission seasons. None of the included studies reported incidence of infection or blood transfusions as outcome
measures. One study reported education outcomes but not school attendance.
Summary of judgements
Evidence from the systematic review (Thwing et al unpublished evidence) and supporting information (Bhattarai et al
unpublished evidence; CS4ME unpublished evidence; Plowe unpublished evidence) was appraised by the GDG in October 2021,
a summary of which is provided in the Evidence-to-Decision table. The GDG determined that SMC has a large beneficial
effect and that the balance of desirable and undesirable effects favours SMC; the costs of implementing SMC are moderate,
although the overall cost would be affected by the drug used and the mode of SMC delivery; SMC is cost-effective, but
expanding SMC to age groups beyond those at highest risk of severe disease or areas of lower malaria transmission, and the
use of more expensive antimalarials could reduce its cost-effectiveness; SMC is an acceptable intervention; SMC delivery
approaches and coverage varied across countries, but were judged to be feasible. In sum, the GDG judged the overall
certainty of the evidence as moderate and strongly recommended SMC for age groups at high risk of severe malaria living in
areas of seasonal malaria transmission to reduce disease burden.
Implementation
Please refer to the Seasonal malaria chemoprevention with sulfadoxine-pyrimethamine plus amodiaquine in children: A field
guide [117].
Evaluation
The effect of introducing SMC may be evaluated using routine hospital, clinic and/or community health worker data.
The WHO chemoprevention efficacy study (CPES) protocol should be used to monitor the efficacy of medicines when used
for chemoprevention. Although the potential effect of chemoprevention on the spread of drug resistance may be monitored
by the analysis of molecular markers associated with treatment outcomes, the correlation between molecular markers and
the efficacy of antimalarials for chemoprevention is unclear and results should be interpreted with caution.
Research Needs
The GDG highlighted the following evidence gaps requiring further research. These relate to:
106 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
School-aged children living in malaria-endemic settings with moderate to high perennial or seasonal transmission can be
given a full therapeutic course of antimalarial medicine at predetermined times as chemoprevention to reduce disease
burden.
• Intermittent preventive treatment in school-aged children (IPTsc) has been evaluated in children aged 5–15 years. The burden
of malaria and benefits of IPTsc may vary across this age range, but evidence is limited.
• National malaria programmes can consider IPTsc if resources allow for its introduction among school-aged children without
compromising chemoprevention interventions for those carrying the highest burden of severe disease, such as children < 5
years old.
• Schools may provide a low-cost means to deliver chemoprevention to school-aged children. However seasonal variation in
malaria transmission and the timing of school terms, as well as equity concerns, may mean alternative delivery channels are
needed to maximize impact.
• First- and second-line malaria treatments should not be used for IPTsc if safe and effective alternatives are available (see
“Practical info”).
• The dosing schedule for IPTsc should be informed by the local malaria epidemiology and timed to give protection during the
period of greatest malaria risk (see “Practical info”).
• Moderate to high malaria transmission settings are defined as areas with P. falciparum parasite prevalence greater than 10%
or an annual parasite incidence greater than 250 per 1000 [30]. These thresholds are indicative and should not be regarded as
absolutes for determining applicability of the IPTsc recommendation.
Practical Info
Antimalarial medicine
Drug regimens evaluated for IPTsc and found to be effective include SP combined with an aminoquinoline (either AQ or
piperaquine), SP+AS, and artemisinin-based combination therapy including an aminoquinoline (AS-AQ or DHAP)1. SP+AQ
has been widely used for chemoprevention in West Africa and has been shown to be efficacious, safe, well tolerated,
available and inexpensive. In order to reduce the risk of drug resistance to life-saving drugs, first- and second-line malaria
treatments should not be used for IPTsc if safe and effective alternatives are available.
The possibility of interactions with other drugs delivered as part of school health programmes should be considered.
Age group
The target age group should be identified using local data on the age distribution of malaria admissions and severe disease.
As young children (≤ 59 months) are the most vulnerable to severe malaria, chemoprevention interventions to protect this
age group should be prioritized over those for school-aged children. If resources allow for introduction of chemoprevention
for school-aged children without compromising chemoprevention in younger children, national malaria programmes can
consider IPTsc.
The majority of IPTsc studies have evaluated the intervention in children under 15 years old. There is some evidence of a
stronger effect on malaria-related anaemia in children younger than 10 years versus those who are 10–15 years. However,
the effect of IPTsc on P. falciparum infection was similar across these two age groups.
If older age groups are included in IPTsc, particular consideration should be given on how best to include girls with a history
of menarche. Certain antimalarials should not be given for chemoprevention without first confirming pregnancy status.
There is insufficient information on the safety, efficacy and pharmacokinetics of most antimalarial agents in pregnancy,
particularly during the first trimester. In IPTsc studies that have included girls with a history of menarche, pregnancy status
has been determined either through self-reporting or the use of pregnancy tests. Further research is needed on how best to
safely include girls of reproductive age in IPTsc.
Dosage
107 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
School-aged children should be given a complete course of antimalarials at their recommended treatment dose as IPTsc. The
drug dosage should be determined by the child’s weight wherever possible, with dosing according to age only in situations
where the child’s weight is unknown or cannot be determined.
Frequency
The IPTsc schedule should be informed by the local malaria epidemiology, particularly transmission intensity and seasonality,
the pharmacokinetics of the drug used, and the feasibility of delivering each additional IPTsc course. IPTsc should be timed
to give protection during the period of greatest malaria risk. Most trials provided IPTsc monthly or each term. In settings
where PMC is being provided, IPTsc may need to be given at regular intervals throughout the year. In perennial transmission
settings, the higher the transmission intensity, the greater the expected value of drugs with longer half-lives or more
frequent dosing, which will increase the proportion of time-at-risk protected by IPTsc. If IPTsc cannot be maintained
throughout the year in perennial transmission settings due to resource constraints, IPTsc may be timed to provide protection
during transmission peaks.
Delivery
IPTsc can be delivered either through schools or through community-based approaches. The method of delivery should
consider the local epidemiology of malaria and whether school-based delivery will offer protection during the period of
greatest malaria risk. All types of schools that cater to children aged up to 15 years in the target area should be included for
IPTsc delivery. National malaria programmes may be able to work with existing health programmes targeting school-aged
children to facilitate delivery of IPTsc. Children not attending school are likely to be at highest risk of malaria and, if school
attendance is not high, special efforts may be needed to target children not attending school. In seasonal transmission areas,
delivery in schools may not align with peak malaria transmission and thus it may be more appropriate to utilize existing
community-based approaches to reach school-aged children, such as those strategies used for SMC. Care is needed to
ensure adequate communication with communities, teachers, caregivers and children to maximize understanding and
acceptability in these key stakeholder groups. If older age groups are included in IPTsc administration, communication with
key stakeholders should pay attention to the inclusion of girls of reproductive age (see ‘Age group’ above).
Drug resistance
The impact of drug resistance on the protection provided by IPTsc is currently unclear. A re-analysis of data on resistance
markers following monthly IPTsc found no suggestion of an increased prevalence of any resistance markers following DHAP
administration2 (Plowe unpublished evidence).
A review of the relationship between the different chemoprevention strategies (IPTp, PMC, SMC, MDA, IPTsc) and drug
resistance concluded that malaria chemoprevention as used to date does not inevitably lead to an increase in resistance, and
even high rates of resistance may not necessarily impair chemoprevention efficacy (Plowe unpublished evidence). However,
expanded use of antimalarial medicines may increase resistance and eventually undermine efficacy. Using different drugs for
chemoprevention and treatment, and combining drugs with counteracting resistance mechanisms may help to preserve
efficacy (Plowe unpublished evidence).
Contraindications
IPTsc is not recommended for individuals receiving other forms of malaria chemoprevention (e.g. SMC or MDA). Children
with sickle cell disease should be included in IPTsc unless they already receive regular chemoprevention due to sickle cell
disease. Co-delivery of IPTsc alongside other school health programmes should consider drug manufacturers’ guidance
regarding whether IPTsc can be safely given with other medicines and whether there are any additional contraindications as
a result. Additionally, there is a need to consider how to include girls of reproductive age who should not be given certain
antimalarials for prophylaxis without first confirming that they are not pregnant (see "Age group' above for further
information).
IPTsc is not recommended in children with severe acute illness, those unable to take oral medication, children who during
the last 30 days received a dose of any drug being used for IPTsc, or those allergic to any of the drugs being used for IPTsc.
IPTsc-SP should not be given to individuals receiving a sulfa-based medication as treatment or prophylaxis, including co-
trimoxazole (trimethoprim–sulfamethoxazole) for HIV.
Other considerations
Information about IPTsc should be fully accessible to school-aged children, their caregivers and key stakeholders, such as
teachers. As with all health interventions, consent should be obtained from the caregiver on behalf of the child prior to
administration of IPTsc and, depending on age, from the child themselves.
108 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
1 Relative risk for P. falciparum infection: SP plus aminoquinoline (0.35; 95% CI: 0.25–0.44); SP+AS (0.04; 95% CI: 0.01–0.07); artemisinin derivative with
aminoquinoline (0.18; 95% CI: 0.11–0.24).
2 The original analysis limited resistance outcomes to the prevalence of pure mutant alleles for each locus of interest among all samples that were positive
for P. falciparum parasitaemia, irrespective of disease. The re-analysis was conducted to compare the proportion of infections containing any resistant
parasites, not just pure mutant alleles, based on the principle that any presence of resistance signals the risk of treatment failure.
Evidence To Decision
None of the studies that met the inclusion criteria for the review systematically collected data on school attendance,
severe malaria, hospital admissions (all-cause and malaria-specific), or mortality (all-cause and malaria-specific)2.
More information on the evidence can be found in the systematic review [125].
2 School achievement was not ranked by the GDG as a critical outcome and therefore was not considered. However, the systematic review found a
marginal effect of IPTsc on cognitive function in children 10–15 years (adjusted mean difference in standardized test scores: 0.36; 95% CI: 0.01–0.71;
p-value for interaction = 0.004), but no significant effect was identified when data were combined across all ages (adjusted relative risk*: 0.12; 95% CI:
-0.20–0.43; p = 0.4564).
The evidence for all the critical outcomes was of low certainty because of serious risk of bias and inconsistency between
the studies included in the review. Therefore, the GDG considered the overall certainty of the evidence for the
outcomes of interest to be low.
More information on the certainty of evidence assessments can be found in the ‘research evidence’ tab associated with
this recommendation online or in the annex of the pdf version.
• consultation with civil society, which indicated that chemoprevention to prevent malaria disease in children was
109 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
seen as important in endemic areas – although children under 5 years old were mentioned as the particular priority
(CS4ME unpublished evidence);
• a synthesis of contextual factors from trials and pilots of IPTsc in sub-Saharan Africa, which found very little data on
values and preferences (Gutman et al unpublished evidence (b)). In one study, parents considered chemoprevention
to be useful and recommended that chemoprevention be expanded to include older children and even adults [126].
The GDG determined that there was probably no important uncertainty or variability in how the outcomes of IPTsc are
valued across contexts.
More information can be found in the summary of contextual factors report (Gutman et al unpublished evidence (b)) and
the civil society consultation report (CS4ME unpublished evidence).
Resources
There are relatively few data on the cost of IPTsc. Key cost drivers were human resources (the provision of training to
teachers) and the drug used, with intervention costs varying substantially based on the selected regimen. In Mali, the
cost of delivering one course of SP+AS was US$ 2.72 per child, which decreased to US$ 1.00 per child for SP+AQ [127].
Modelling of IPTsc costs in Kenya estimated the intervention cost to be US$ 1.88 per child treated per year, with US$
0.25 per child in set-up costs and US$ 1.63 per child in recurrent costs.
The modelled cost-effectiveness of IPTsc in Kenya was US$ 5.36 per P. falciparum infection averted and US$ 29.84 per
case of anaemia averted [128]. The largest drivers of cost-effectiveness were the effectiveness of the intervention and
the prevalence of anaemia.
The GDG determined that the resources required to implement IPTsc varied, and the certainty of the evidence on the
resources required was low. The GDG concluded that IPTsc is probably a cost-effective intervention, and if existing
health interventions are being delivered through schools, integrating IPTsc could yield some cost savings. The overall
effectiveness of IPTsc is likely to be influenced by the local malaria epidemiology and age burden of disease: if school
children are at high risk during the school term, then the cost-effectiveness of IPTsc is likely to increase.
More information on the evidence can be found in the summary of contextual factors report (Gutman et al unpublished
evidence (b)).
Equity
There is very limited data on how a school-based platform for delivery of malaria chemoprevention to children would
affect equity and health equality.
The GDG considered the equity of IPTsc to vary, depending on the proportion of children attending school. As those
absent from school are more likely to be from lower socioeconomic groups and female, delivering IPTsc solely through
schools may affect the equity of the strategy. There is some evidence that the effect of IPTsc on school performance
may differ between girls and boys [129].
More information on the evidence can be found in the summary of contextual factors report (Gutman et al unpublished
evidence (b)).
Acceptability
Few studies directly assessed the acceptability of IPTsc. Community sensitization was identified as important for
improving the acceptability of IPTsc. In one study, 93% of children reported that they would be willing or very willing to
take the tablets for IPTsc each school term [123]. Another study, which evaluated IPTsc among other interventions (iron
fortification and anthelmintics), delivered two rounds of IPTsc-SP three months apart. Only one person (0.15%)
approached for enrolment refused to participate, and there was high compliance (93.7%) among those who participated,
suggesting that treatment was acceptable [130]. In a study that added malaria treatment to an existing school-based
110 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
MDA programme, 87% of children received IPTsc, suggesting that it might be acceptable to combine the intervention
with ongoing health programmes [131]. In another study, staff noted issues with acceptance from parents, particularly
when there were side effects from the drugs. Consequently, parents would refuse the second and third days of
treatment, and acceptance was lower with subsequent rounds [126].
More information on the evidence can be found in the summary of contextual factors report (Gutman et al unpublished
evidence (b)).
Feasibility
The feasibility of IPTsc is influenced by the choice of drug regimen. One study suggested that using a simpler
antimalarial regimen would enhance compliance, as single-dose regimens could be administered as DOT. Additionally,
feasibility may be adversely impacted in girls of reproductive age, given the need to confirm that they are not pregnant
before giving certain antimalarials as IPTsc [132]. Poor uptake of IPTsc in one study was attributed to poor community
perceptions about IPTsc and the requirement for parental informed consent [132]. School-based delivery is likely to be
more feasible than community-based delivery of IPTsc, but enrolment rates and absenteeism could pose barriers to
reaching children through schools [132]. In some countries, schools already provide nutrition services and are sites of
targeted insecticide-treated net (ITN) distribution and deworming programmes (Gutman et al unpublished evidence (b)).
More information on the evidence can be found in the summary of contextual factors report (Gutman et al unpublished
evidence (b)).
Justification
This recommendation was developed using the Grading of Recommendations Assessment, Development and Evaluation
(GRADE) framework [103].
Sources of information
Recommendation development was informed by a systematic review [125], independently evaluated using the AMSTAR-2
Checklist (Gutman et al unpublished evidence (c))[114], and a report summarizing evidence from published studies on
contextual factors related to IPTsc implementation (Gutman et al unpublished evidence (b)), including cost-effectiveness,
feasibility, equity, values and acceptability. These sources of information were supplemented by a cross-cutting review on
chemoprevention and drug resistance (Plowe unpublished evidence), a civil society consultation report on chemoprevention
(CS4ME unpublished evidence) and contributions from the GDG membership, which included former and current national
malaria programme representatives.
The systematic review addressed the GDG’s PICO (population, intervention, comparison, outcome) question regarding
whether school-aged children living in settings with malaria transmission should be given antimalarial medicines as
chemoprevention to reduce disease burden. The main outcome of interest was the impact of IPTsc on confirmed clinical
malaria. Other outcomes of interest included anaemia, school attendance, parasite prevalence, severe malaria, hospital
admissions (all-cause and malaria-specific), adverse events, and mortality (all-cause and malaria-specific). Thirteen
randomized trials were included in the review, 11 of which contributed data to an individual participant data meta-analysis.
All the trials were conducted in sub-Saharan Africa: Côte d'Ivoire, Democratic Republic of the Congo, Ghana, Kenya, Mali,
Senegal and Uganda. Drug regimens evaluated in the individual studies included DHAP in three trials, SP in three trials,
SP+AQ in three trials, SP+AS in two trials, SP with piperaquine in one trial, AL in two trials, AS+AQ in two trials, doxycycline
in one trial, primaquine in one trial, mefloquine plus multivitamin in one trial, and proguanil plus chloroquine in one trial. The
systematic review grouped the treatment regimens by drug class and pharmacokinetic features: SP alone, SP combined with
an aminoquinoline (either AQ or piperaquine), SP+AS, artemisinin-based combination therapy including an aminoquinoline
(AS+AQ or DHAP), and AL. Treatment intervals ranged from daily (with subtherapeutic doses of primaquine and doxycycline)
to every four months, with the majority of studies providing IPTsc monthly or each term (i.e. 3-4 month intervals). The
systematic review included trials that studied IPTsc in children aged 5 to 15 years old, with the follow-up period ranging
from six to 103 weeks and most studies delivering 1–12 courses of antimalarial treatment. The authors of the review
estimated the proportion of the follow-up period protected by treatment for each of the individual studies, and this ranged
from 2% to 100%. The AMSTAR-2 Checklist assessment concluded that the systematic review was of sufficient quality, and
111 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
the inclusion of one new study identified since the systematic review was published did not substantially change the
conclusions (Gutman et al unpublished evidence (c)). Four outcomes of interest to the GDG were not covered by the
systematic review, namely school attendance, severe malaria, hospital admissions (all-cause and malaria-specific) and
mortality (all-cause and malaria-specific).
Summary of judgements
The Evidence-to-Decision framework captures the evidence from the systematic review considered by the GDG. The GDG
considered the balance between desirable and undesirable effects to probably favour IPTsc; costs associated with IPTsc
implementation to vary; and the certainty of the evidence on resources required to be low. In addition, IPTsc was considered
probably cost-effective; the equity of IPTsc was judged to vary, depending on the proportion of children attending school;
and IPTsc was judged as probably acceptable to key stakeholders and probably feasible to implement. The GDG concluded
that a conditional recommendation should be made for IPTsc for school-aged children in moderate to high burden malaria
transmission settings given IPTsc’s moderate beneficial effects and small undesirable effects.
Implementation
A guide to support implementation of IPTsc will be developed in due course, and a manual for subnational tailoring of
malaria interventions is under development and expected for publication in 2022.
Evaluation
The safety and impact of IPTsc programmes should be routinely monitored. The effect of introducing IPTsc may be evaluated
using routine hospital, clinic and/or community health worker data. School surveys provide an opportunity to evaluate
outcomes related to school attendance and achievement.
The WHO chemoprevention efficacy study (CPES) protocol should be used to monitor the efficacy of medicines used for
chemoprevention. Although the potential effect of chemoprevention on the spread of drug resistance may be monitored by
the analysis of molecular markers associated with treatment outcomes, the correlation between molecular markers and the
efficacy of antimalarials for chemoprevention is unclear and results should be interpreted with caution.
Research Needs
The GDG highlighted the following evidence gaps requiring further research. These relate to:
• the efficacy of alternative (e.g. monthly versus each term) IPTsc drug regimens at different transmission intensities;
• the value of IPTsc in children 10 years and under compared to the value in children over 10 years old;
• the full economic and financial costs (including the cost of engaging communities, parents, school teachers, etc.) of
introduction and deployment of IPTsc;
• the cost-effectiveness of combining IPTsc with other school health programmes;
• the costs and feasibility of alternative strategies to deliver malaria chemoprevention to school-aged children;
• the development of drugs suitable for use as chemoprevention in school-aged children;
• the effect of IPTsc on community-level transmission;
• the impact of IPTsc on cognition and school performance;
• the development of drugs for malaria chemoprevention that can be administered as a single dose;
• evaluating approaches to safely include girls of reproductive age in IPTsc, including exploring alternative regimens that
are safe through pregnancy.
112 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Children admitted to hospital with severe anaemia living in settings with moderate to high malaria transmission can be given
a full therapeutic course of an antimalarial medicine at predetermined times following discharge from hospital to reduce re-
admission and death.
• Post-discharge malaria chemoprevention (PDMC) should be given to children following admission with severe
anaemia [133] that is not due to blood loss following trauma, surgery, malignancy or a bleeding disorder.
• PDMC implementation should be tailored to admissions of children with severe anaemia and consider the duration of
protection of the selected antimalarial, and the feasibility and affordability of delivering each additional PDMC course (see
“Practical info”).
• Moderate to high perennial malaria transmission settings are defined as areas with a P. falciparum parasite prevalence greater
than 10% or an annual parasite incidence greater than 250 per 1000 [30]. These thresholds are indicative and should not be
regarded as absolute for determining applicability of the PDMC recommendation.
Practical Info
Antimalarial medicine
Medicines used for PDMC can be the same as the first-line malaria treatment, but an alternative medicine is preferred. SP,
AL and DHAP were used in three trials and all regimens were found to be effective for PDMC (Phiri et al unpublished
evidence).
Age group
Local data on the age distribution of severe anaemia should be referenced when determining the target age group for
PDMC. Two studies evaluated PDMC doses in children under 59 months [134][135], and one study evaluated doses in
children aged 3 months to 9 years [136].
Dosage
Children on PDMC should receive a complete course of antimalarials at the recommended treatment dose. The drug dosage
should be determined by the child’s weight wherever possible, with dosing according to age only in situations where the
child’s weight is unknown or cannot be determined.
Frequency
The frequency of PDMC administration should be informed by the length of protective efficacy of the selected drug, the
duration of the transmission season, and the feasibility of delivering each additional PDMC treatment. Two of the three trials
providing evidence for this recommendation provided three PDMC treatments. One trial administered SP monthly starting
seven days post-discharge until the end of the transmission season [136]; another trial administered AL at discharge then
twice at four and eight weeks post-discharge [134]; and the third trial administered AL at discharge and then DHAP three
times starting 14 days post-discharge and then monthly [135].
Delivery
Two delivery approaches for PDMC were evaluated in one effectiveness study: community-based and facility-based delivery
strategies. For community-based delivery, caregivers received all courses of PDMC on discharge, whereas for facility-based
delivery, the caregiver had to collect the PDMC drugs from a health facility each month. Community-based delivery was
preferred by caregivers and associated with increased adherence compared to facility-based strategies (community:70.6%
vs. facility: 52.0%, p = 0.006) [137]. Caregivers felt that the instructions on PDMC administration written on the child’s
health card were sufficient without reminders via text message or from community health workers (CHWs). There was no
statistical evidence that SMS reminders resulted in greater adherence (incidence rate ratio: 1.03; 95% CI: 0.88–1.21; p =
0.68) [138].
Drug resistance
The impact of drug resistance on the protection provided by PDMC is currently unclear. A relatively small proportion of the
population is eligible for PDMC compared to other malaria chemoprevention interventions such as SMC, PMC or IPTp.
Hence, the selective pressure exerted by PDMC on the parasite population, and consequent risk of PDMC increasing
resistance to antimalarials across the population, is likely to be small.
Contraindications
113 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Individuals should not receive both PDMC and other forms of malaria chemoprevention (e.g. SMC, PMC or MDA). If other
malaria chemoprevention programmes are unable to effectively screen and exclude individuals receiving PDMC, then PDMC
should not be administered during periods when SMC, PMC or MDA are being provided. Children with sickle cell disease
should be included in PDMC, unless they are already receiving regular chemoprevention due to sickle cell disease.
PDMC is not recommended in children who develop severe acute illness following discharge, those who are unable to take
oral medication, children who during the last 30 days received a dose of any of the drugs being used for PDMC, or those
allergic to any of the drugs being used for PDMC. PDMC-SP should not be given to individuals receiving a sulfa-based
medication as treatment or prophylaxis, including co-trimoxazole (trimethoprim–sulfamethoxazole) for HIV.
Other considerations
Information about PDMC should be fully accessible to caregivers. As with all health interventions, consent should be
obtained from the caregiver on behalf of the child prior to administration of PDMC.
Evidence To Decision
• Re-admission (all-cause and severe anaemia): PDMC probably reduces all-cause re-admission during the
intervention period (risk ratio: 0.42; 95% CI: 0.34–0.52; moderate-certainty evidence). In the post-intervention
period, the effect of PDMC varies and may result in little to no difference in all-cause re-admission (hazard ratio:
1.04; 95% CI: 0.83–1.30; moderate-certainty evidence). PDMC probably reduces re-admission for severe anaemia
during the intervention period (hazard ratio: 0.38; 95% CI: 0.26–0.56; moderate-certainty evidence) and during the
post-intervention period (hazard ratio: 0.74; 95% CI: 0.52–1.05; moderate-certainty evidence). PDMC probably
reduces re-admission for severe malaria during the intervention period (hazard ratio: 0.32; 95% CI: 0.22–0.48;
moderate-certainty evidence), but may have little effect during the post-intervention period (hazard ratio: 1.06;
95% CI: 0.81–1.39; moderate-certainty evidence).
• Death (all-cause): PDMC reduces all-cause mortality during the intervention period (risk ratio: 0.23; 95% CI:
0.08–0.70; high-certainty evidence). The effect in the post-intervention period varies and may result in little or no
difference in all-cause mortality (risk ratio: 1.61; 95% CI: 0.81–3.19; moderate-certainty evidence). Overall, PDMC
probably reduces all-cause mortality (risk ratio: 0.77; 95% CI: 0.47–1.28; moderate-certainty evidence).
• Clinical malaria: PDMC probably reduces clinical malaria (hazard ratio: 0.64; 95% CI: 0.58–0.72; moderate-certainty
evidence), with most of the benefit accruing during the intervention period (hazard ratio: 0.43; 95% CI: 0.36–0.50;
versus 0.96; 95% CI: 0.83–1.11 during the post-intervention period; both moderate-certainty evidence).
• Adverse events: The three randomized controlled studies provided moderate-certainty evidence on adverse events
associated with using different antimalarials: SP, AL, and DHAP. Minor symptoms recorded for those in the SP arm
30 days after the administration of each treatment were similar to those seen in the placebo arm [136]. DHAP
administration was associated with vomiting within 60 minutes after drug intake (12.4%, compared to placebo
3.8%) [135]. No drug-related serious adverse events were reported in the study arm receiving monthly AL [134].
DHAP was associated with an 18.6 ms (95% CI: 15.6–21.8; moderate-certainty evidence) increase of the QTc
interval (Fridericia correction) after the third dose of each course. All events of QTc interval prolongation were
asymptomatic and none of the children in the DHAP group had QTc interval values of more than 500 ms (Fridericia-
corrected).
No information was provided in the systematic review on severe malaria, anaemia or severe anaemia not associated
with re-admission, blood transfusion or parasite prevalence outcomes.
More information on the evidence can be found in the systematic review (Phiri et al unpublished evidence).
The certainty of the evidence across all critical outcomes ranged from moderate to high. Only the evidence on the effect
114 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
of PDMC on all-cause mortality during the 2–14-week intervention period was of high certainty. The GDG
consequently considered the certainty of the evidence overall to be moderate.
More information on the certainty of evidence assessments can be found in the ‘research evidence’ tab associated with
this recommendation online or in the annex of the pdf version.
• consultation with civil society, which indicated that chemoprevention to prevent malaria disease in children under 5
years was seen as a priority in endemic areas, although there was no specific mention of the need during the post-
discharge period (CS4ME unpublished evidence);
• a synthesis of contextual factors from trials of PDMC in sub-Saharan Africa (Lange et al unpublished evidence). The
report showed that caregivers had generally positive views of PDMC. Caregivers understood the value of giving
preventive malaria medicines during the post-discharge period, given that their children had recently been in
hospital [139]. CHWs also viewed PDMC as an important and beneficial intervention [137].
The GDG determined that there was probably no important uncertainty or variability in how the outcomes of PDMC are
valued across contexts.
More information can be found in the summary of contextual factors report (Lange et al unpublished evidence) and the
civil society consultation report (CS4ME unpublished evidence).
Resources
The mean estimated cost of implementing community-based PDMC was between US$ 22.91 and US$ 28.33 per child
treated in the three countries where the studies were conducted. Implementation costs for community-based PDMC
were outweighed by cost savings for re-admission compared to standard care, with a mean expected saving per child
between US$ 22.08 and US$ 45.24. Health care providers’ net cost saving per child receiving PDMC, including health
care (especially blood transfusion) and societal costs, was between US$ 19.12 and US$ 25.71. Two approaches for
delivering PDMC were evaluated: (i) facility-based, in which children had to be brought to a health facility to receive
subsequent doses of PDMC, and (ii) community-based, in which the caregiver received all doses for PDMC on discharge
with instructions and dates for administration written on the child’s health card, and with CHWs reminding caregivers
when to administer doses, SMS reminders, or no reminders. Community-delivered PDMC was found to be more cost-
saving compared to health facility-based delivery due to costs from repeated travel for drug collection, which also posed
a disincentive to adherence.
The GDG judged that PDMC probably results in moderate savings and is therefore probably cost-effective, but the
certainty of the evidence regarding the resources required was low.
More information on the evidence can be found in the report on PDMC cost-effectiveness (Kϋhl et al unpublished
evidence).
Equity
None of the studies included in the PDMC contextual factors report were designed to capture issues related to equity.
However, caregivers whose children received PDMC from the health facility reported that repeated travel to the
hospital to collect medicines was costly and time-consuming. Caregiver literacy was identified as a potential challenge
for equitable PDMC delivery among participants who received all medicines when their child was discharged
(community-based delivery), as some caregivers may not be able to read the PDMC administration dates recorded on
their child’s health card. SMS reminders (see “Feasibility” below) may also raise concerns over equity.
The GDG considered that PDMC has a variable effect on health equity and noted that PDMC likely reinforces existing
115 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
health inequities, given that it is administered to children who have already accessed a hospital. Nevertheless, among
those who have already accessed a hospital, the intervention is likely to be equitable; however, this may be dependent
on how PDMC is administered, with community-based delivery being potentially more equitable than facility-based
delivery.
More information on the evidence can be found in the summary of contextual factors report (Lange et al unpublished
evidence).
Acceptability
One study showed that community-based PDMC resulted in higher self-reported adherence than facility-based PDMC
(71% vs 52% adherence to the full three courses). Community-based adherence may have been influenced by the
anticipation of study staff visits for pill counts after each treatment course. Potential stigma from repeated CHW visits
may be a potential issue for community-based adherence.
More information on the evidence can be found in the summary of contextual factors report (Lange et al unpublished
evidence).
Feasibility
For community-based delivery of PDMC, CHWs reported a high level of intrinsic motivation to conduct home visits to
remind caregivers to administer PDMC doses. Nevertheless, adherence to the required number of home visits was poor,
with less than half of the CHWs conducting the required home visit reminders. Positive factors that encouraged CHWs
to conduct home visits were the knowledge and perception of PDMC effectiveness, and recognition from the
community and the health system. Reported barriers to CHWs conducting home visits included poor training, lack of
supervision, and high workload.
Written reminders of PDMC treatment dates on children’s health cards were positively viewed by participants. Most
caregivers preferred SMS reminders over CHW visits, but those who didn’t own a phone had to receive reminders
through neighbours and/or family members, which caused delays. Although PDMC adherence was higher among SMS
recipients (66.2%) compared to non-SMS participants (56.9%), there was no statistical evidence that SMS reminders
resulted in greater adherence (incidence rate ratio: 1.03; 95% CI: 0.88–1.21; p = 0.68).
The GDG concluded that it was unclear whether PDMC implementation was broadly feasible, given that there is
currently only evidence from three trials, including one implementation study. The optimal approach to PDMC
implementation may vary in different places and, where CHWs are involved, may benefit from a direct link between
health facilities and community-based care.
More information on the evidence can be found in the summary of contextual factors report (Lange et al unpublished
evidence).
Justification
This recommendation was developed using the Grading of Recommendations Assessment, Development and Evaluation
(GRADE) framework [103].
Sources of information
Recommendation development was informed by a systematic review (Phiri et al unpublished evidence), independently
evaluated using the AMSTAR-2 Checklist [114](Gutman et al unpublished evidence (d)), and a report summarizing evidence
from published studies on contextual factors related to PDMC implementation (Lange et al unpublished evidence), including
feasibility, equity, values and acceptability, as well as a cost-effectiveness analysis (Kϋhl et al unpublished evidence). These
sources of information were supplemented by a cross-cutting review on chemoprevention and drug resistance (Plowe
unpublished evidence), a civil society consultation report on chemoprevention (CS4ME unpublished evidence) and
contributions from the GDG membership, which included former and current national malaria programme representatives.
The systematic review addressed the GDG’s PICO (population, intervention, comparison, outcome) question regarding
116 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
whether children hospitalized with severe anaemia in malaria-endemic settings should be given antimalarial medicines as
chemoprevention post-discharge. The main outcomes of interest were the impact of PDMC on re-admission (all-cause and
severe anaemia), mortality (all-cause), severe anaemia, and blood transfusion. Other outcomes of interest included confirmed
clinical malaria, severe malaria, anaemia, adverse events, and parasite prevalence. Three randomized double-blind placebo-
controlled trials were included in the review. All the trials were conducted in sub-Saharan Africa: Gambia, Kenya, Malawi and
Uganda. One trial evaluated monthly SP until the end of the malaria transmission season; another trial evaluated monthly AL
at four and eight weeks post-discharge; and the third trial evaluated monthly DHAP at 14, 42 and 70 days post-discharge.
The systematic review included trials that compared PDMC with no intervention in children aged < 9 years with anaemia,
defined as haemoglobin < 7 g/dL (one trial), or severe anaemia, defined as haemoglobin < 5 g/dL. The intervention period
started from the first dose of the first course of PDMC and continued until four weeks after the first dose of the last course
of PDMC, a follow-up period of 2–14 weeks. The post-intervention period started the day after the completion of the
intervention period and continued up to 26 weeks. The AMSTAR-2 Checklist assessment concluded that the systematic
review was good quality overall (Gutman et al unpublished evidence (d)). Five outcomes of interest were not covered by the
systematic review, namely severe malaria, anaemia, severe anaemia, blood transfusion and parasite prevalence.
Summary of judgements
The Evidence-to-Decision framework captures the evidence from the systematic review considered by the GDG. The GDG
determined that the balance between desirable and undesirable effects favoured PDMC; moderate cost savings were
probably associated with PDMC implementation; PDMC is therefore probably cost-effective, although the certainty of
evidence regarding required resources was low; and PDMC is probably acceptable to key stakeholders, but the feasibility of
implementing PDMC at scale is not known. The GDG concluded that a conditional recommendation should be made for
PDMC based on the moderate- to high-certainty evidence of large beneficial effects and likely low costs.
Implementation
A guide to support implementation of PDMC will be developed in due course, and a manual for subnational tailoring of
malaria interventions is under development and expected for publication in 2022.
Evaluation
PDMC programmes should be routinely monitored for safety, efficacy, drug resistance and effectiveness. The impact of
introducing PDMC may be evaluated using routine hospital, clinic and/or CHW data.
The potential effect of PDMC on the spread of drug resistance is likely to be modest, given the small proportion of the
population receiving the intervention. Resistance may be monitored by the analysis of molecular markers associated with
treatment outcomes, although the correlation between molecular markers and the efficacy of antimalarials for
chemoprevention is unclear and should be interpreted with caution.
Further guidance will be made available in the PDMC implementation guide, which will be developed in due course.
Research Needs
The GDG identified the following evidence gaps as requiring further research. These relate to:
• the optimal duration for PDMC in different geographical and transmission settings, and understanding of the short-,
medium- and long-term benefits of PDMC of different durations; these evaluations should recognize the underlying
pattern of post-discharge death and/or re-admission, and the higher risk of some groups dying soon after discharge; to
minimize bias, the overall impact during the whole intervention and follow-up period should be considered;
• a better understanding of risk factors (including age) for adverse outcomes following discharge with severe anaemia,
and potential differential effects of PDMC in different risk groups;
• patient adherence to PDMC when deployed at scale;
• costs of and coverage achieved by alternative approaches to delivering PDMC;
• feasibility of different coordination mechanisms between hospital and outpatient/community settings for PDMC;
• feasibility of implementing PDMC in parallel with other malaria chemoprevention interventions (e.g. SMC and PMC);
• the long-term (e.g. 12 months and longer) impact of PDMC on child survival;
• the effectiveness of PDMC on severe anaemia of different etiologies;
• the effectiveness of PDMC for children diagnosed with severe anaemia and malaria in low transmission settings;
• the feasibility, costs and effects of combining PDMC with additional interventions (e.g. ITNs) to reduce the household’s
risk of further infection and adverse health outcomes.
117 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
medicine at approximately the same time, and often at with an antimalarial medicine that includes an
repeated intervals, to all age groups of a population in a 8-aminoquinoline to clear hypnozoites. An 8-aminoquinoline
defined geographical area. Antimalarial medicines are medicine has the potential to cause severe haemolysis in
administered without prior malaria testing and therefore persons deficient for the glucose-6-phosphate dehydrogenase
regardless of the malaria infection status of individuals. (G6PD) enzyme. Safe administration of an 8-aminoquinoline
Consequently, any existing infections are treated and new requires G6PD testing, an effective pharmacovigilance system
infections are prevented for the duration of the drug’s and emergency access to blood transfusion services. The two
prophylactic period. MDA has been an important component GDGs that reviewed evidence for the impact of MDA on P.
of malaria control and elimination programmes for vivax prioritized different outcome measures and arrived at
decades [140]. Some earlier WHO documents referred to “age- different recommendations. Whereas the evidence was
targeted MDA”: however, such use cases are no longer considered insufficient to recommend MDA for the reduction
considered MDA and recommendations for such targeted use of P. vivax disease, it was recognized that, in some situations,
are presented separately – see recommendations for perennial MDA may usefully contribute to the reduction of P. vivax
malaria chemoprevention (PMC) (section 4.2.2) and seasonal transmission. Malaria programmes should, therefore, review
malaria chemoprevention (SMC) (section 4.2.3). The use of the MDA recommendations for P. vivax and decide whether or
chemoprevention in occupationally vulnerable groups, such as not an MDA intervention is likely to lead to a successful
forest workers, is considered targeted drug administration outcome in their setting.
(TDA) and not MDA. Similarly, use of chemoprevention around
a confirmed case in areas approaching elimination or post- A chemoprevention strategy related to MDA that is intended
elimination preventing re-establishment is known as reactive to reduce transmission of P. vivax is mass relapse prevention
drug administration (RDA). Although not called MDA, all of (MRP). MRP is similar to MDA in that the entire population of
these strategies share a common underlying principle – that a delimited geographical area is provided with an antimalarial
the provision of a treatment dose of antimalarial medicine will medicine at approximately the same time. In the case of MRP,
cure existing infections and prevent new ones. however, only an 8-aminoquinoline drug is provided. In the
past, the strategy used primaquine and was referred to as
Historically, MDA has been given either to reduce malaria “mass primaquine prophylactic treatment”. However, the name
disease burden or to reduce malaria transmission. The of this strategy has since been expanded to include the
distinction between the two MDA use cases for P. falciparum is potential for new drugs with similar anti-relapse properties.
to some extent artificial, as any intervention that reduces Generally deployed in areas with cold winters and highly
transmission will also reduce disease burden, and burden- seasonal transmission of P. vivax, the medicine is provided to
reducing interventions that reach a sufficient proportion of the the population in early spring, when there is no or very low
population will also reduce transmission. Nevertheless, the transmission of the parasite, to treat hypnozoites and prevent
evidence on the use of MDA for disease burden and relapses that could infect a new population of mosquitoes in
transmission reduction was considered separately by two the summer months.
Guideline Development Groups (GDGs). The two GDGs
broadly recommended that programmes may consider MDA to WHO recommends that malaria programmes tailor
reduce P. falciparum transmission in very low to low intervention packages to their local context. The MDA
transmission settings, and to reduce disease burden in recommendations are subject to considerations, identified by
moderate to high transmission settings. A P. falciparum the GDGs, which will influence the likelihood of successful
prevalence (PfPR2-10) of around 10% (or incidence of infection outcomes. These contextual considerations are outlined in
remarks under the recommendations and in the “Practical info”
around 250 per 1000 population per year) may be used to
sections.
differentiate areas of low to very low transmission from areas
of moderate to high transmission. These thresholds should not • Recommendations regarding the use of MDA for burden
be considered absolute cut-offs and it is biologically plausible reduction are presented in section 4.2.6.1 MDA for burden
that MDA in settings near the 10% threshold may reduce both reduction; and recommendations for burden reduction in
disease burden and transmission intensity. However, the emergency settings are presented in section 4.2.6.2 MDA
relative effects of burden reduction versus transmission for burden reduction in emergency settings;
reduction differ along the transmission spectrum. Malaria • Recommendations for transmission reduction are found in
programmes should therefore review the MDA section 4.2.6.3 MDA to reduce transmission of P.
recommendations and practical information for both burden falciparum in very low to low transmission settings; section
and transmission reduction and decide whether or not an 4.2.6.4 MDA to reduce transmission of P. falciparum in
MDA intervention is likely to lead to a successful outcome in moderate to high transmission settings; and section 4.2.6.5
their setting. MDA to reduce transmission of P. vivax.
• The recommendation for MRP is found in section 4.2.6.6
The use of MDA for P. vivax is more complicated, as P. vivax
MRP to reduce transmission of P. vivax.
infections may relapse within a few months unless treated
118 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Antimalarial medicine can be given as chemoprevention through mass drug administration (MDA) in areas of moderate
to high transmission of P. falciparum to provide short-term reductions in disease burden.
• MDA may quickly reduce clinical malaria incidence in settings with moderate to high P. falciparum transmission, but the
effect wanes within 1–3 months. Therefore, if MDA is implemented, it should be one of several components of a robust
malaria control programme (including good coverage of effective case management and appropriate prevention tools and
strategies).
• Malaria programmes should judge the suitability of using MDA in their context based on the desired impact, level of
endemicity, and resources required. MDA for burden reduction should be targeted at moderate to high transmission
settings, regardless of seasonality (see “Practical info”).
• Moderate to high malaria transmission settings are defined as areas with P. falciparum parasite prevalence greater than
10%, or incidence greater than 250 P. falciparum cases per 1000 population per year [30]. These thresholds should not be
regarded as absolutes for determining applicability of MDA implementation. It is biologically plausible that MDA in
intermediate transmission settings may reduce both disease burden and transmission intensity.
Practical Info
Transmission setting
The impact of MDA on disease burden varies between high and low malaria transmission settings. In high transmission
settings, the impact of MDA on disease is likely to be large and may be cost-effective due to the high background
disease burden. However, as transmission intensity and the corresponding disease burden decrease, the impact of MDA
also decreases and MDA becomes less cost-effective for disease burden reduction. The effect on other outcomes,
parasite incidence and prevalence, and incidence of severe disease also appears to vary by transmission intensity. There
are no studies directly comparing the impact of MDA for burden reduction with the impact of more targeted approaches
to chemoprevention (e.g. SMC) (Schneider et al unpublished evidence (a)). MDA for burden reduction should be targeted
at moderate to high transmission settings, regardless of seasonality.
Antimalarial medicine
WHO recommends the use of a combination medicine for MDA that is different from that used as first-line malaria
treatment. The component medicines should have closely matched pharmacology, such that no component is present in
the absence of other components for more than a minimal amount of time in order to reduce the risk of new infections
encountering only a single drug. A drug regimen that can be administered as a directly observed single dose is preferable
to a multi-day regimen. Data were insufficient to discern a specific effect of single-dose primaquine. Available evidence
suggests that maximum benefits are seen within 1–3 months after the last round of the intervention (Schneider et al
unpublished evidence (a)).
Dosage
A complete therapeutic course of antimalarials, at doses recommended by the manufacturer, should be given to all
eligible adults and children within a defined geographical area. Drug dosage should be determined by weight wherever
possible, with dosing according to age only in situations where the person’s weight is unknown.
Frequency
The frequency of MDA rounds should take into account the local malaria epidemiology, the half-life of the antimalarial
used, and the feasibility and cost of delivering each additional round. Consistent with trial data, mathematical models
predict that a single round of MDA would lead to an initial decrease in infections, but that the duration of effect would
be short-lived. Application of additional rounds is predicted to substantially improve the impact and duration of effect.
MDA should not be given to individuals receiving other forms of malaria chemoprevention (e.g. SMC, PMC, or IPTp)
(Schneider et al unpublished evidence (a)).
Drug resistance
There is limited evidence to date on whether MDA accelerates the development and spread of antimalarial drug
resistance. However, where data were collected, MDA had little to no effect on drug resistance markers (PfKelch13 and
Pfplasmepsin2/3 copy number) among P. falciparum infections (Schneider et al unpublished evidence (a); Plowe unpublished
evidence).
119 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Contraindications
Depending on the medicine chosen, certain population groups may need to be excluded from MDA. These include
pregnant women in their first trimester; infants <6 months of age or weighing <5kg; people recently treated with the
same medicine; people with a known allergy to the medicine; anyone with severe acute illness or who is unable to take
oral medication; people taking medicine known to interact with the medicine used for MDA; and people with specific
contraindications to the medicine used [141].
Other considerations
Information about MDA should be fully accessible to caregivers, health workers and key stakeholders, such as
government officials and religious leaders. As with all health interventions, consent should be obtained, including from
the carers of children, prior to administration of MDA.
Evidence To Decision
• Clinical malaria: MDA may reduce clinical malaria incidence 1–3 months post-MDA1 (rate ratio: 0.41; 95% CI:
0.04–4.42; low-certainty evidence). There was limited evidence available on the effect on malaria burden 4–12
months post-MDA or 12–24 months post-MDA.
• All-cause mortality: It is very uncertain whether MDA affects mortality within the first month post-MDA (risk
ratio: 0.68; 95% CI: 0.57–0.81; very low-certainty evidence) or 1–3 months post-MDA (odds ratio: 1.77; 95%
CI: 1.54–2.04; very low-certainty evidence). No evidence was available from randomized trials and the certainty
of evidence from non-randomized trials was graded very low.
• Parasitaemia: MDA probably reduces the incidence of P. falciparum infection 1–3 months post-MDA (rate ratio:
0.61; 95% CI: 0.40–0.92; moderate-certainty evidence), but may have little to no effect on incidence 4–12
months post-MDA as the evidence is very uncertain (rate ratio: 0.91; 95% CI: 0.55–1.50; very low-certainty
evidence). MDA may result in little to no difference in P. falciparum prevalence 1–3 months (risk ratio: 1.76; 95%
CI: 0.58–5.36; low-certainty evidence) or 4–12 months post-MDA (risk ratio: 1.18; 95% CI: 0.89–1.56; low-
certainty evidence). Evidence from non-randomized trials suggests: MDA may reduce parasite prevalence
12–24 months post-MDA (risk ratio: 0.77; 95% CI: 0.70–0.84; low-certainty evidence), 1–3 months post-MDA
(risk ratio: 0.85; 95% CI: 0.78–0.93; very low-certainty evidence) and 4–12 months post-MDA (risk ratio: 0.60;
95% CI: 0.55–0.67; very low-certainty evidence), but the evidence is very uncertain.
• Adverse events: We are uncertain whether MDA increases or decreases adverse events 1–3 months post-MDA
(odds ratio: 3.25; 95% CI: 0.68–15.53; very low-certainty evidence). No data were available to assess the effect
of MDA on serious adverse events in moderate to high transmission settings, but the absolute risk is very low
(0.01 per 1000 doses).
• Anaemia, drug resistance, hospitalization, severe malaria, or blood transfusions: In the studies that met the
inclusion criteria, none systematically collected data on these outcomes for moderate to high transmission
areas, beyond what was reported as severe adverse events.
• Clinical malaria: MDA may reduce the incidence of clinical malaria due to P. falciparum 1–3 months post-MDA
(rate ratio: 0.58; 95% CI: 0.12–2.73; low-certainty evidence) and 12–24 months post-MDA (rate ratio: 0.77;
95% CI: 0.2–3.03; low-certainty evidence). It is uncertain whether MDA reduces clinical malaria 4–12 months
post-MDA, as the evidence is very uncertain (rate ratio: 0.47; 95% CI: 0.21–1.03; very low-certainty evidence).
• Anaemia: MDA increases mean haemoglobin (mean difference: 0.53; 95% CI: 0.27–0.79; high-certainty
evidence).
• Parasitaemia: MDA probably reduces the incidence of P. falciparum infection 1–3 months post-MDA (rate ratio:
0.37; 95% CI: 0.21–0.66; moderate-certainty evidence). MDA may reduce P. falciparum prevalence 0–1 month
post-MDA (risk ratio: 0.12; 95% CI: 0.03–0.52; moderate-certainty evidence) and probably reduces P. falciparum
prevalence 1–3 months post-MDA (risk ratio: 0.25; 95% CI: 0.15–0.41; moderate-certainty evidence). MDA
may reduce P. falciparum prevalence 4–12 months post-MDA (risk ratio: 0.82; 95% CI: 0.56–1.22; low-certainty
evidence). MDA may reduce P. falciparum prevalence 12–24 months post-MDA, but the evidence is very
uncertain (risk ratio: 0.34; 95% CI: 0.06–1.97; very low-certainty evidence).
• Drug resistance: There was no evidence of an effect on Pfkelch13 or on multi-copy Pfplasmepsin2/3 drug
120 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
resistance markers among those who received three rounds of MDA over three months, compared to the
control.
• Adverse events: MDA may increase the number of serious adverse events within three months (odds ratio:
3.61; 95% CI: 0.43–30.03; moderate-certainty evidence) and 4–12 months post-MDA (odds ratio: 1.47; 95% CI:
0.68–3.20; moderate-certainty evidence). However, the absolute event rate is very low (0.03 per 1000). Four
studies only presented narrative summaries of adverse events. No data were available to assess the effect of
MDA on adverse events in very low to low transmission settings.
• All-cause mortality, hospitalization, severe malaria, or blood transfusions: In the studies that met the inclusion
criteria, none systematically collected data on these outcomes for very low to low transmission areas, beyond
what was reported as severe adverse events.
P. vivax
• Clinical malaria: It is uncertain whether MDA increases or reduces P. vivax malaria 4–12 months post-MDA, as
the evidence is very uncertain (rate ratio: 1.38; 95% CI: 0.97–1.95; very low-certainty evidence). Non-
randomized trials showed that MDA may reduce the incidence of P. vivax malaria at <1 month (rate ratio: 0.23;
95% CI: 0.21–0.25; very low-certainty evidence), 1–3 months (rate ratio: 0.29; 95% CI: 0.26–0.31; very low-
certainty evidence), 4–12 months (rate ratio: 0.72; 95% CI: 0.68–0.76; very low-certainty evidence) or 12–24
months post-MDA (rate ratio: 0.04; 95% CI: 0.02–0.07; very low-certainty evidence), but the evidence is very
uncertain.
• Parasitaemia: MDA probably reduces P. vivax prevalence 0–1 month post-MDA (risk ratio: 0.18; 95% CI:
0.08–0.40; moderate-certainty evidence), and may reduce P. vivax prevalence 1–3 months (risk ratio: 0.15; 95%
CI: 0.10–0.24; low-certainty evidence) and 12–24 months post-MDA (risk ratio: 0.81; 95% CI: 0.44–1.48; low-
certainty evidence). However, MDA may result in little or no difference 4–12 months post-MDA (risk ratio:
1.01; 95% CI: 0.87–1.18; low-certainty evidence). Evidence from non-randomized trials for incidence of P. vivax
infection show that MDA may reduce incidence <1 month after MDA (rate ratio: 0.15; 95% CI: 0.12–0.19; low-
certainty evidence). MDA may reduce P. vivax incidence at 1–3 months (rate ratio: 0.37; 95% CI: 0.32–0.43;
very low-certainty evidence) and 4–12 months post-MDA (rate ratio: 0.15; 95% CI: 0.07–0.34; very low-
certainty evidence), but the evidence is very uncertain.
• Adverse events: With the drugs used in the studies included in the review, MDA probably increases the
frequency of serious adverse events post-MDA (0–3 months post-MDA: odds ratio: 3.61; 95% CI: 0.43–30.03;
moderate-certainty evidence; 4–12 months post-MDA: odds ratio: 1.47; 95% CI: 0.68–3.20; moderate-
certainty evidence).
• Anaemia, all-cause mortality, drug resistance, hospitalization, severe malaria, or blood transfusions: In the
studies that met the inclusion criteria, none systematically collected data on these outcomes for P. vivax
transmission areas, beyond what was reported as severe adverse events.
More information on the evidence can be found in the systematic review (Schneider et al unpublished evidence (a)).
1 In studies with multiple rounds, “post-MDA” refers to after the last round of MDA in a given transmission season or year.
The GDG considered the overall certainty of the evidence for the outcomes of interest to be low. The certainty of
evidence, as summarized under “Benefits and harms”, ranged from very low to high. The priority outcome of
confirmed clinical malaria was assessed as having predominantly low-certainty evidence for P. falciparum
transmission settings and very low-certainty evidence for P. vivax transmission settings. Most studies reported on
outcomes after the last round of MDA, rather than during the intervention period. Studies with multiple rounds of
MDA may not have captured important effects that occurred between the first and last rounds of MDA, and
outcomes may reflect a cumulative effect for MDA. There is a lack of evidence on clinical outcomes during the 0–1
months post-intervention, when impact may be expected to be the greatest. There is no information on
121 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
More information on the certainty of evidence assessments can be found in the ‘research evidence’ tab associated
with this recommendation online or in the annex of the pdf version.
• consultation with civil society, which indicated that chemoprevention to prevent malaria disease is broadly
considered a priority, especially in children under 5 years and women in pregnancy;
• synthesis of contextual factors from trials and pilots of MDA. One study that surveyed participants’ values
found that the most common explanation for the uptake of MDA was the desire to protect their family or
community from future malaria infections.
The GDG determined that there was possibly important uncertainty or variability in how the main outcomes are
valued across contexts, dependent on the transmission setting and burden of disease.
More information on the evidence can be found in the systematic review (Schneider et al unpublished evidence (a))
and the civil society consultation report (CS4ME unpublished evidence).
Resources
The estimated costs per person per round varied from approximately US$ 1.04 to US$ 19.40; one study estimated
that drugs accounted for 70% of the cost of MDA (Schneider et al unpublished evidence (a)). The costs associated
with MDA are likely to vary depending on the extent to which the intervention could leverage existing campaigns
and platforms.
More information can be found in the systematic review (Schneider et al unpublished evidence (a)).
Equity
There was no evidence of a direct impact of MDA on health equity, although the GDG judged that it would likely
increase health equity by enhancing access to medicines for those at risk of malaria. Specific effort may be needed
to reach high-risk communities, among whom uptake tends to be lower, and ethnic minority communities that may
suffer geographical isolation.
More information can be found in the systematic review (Schneider et al unpublished evidence (a)).
Acceptability
MDA is probably acceptable to key stakeholders. Studies have shown that sensitization, education, and inclusion of
local leaders, such as government figures, religious leaders and health authorities, are very important in improving
acceptability. The most common barrier to acceptability is fear of perceived adverse events. Two studies found that
participants were concerned that adverse events may inhibit their economic productivity, although, in another study,
122 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
respondents felt that malaria infection was more likely to limit their economic activity than adverse events.
Previous experience reinforced initial perceptions of MDA: individuals who had been part of previous MDA trials
shared stories in their communities; if those experiences were poor, community members had negative impressions
of MDA. In areas where other malaria interventions had been implemented effectively, MDA for malaria was viewed
more positively.
More information can be found in the systematic review (Schneider et al unpublished evidence (a)).
Feasibility
The feasibility of implementing MDA varies and is highly context-specific, with more remote or mobile populations
being harder to reach.
More information can be found in the systematic review (Schneider et al unpublished evidence (a)).
Justification
This recommendation was developed using the Grading of Recommendations Assessment, Development and Evaluation
(GRADE) framework [103].
Sources of information
A systematic review of existing evidence was commissioned to inform this guidance on the use of MDA to reduce the
burden of malaria disease (Schneider et al unpublished evidence (a)). The review team produced a separate report to
address the needs of the GDG developing the MDA recommendation for transmission reduction. The main objective of
the review was to synthesize evidence on the efficacy and safety of giving a full therapeutic course of antimalarial
medicine at approximately the same time to people residing in defined geographical areas with ongoing human malaria
transmission to reduce the burden of clinical disease from P. falciparum and P. vivax. Secondary objectives included
summarizing evidence on contextual factors that affect the implementation of MDA and findings from mathematical
modelling studies with respect to the impact of different operational factors on MDA efficacy. The primary outcome of
interest was confirmed clinical malaria at 0–1 months, 1–3 months, 4–12 months, and 12–24 months post-MDA.
Secondary outcomes of interest included: hospital admissions (all-cause and malaria-specific); all-cause mortality;
parasite prevalence; adverse events; anaemia; drug resistance; severe malaria; and blood transfusions. The systematic
review was supplemented by a cross-cutting review on chemoprevention drug resistance (Plowe unpublished evidence), a
civil society consultation report on chemoprevention (CS4ME unpublished evidence) and contributions from the GDG
membership, which included national malaria programme representatives.
The systematic review identified 20 studies: eight provided data on P. falciparum (five cluster-randomized controlled
studies and three non-randomized studies); five cluster-randomized controlled trials provided data on both P. falciparum
and P. vivax; and an additional seven studies provided data on P. vivax only (all non-randomized, before-after studies)
(Schneider et al unpublished evidence (a)). The drugs used for MDA in studies evaluating an effect on P. falciparum
included: amodiaquine (1); AS-AQ (1); chloroquine (1); DHAP (8); pyronaridine-artesunate (1); sufalene-pyrimethamine
(1); and SP+AS (2). The drugs used for MDA in studies evaluating an effect on P. vivax included: atebrin (1); chloroquine
(2); chloroquine plus pyrimethamine (1); DHAP (5); and pyrimethamine (3). Seven of the 13 studies evaluating an effect
on P. falciparum included an 8-aminoquinoline, such as low-dose primaquine, as did seven of the 12 studies evaluating an
effect on P. vivax. P. falciparum gametocytes and P. vivax hypnozoites are eliminated by 8-aminoquinolines, but these
drugs may cause haemolysis in people with G6PD deficiency. None of the P. vivax studies included anti-relapse
treatment. Follow-up ranged from 0 to 24 months post-MDA for studies investigating P. falciparum and studies looking
at both P. falciparum and P. vivax, whereas for P. vivax studies, follow-up ranged from 0 to 12 months post-MDA. Studies
that reported data on P. falciparum were stratified into areas of moderate to high (>10% prevalence of P. falciparum
infection) versus low to very low (≤10% prevalence of P. falciparum infection) transmission due to heterogeneity in the
outcomes. Three studies were not included in the review due to an imbalance of background interventions. In addition,
large-scale operational experience of MDA in Central Asia, China and Russian Federation, among others, was not
captured, although MDA has been a prominent feature of control and elimination efforts in those settings.
Summary of judgements
Evidence from the systematic review (Schneider et al unpublished evidence (a)) and supporting information (CS4ME
unpublished evidence; Plowe unpublished evidence) was appraised by the GDG in October 2021. The evidence and their
123 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
judgements are captured in the Evidence-to-Decision table. Where the GDG felt there were differences in moderate to
high versus very low to low transmission areas, a separate assessment was made for each transmission setting. The GDG
determined that the balance of effects favoured MDA for short-term disease burden reduction in moderate to high P.
falciparum transmission settings, given the moderate-certainty evidence that MDA reduces the incidence of P. falciparum
infection 1–3 months post-MDA and has a consistent-sized effect on clinical outcomes. The GDG also considered it
plausible that a reduction in the incidence of infection would translate into an impact on disease. The balance of effects
with regard to burden reduction thus favoured implementation of MDA in moderate to high transmission P. falciparum
settings for short-term reduction of disease burden. There was insufficient evidence from field trials on the impact of
MDA as a long-term (e.g. >1 year) intervention on disease burden in moderate to high transmission areas. In very low to
low P. falciparum transmission settings, the GDG favoured standard care over MDA for malaria disease burden reduction,
given the low certainty of evidence of desirable effects and the low disease burden in low P. falciparum transmission
settings: burden reduction alone was not considered adequate justification for implementing MDA in such settings due
to the small gains in burden reduction from MDA. The overall balance of effects for MDA for burden reduction in P. vivax
transmission settings was not considered by the GDG, given the weak and conflicting available evidence. The GDG
considered that implementation of MDA was associated with moderate costs and that MDA was considered cost-
effective to reduce disease burden in moderate to high transmission settings; however, it was not considered cost-
effective for burden reduction in very low to low transmission settings due to the fewer cases averted in these contexts.
MDA was probably acceptable to key stakeholders, and the feasibility of MDA implementation was deemed variable, as
this is highly context-specific.
Studies evaluating MDA have generally explored the potential of MDA to reduce transmission. Such studies prioritize
infection end-points and this may limit their ability to detect clinical outcomes. The certainty of evidence on clinical
outcomes was considered low, and confidence intervals crossed the null. However, the GDG considered it biologically
plausible that a reduction in the incidence of infection would translate into impact on disease, and recognized that the
point estimates of effect sizes against these end-points were consistent with each other. The GDG concluded that a
conditional recommendation should be made for MDA for short-term burden reduction in moderate to high transmission
settings, given the large impact on burden reduction, low risk of adverse events, moderate costs, likelihood of increasing
equity in terms of access to health interventions, and likely acceptability of short-term MDA in most settings. However,
the feasibility of delivering the intervention could vary and warrants careful consideration in each setting. The GDG
determined that the recommendation should apply to areas with mainly P. falciparum transmission, as there was little and
contradictory evidence for P. vivax.
Implementation
Please refer to the Mass drug administration for falciparum malaria: a practical field manual [137].
Evaluation
Mass drug administration for falciparum malaria: a practical field manual [137] should be used to monitor MDA programmes
for burden reduction. Programmes should include monitoring of efficacy, drug safety and adverse events, drug resistance
and the impact of MDA on morbidity and mortality. Malaria programmes are also encouraged to evaluate the operational
effectiveness and costs of implementation of MDA within their contexts.
Research Needs
Evidence gaps requiring further research include:
• the comparative value of age-targeted chemoprevention (e.g. SMC) vs MDA in terms of disease burden reduction;
• the relative cost-effectiveness of MDA vs targeted chemoprevention (e.g. SMC) for burden reduction;
• the effectiveness of MDA based on different dosing schedules and duration;
• MDA drug choice options for young infants;
• MDA drug choice options for women in their first trimester of pregnancy.
124 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
During emergencies or periods of health service disruption, antimalarial medicine can be used for mass drug
administration (MDA) in defined geographical areas to provide short-term reductions in the burden of disease caused by
P. falciparum.
• MDA may quickly reduce clinical malaria incidence in settings with moderate to high P. falciparum transmission, but the
effect wanes within 1–3 months. As far as possible, MDA should be implemented as part of a package of malaria control
measures (including effective case management and appropriate prevention tools and strategies).
• Malaria programmes should judge the suitability of using MDA in their context based on the desired impact, level of
endemicity, and resources required (see “Practical info”).
• There is very limited evidence on the impact of MDA on disease in emergency settings. However, the biological effects of
MDA on disease in non-emergency settings are likely to translate to MDA recipients in emergency settings. The size of
effect will vary according to the type of emergency and level of disruption to health services, as well as underlying
transmission intensity, choice of drug, delivery method and other factors.
Practical Info
See section 4.2.6.1 for the recommendation on MDA for burden reduction for further practical considerations.
Evidence To Decision
More information on the evidence can be found in the systematic review (Sayre et al unpublished evidence).
The GDG judged the overall certainty of evidence for all critical outcomes to be low.
More information on the certainty of evidence assessments can be found in the ‘research evidence’ tab associated
with this recommendation online or in the annex of the pdf version.
Resources
There was limited evidence on the cost-effectiveness of MDA in emergency settings. One study estimated that
125 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
More information on the evidence can be found in the systematic review (Sayre et al unpublished evidence).
Equity
No evidence was available to assess equity.
Acceptability
Acceptability of MDA was high, despite challenges to implementation in emergency settings.
More information on the evidence can be found in the systematic review (Sayre et al unpublished evidence).
Feasibility
Accurate estimation of the target population, supervision of field staff, and inconsistencies in drug supply were
among the challenges cited in reports of MDA use in emergency settings.
More information on the evidence can be found in the systematic review (Sayre et al unpublished evidence).
Justification
This recommendation was developed using the Grading of Recommendations Assessment, Development and Evaluation
(GRADE) framework [103].
Sources of information
WHO commissioned a systematic review to inform this recommendation on MDA in emergencies or periods of health
service disruption. The systematic review aimed to determine whether people residing in malaria-endemic settings
during an emergency, in a period of health service disruption, or during a febrile illness epidemic should be given an
antimalarial for chemoprevention through MDA. Secondary objectives included summarizing evidence on contextual
factors that affect the implementation of MDA in emergencies. Two studies were included in the quantitative
assessment – neither of which was a randomized controlled trial. These studies were conducted in Sierra Leone and the
Democratic Republic of the Congo with, respectively, two rounds of artesunate-amodiaquine (AS-AQ) given five weeks
apart and two rounds of AS-AQ followed by one round of pyronaridine-artesunate 4–7 weeks apart (Sayre et al
unpublished evidence). The evidence was reviewed by the GDG using the Evidence-to-Decision framework in October
2021.
The overall certainty of the evidence regarding the use of MDA in emergency settings was low and the complexity of
conducting research in emergency settings was noted by the GDG. Despite the limited evidence of MDA impact on
disease in emergency settings, the GDG considered that the biological effects of MDA on disease in non-emergency
settings would likely translate to MDA recipients in emergency settings. The size of effect will likely vary according to the
type of emergency and level of disruption to health services, as well as factors affecting MDA impact such as underlying
transmission intensity, delivery method, and other factors.
Summary of judgements
The GDG determined that the balance between desirable and undesirable effects favoured MDA in emergency settings,
and resource requirements would likely vary depending on the nature of the emergency and the setting. In addition, the
GDG judged that MDA in emergency settings is probably cost-effective; can be feasible, although this will vary
depending on the context; would increase health equity; and is probably acceptable to key stakeholders. Consequently,
the GDG concluded that a conditional recommendation should be made for MDA in emergency settings, highlighting the
strong ethical and moral imperative for malaria prevention in these contexts.
Evaluation
It is acknowledged that the monitoring and evaluation of MDA in emergencies is particularly challenging. However,
programmes should actively consider including systems for monitoring and evaluation to provide evidence for future
126 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
MDA to reduce transmission of P. falciparum in very low to low transmission settings (2022)
In areas with very low to low levels of P. falciparum transmission, antimalarial medicine can be given as chemoprevention
through mass drug administration (MDA) to reduce transmission.
• MDA may quickly reduce transmission of P. falciparum in very low to low transmission areas, but the effect wanes within
1–3 months. Therefore, if MDA is implemented, it should be one of several components of a robust malaria elimination
programme (including, at minimum, good coverage of case-based surveillance with parasitological diagnosis, effective
antimalarial treatment, and appropriate prevention tools and strategies) in order to reduce the risk of resurgence after the
MDA programme has ended.
• MDA should be considered only for geographical areas where there is limited risk of importation of malaria either from
adjacent communities or through travel of the population to endemic areas.
• Malaria programmes should consider whether sufficient resources are available to implement MDA without affecting other
components of a robust malaria elimination programme.
• Very low to low transmission settings are defined as areas with P. falciparum parasite prevalence less than 10%, or P.
falciparum incidence less than 250 cases per 1000 population per year [30]. These thresholds should not be regarded as
absolutes for determining applicability of MDA implementation for transmission reduction. MDA implemented in areas
with levels of transmission near these cut-offs may reduce both disease burden and transmission intensity.
Practical Info
The WHO guidance document, Mass drug administration for falciparum malaria: a practical field manual provides technical
and operational guidance on the practical aspects of organizing a successful MDA program [141].
MDA has been found to have a short-term (1–3 months) impact on P. falciparum transmission in very low to low
transmission areas. For MDA to contribute meaningfully towards achievement of malaria elimination, activities must
already be in place to capitalize on the reduction in transmission achieved through the strategy. For that reason, if MDA
is implemented, it should be as one component of a robust malaria elimination programme that includes, at minimum,
good coverage of case-based surveillance, quality-assured parasitological diagnosis, effective antimalarial treatment and
additional prevention strategies such as vector control. MDA will have maximal benefit to an elimination programme if
the aim is to reduce transmission to the level that intensive surveillance and follow-up of every case can begin.
MDA is likely to be most effective at reducing transmission in geographical areas where there is limited risk of
importation of malaria either from adjacent communities or through travel of the population to endemic areas.
Additionally, MDA rounds should be scheduled for time periods when populations exhibit low levels of movement in and
out of the area in order to increase coverage of the intervention and reduce risk of importation. The impact of MDA will
be greater, and last longer, if a large proportion of the population present in the area benefits from the treatment and
prophylaxis provided by the medicine and if the rate of parasite importation is low.
The frequency of rounds and duration of the MDA programme should take into account the local malaria epidemiology,
the length of the prophylactic period provided by the antimalarial used, and the feasibility and cost of delivering each
additional round. Consistent with trial data, mathematical models predicted that a single round of MDA would lead to an
initial decrease in infections, but that the duration of effect would be short lived. Application of additional rounds is
predicted to substantially improve the impact and duration of effect, but attempts should be made in later rounds to
reach individuals who did not participate in earlier rounds.
Achieving high coverage of the population and good adherence to the antimalarial medicine are critical aspects of MDA
programmes. MDA programmes ask many asymptomatic, healthy people to take a medicine when they do not feel ill,
with the potential for adverse reactions to occur. Improving coverage and adherence requires development of
understanding and trust in the institutions implementing the programme. Community engagement is thus a key factor in
127 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
determining the success of MDA in order to improve participation rates and adherence to the full treatment course of
the medicine.
A complete therapeutic course of antimalarial medicine, at doses recommended by the manufacturer, should be given to
all eligible adults and children within the defined geographic area. Drug dosage should be determined by weight
wherever possible, with dosing according to age only in situations where the person’s weight is unknown. The
antimalarial medicines chosen for use in MDA should: a) be WHO recommended and prequalified; b) be efficacious
against local parasites; c) be different from the medicine used as first-line treatment, where possible c) have a superior
safety and tolerability profile; d) provide a longer duration of post-treatment prophylaxis with component medicines that
have closely matched pharmacology to reduce the risk of new infections encountering only a single drug; e) have a
positive public reputation and acceptability and f) be available and low-cost. Programmes may consider including a
single, low-dose of primaquine in MDA programmes in order to increase the gametocytocidal effect, although the
evidence was insufficient to discern an additional benefit of single low-dose primaquine. A drug regimen that can be
administered as a directly-observed single dose is preferred to multi-day regimens.
Depending on the medicine chosen, certain population groups may need to be excluded from MDA, such as: pregnant
women in their first trimester; infants < 6 months of age or weighing < 5kgs; people recently treated with the same
medicine; people with a known allergy to the medicine; anyone with severe acute illness or unable to take oral
medication; people taking medication known to interact with the medicine used for MDA; and people with specific
contraindications to the medicine used [141]. MDA should not be given to individuals receiving other forms of malaria
chemoprevention (e.g. seasonal malaria chemoprevention, perennial malaria chemoprevention, or intermittent preventive
treatment during pregnancy).
Evidence To Decision
• MDA probably reduces P. falciparum prevalence (risk difference [RD]: -18 cases per 1000 persons; 95% CI: -20
to -14 per 1000 persons; eight cRCTs; moderate-certainty evidence).
• MDA probably reduces the incidence of P. falciparum (RD: -8 cases per 1000 p-y; 95% CI: -10 to -4 per 1000 p-
y; one cRCT; moderate-certainty evidence).
• MDA may result in little to no difference in the incidence of P. falciparum clinical malaria (RD: -3 cases per 1000
p-y; 95% CI: -5 to 11 per 1000 p-y; two cRCTs; low-certainty evidence).
• MDA may result in little to no difference in P. falciparum prevalence (RD: -3 per 1000 persons; 95% CI: -8 to 4
per 1000 persons; six cRCTs; low-certainty evidence).
• The evidence is very uncertain about the effect of MDA on the incidence of P. falciparum clinical malaria (RD: -6
per 1000 p-y; 95% CI: -9 to 0 per 1000 p-y; four cRCTs; very low-certainty evidence).
• The evidence is very uncertain about the effect of MDA on the prevalence of P. falciparum (RD: -21 per 1000
persons; 95% CI: -30 to 31 per 1000 persons; one cRCT; very low-certainty evidence).
• MDA may reduce the incidence of P. falciparum clinical malaria (RD: -4 per 1000 p-y; 95% CI: 14 to 34 per
1000 p-y; one cRCT; low-certainty evidence).
128 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• At 0–3 months, MDA probably has little to no effect on serious adverse events (RD: 1 per 1000 persons; 95%
CI: 0 to 11 per 1000 persons; one cRCT; moderate-certainty evidence).
• At 4–12 months, MDA may increase serious adverse events slightly (RD: 2 per 1000 persons; 95% CI: -1 to 8
per 1000 persons; one cRCT; moderate-certainty evidence).
• Among people who participated in MDA, the rate of serious adverse events was 0.03 per 1000 doses of
antimalarial medicine (four cRCTs; not GRADEd because no information was available from the comparator
arm).
Adverse events
• At 1–3 months, the evidence is very uncertain about the effect of MDA on adverse events (RD: 300 per 1000
persons; 95% CI: -43 to 1 937 per 1000 persons; one cRCT; very low-certainty evidence).
• Among people who participated in MDA, the rate of adverse events was 4.6 per 1000 doses of antimalarial
medicine (four cRCTs; not GRADEd because no information was available from the comparator arm).
• At 1–3 months after the last round, the evidence is very uncertain about the effect of MDA on artemisinin
resistance markers (PfKelch13) among P. falciparum infections (RD: -109 per 1000 persons; 95% CI: -334 to 310
per 1000 persons; one cRCT very low-certainty evidence).
• At 1–3 months after the last round MDA may reduce the proportion of artemisinin resistance markers
(PfKelch13) among all participants (RD: -56 per 1000 persons; 95% CI: -61 to -45 per 1000 persons; one cRCT;
low-certainty evidence).
• At 4–12 months after the last round, the evidence is very uncertain about the effect of MDA on the proportion
of infections with artemisinin resistance markers (PfKelch13) among all P. falciparum infections (RD: 98 per
1000 persons; 95% CI: -104 to 372 per 1000 persons; one cRCT; very low-certainty evidence).
• At 4–12 months after the last round, MDA may reduce the proportion of artemisinin resistance markers
(PfKelch13) among all participants (RD: -15 per 1000 persons; 95% CI: -21 to -4 per 1000 persons; one cRCT;
low-certainty evidence).
• At 12–24 months after the last round, the evidence is very uncertain about the effect of MDA on the
proportion of infections with artemisinin resistance markers (PfKelch13) among all P. falciparum infections (RD:
50 per 1000 persons; 95% CI: -129 to 286 per 1000 persons; one cRCT; very low-certainty evidence).
• At 12–24 months after the last round, MDA may reduce the proportion of artemisinin resistance markers
(PfKelch13) among all participants (RD: -9 per 1000 persons; 95% CI: -15 to 3 per 1000 persons; one cRCT;
low-certainty evidence).
129 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The GDG judged that there may be important uncertainty or variability in the preferences or values that could not
be determined due to the lack of studies.
Resources
The systematic review identified four studies with information on resource needs for MDA (Schneider et al
unpublished evidence (b)). The cost of MDA varied from ~US$ 1.04 to US$ 19.40 per person per round; one study
estimated that drugs accounted for 70% of the cost of MDA. Compared to reactive drug administration (RDA),
MDA was superior in all cost-effectiveness measures, including cost per infection averted, cost per case averted,
cost per death averted, and cost per disability-adjusted life year (DALY) averted. Furthermore, the cost of MDA per
person reached was substantially lower in an operational setting (US$ 2.90) than in a research setting (US$ 4.71).
The GDG judged the resources required to implement MDA to be large. The GDG found it difficult to judge the cost-
effectiveness of MDA as the evidence of an effect was of low certainty, and both the effectiveness and cost of the
intervention are likely to vary depending on the time period over which outcomes are measured and whether
elimination is achieved. However, the GDG concluded that cost-effectiveness in very low to low transmission areas
probably favoured the intervention.
Equity
No studies were identified that addressed the issue of whether MDA increased or decreased health equity.
The GDG judged that the impact of MDA on equity is likely to vary. While MDA has the potential to reach people
who might have difficulty accessing other malaria prevention and treatment services, MDA might also expose many
people to antimalarials who were not infected. The GDG felt that MDA could exacerbate inequity if not
implemented appropriately or if implementation resulted in only a small, temporary effect. However, if
implementation of MDA contributed to elimination of P. falciparum, then the intervention would likely improve
equity.
Acceptability
The systematic review identified 18 studies with information on acceptability (Schneider et al unpublished evidence
(b)). The most common barrier to acceptability of MDA reported in the literature was fear of adverse events. Two
studies found that participants were concerned that adverse events from MDA might inhibit their economic
productivity, although in another study respondents felt that malaria infection was more likely to limit economic
activity than adverse events from MDA.
One study found that, in addition to sensitization on the benefits of MDA, providing healthcare to communities
participating in MDA helped to reduce concerns about adverse effects; however, another study found that the
presence of expatriate physicians, an ambulance, and the unfamiliar informed consent process elevated rather than
reduced concerns. Previous experience reinforced initial perceptions of MDA: individuals who had been part of
previous MDA trials shared stories in their communities; if those experiences were poor, community members had
negative impressions of MDA. In areas where other malaria interventions had been implemented effectively, MDA
for malaria was viewed more positively. One study found that reported acceptability of MDA increased from 62%
before the intervention to 98% after, while the proportion of respondents who answered that MDA could cause side
effects decreased from 30% to 20% in the same timeframe.
Common themes in analyses of drivers of acceptance were sensitization or education about the intervention,
support from a range of local authority figures, and additional health support. One study reported that “Respondents
who felt that they have received enough information… were more likely to participate in all rounds of MDA,” a theme
that was reiterated in five other studies.
One study found that a lack of engagement with local healthcare providers limited adherence due to conflicting
messages around the efficacy of MDA.
130 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The GDG judged the acceptability of MDA for P. falciparum in very low to low transmission settings to vary
depending on whether factors that affect community and individual acceptability have been appropriately addressed
in the design of the intervention. The GDG considered that a country’s previous experience with MDA, whether
positive or negative, was likely to affect their level of acceptance of the intervention. The GDG suggested that a key
consideration was whether malaria programme staff find MDA to be an acceptable intervention, but no surveys of
this key stakeholder were identified.
Feasibility
The systematic review identified 13 studies providing information on the feasibility of implementation of MDA
(Schneider et al unpublished evidence (b)). Ten studies described barriers to implementing MDA due to residents’
absence. Of these, three studies noted that absenteeism was one of the major driving forces of non-adherence to
medicine. One study noted that determining participants’ seasonal mobility prior to the MDA campaign had
contributed to the success of the campaign. Three studies noted difficulties related to determining the optimal
timing of the MDA campaign: weather-related challenges, agricultural activities, overlaps with religious events,
especially those involving fasting, unpredictable policy changes at the national level and the school year. Feasibility
concerns related to participants’ religion were further noted in one study that attempted to implement directly
observed drug administration but found that some women were unwilling to remove their face coverings in front of
strangers. This issue was resolved by creating sequestered administration sites staffed by accepted local staff.
The GDG judged the feasibility of implementing MDA to vary depending on the size of the population, with
improved feasibility in smaller populations and island communities.
Justification
The systematic review of the impact of MDA on P. falciparum identified significant heterogeneity in the meta-analysis of
a key outcome (prevalence of infection 1–3 months after the last round of MDA) (Schneider et al unpublished evidence
(b)). A subgroup analysis found that the heterogeneity between studies could be explained by differences between
higher and lower transmission settings. In the systematic review, a cut-off of 10% prevalence of P. falciparum infection
and incidence of 250 P. falciparum cases per 1000 population per year was used to differentiate between areas of very
low to low transmission and areas of moderate to high transmission. As higher transmission settings have a larger
parasite reservoir, higher rate of new infections and often greater vectorial capacity than lower transmission settings, it is
biologically plausible for MDA to have a differential impact on transmission reduction depending on the transmission
setting. As a result, the systematic review stratified all analyses by transmission setting, and separate recommendations
were developed on the use of MDA for reducing transmission of P. falciparum in very low to low and moderate to high
transmission areas.
The GDG concluded that the balance of effects probably favoured implementation of MDA to reduce P. falciparum
transmission in very low to low transmission settings although there were concerns about the sustainability of impact if
only one or two rounds are implemented. The GDG judged that the resources required for implementation of MDA were
large and could impact negatively on the implementation of other recommended malaria prevention strategies. While
there were limited data on cost-effectiveness, the GDG judged that cost-effectiveness probably favoured MDA but
would depend on the time period over which outcomes were measured; if elimination were achieved, in part, through
MDA, the cost-effectiveness would be very high. The GDG judged that the acceptability of the intervention was likely to
vary depending on the stakeholder group and the population’s previous experience with MDA. The feasibility of
implementing the intervention was judged to vary depending on the size of the population to be covered. The GDG
concluded that a conditional recommendation for MDA for P. falciparum in very low to low transmission settings should
be issued given the moderate-certainty evidence for a short-term benefit, variability around issues such as acceptability
and feasibility and large resource requirements.
Research Needs
• Further evidence is needed on the impact (incidence or prevalence of malaria infection at the community level) and
potential harms/unintended consequences of MDA for P. falciparum in very low to low transmission areas, including
resistance to antimalarial medicines. Evidence of impact disaggregated by sex, age and socioeconomic status is
needed to understand whether there are any equity considerations.
• Determine the optimal timing and number of MDA rounds to maximize the impact (incidence or prevalence of
131 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
malaria infection at the community level) of MDA on P. falciparum in very low to low transmission areas.
• Determine the minimum effective coverage of MDA in the population to maximize the impact (incidence or
prevalence of malaria infection at the community level) of MDA on P. falciparum in very low to low transmission
areas.
• Determine whether multiple years of effective coverage of MDA as part of an elimination programme is feasible and
acceptable and whether it can contribute to interrupting P. falciparum transmission in very low to low transmission
areas.
• Investigate approaches to improving the acceptability of MDA and adherence to antimalarial medicines in very low
to low transmission areas.
• Determine whether the addition of single, low-dose primaquine modifies the impact (incidence or prevalence of
malaria infection at the community level) of MDA on P. falciparum in very low to low transmission areas.
In areas with moderate to high levels of P. falciparum transmission, providing antimalarial medicine through mass drug
administration (MDA) to reduce transmission is not recommended.
• The studies included in the systematic review did not demonstrate evidence that MDA has either a short- or long-term
effect on P. falciparum transmission in moderate to high transmission settings.
• Recommendations on MDA to reduce the burden of malaria in moderate to high transmission settings can be found in
section 4.2.4.1 MDA for burden reduction. Moderate to high transmission settings are defined as areas with P. falciparum
parasite prevalence greater than 10%, or P. falciparum incidence above 250 cases per 1000 population per year [30].
These thresholds should not be regarded as absolutes for determining applicability of MDA.
Evidence To Decision
• MDA may result in little to no difference in P. falciparum prevalence (RD: 38 cases per 1000 persons; 95% CI:
-21 to 219 per 1000 persons; one cRCT; low-certainty evidence).
• The evidence is very uncertain about the effect of MDA on P. falciparum prevalence (RD: -108 per 1000
persons; 95% CI: -159 to -51 per 1000 persons; one NRS; very low-certainty evidence).
• MDA probably reduces the incidence of P. falciparum parasitaemia (RD: -22 per 1000 p-y; 95% CI: -34 to -5 per
1000 p-y; one cRCT; moderate-certainty evidence).
• MDA may result in little to no difference in the incidence of P. falciparum clinical malaria (RD: -1 per 1000 p-y;
95% CI: -2 to 8 per 1000 p-y; one cRCT; low-certainty evidence).
• MDA may result in little to no difference in P. falciparum prevalence (RD: -87 per 1000 persons; 95% CI: -53 to
271 per 1000 persons; one cRCT; low-certainty evidence).
132 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• MDA may reduce P. falciparum prevalence (RD: -167 per 1000 persons; 95% CI: -188 to -138 per 1000 persons;
one NRS; low-certainty evidence)
• The evidence is very uncertain about the effect of MDA on the incidence of P. falciparum parasitaemia (RD: -10
per 1000 p-y; 95% CI: -49 to 54 per 1000 p-y; one cRCT; very low-certainty evidence).
• MDA may reduce P. falciparum prevalence (RD: -99 per 1000 p-y; 95% CI: -129 to -69 per 1000 p-y; one NRS;
low-certainty evidence).
• Among people who participated in MDA, the rate of serious adverse events was 0.01 per 1000 doses of
antimalarial medicine (one cRCT; not GRADEd because no information was available from the comparator arm).
Adverse events
• The evidence is very uncertain about the effect of MDA on adverse events (RD: 200 per 1000 persons; 95% CI:
-39 to 572 per 1000 persons; one cRCT; very low-certainty evidence).
• Among people who participated in MDA, the rate of adverse events was 2.0 per 1000 doses of antimalarial
medicine (one cRCT; not GRADEd because no information was available from the comparator arm).
The GDG judged that there may be important uncertainty or variability in preferences or values that could not be
determined due to the lack of studies.
Resources
The systematic review identified four studies with information on resource needs for MDA (Schneider et al
unpublished evidence (b)). The cost of MDA varied from ~US$ 1.04 to US$ 19.40 per person per round; one study
estimated that drugs accounted for 70% of the cost of MDA. Compared to reactive drug administration (RDA),
MDA was superior in all cost-effectiveness measures, including cost per infection averted, cost per case averted,
cost per death averted, and cost per disability-adjusted life year (DALY) averted. Furthermore, the cost of MDA per
person reached was substantially lower in an operational setting (US$ 2.90) than in a research setting (US$ 4.71).
The GDG judged the resources required to implement MDA to be large. The GDG judged that cost-effectiveness
133 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
probably favoured no MDA but found it difficult to judge the cost-effectiveness of MDA as the evidence for an
effect was of very low certainty and both the effectiveness and cost of the intervention are likely to vary depending
on the time period over which they are measured.
Equity
The systematic review did not identify any research that addressed the issue of how MDA affects health equity.
The GDG judged the impact of implementing MDA on equity to vary. While MDA had the potential to reach people
who might have difficulty accessing other malaria prevention and treatment services, it also exposes uninfected
people to the potential adverse effects of antimalarials. The GDG felt that MDA could exacerbate inequity if not
implemented appropriately or if implementation resulted only in a small, temporary effect.
Acceptability
The systematic review identified 18 studies with information on acceptability (Schneider et al unpublished evidence
(b)). The most common barrier to acceptability of MDA reported in the literature was fear of adverse events. Two
studies found that participants were concerned that adverse events from MDA might inhibit their economic
productivity, although in another study respondents felt that malaria infection was more likely to limit economic
activity than adverse events.
One study found that, in addition to sensitization on the benefits of MDA, providing healthcare to communities
participating in MDA helped to reduce concerns about adverse effects; however, another study found that the
presence of expatriate physicians, an ambulance, and the unfamiliar informed consent process elevated rather than
reduced concerns. Previous experience reinforced initial perceptions of MDA: individuals who had been part of
previous MDA trials shared stories in their communities; if those experiences were poor, community members had
negative impressions of MDA. In areas where other malaria interventions had been implemented effectively, MDA
for malaria was viewed more positively. One study found that reported acceptability of MDA increased from 62%
before the intervention to 98% after, while the proportion of respondents who answered that MDA could cause side
effects decreased from 30% to 20% in the same timeframe.
Common themes in analyses of drivers of acceptance were sensitization or education about the intervention,
support from a range of local authority figures, and additional health support. One study reported that “Respondents
who felt that they have received enough information… were more likely to participate in all rounds of MDA,” a theme
that was reiterated in five other studies.
One study found that a lack of engagement with local healthcare providers limited adherence due to conflicting
messages around the efficacy of MDA.
The GDG judged the acceptability of MDA for P. falciparum in moderate to high transmission settings to depend on
whether factors that affect community and individual acceptability have been appropriately addressed in the design
of the intervention. The GDG considered that a country’s previous experience with MDA, whether positive or
negative, was likely to affect their level of acceptance of the intervention. The GDG suggested that a key
consideration was whether malaria programme staff find MDA to be an acceptable intervention, but no surveys of
this key stakeholder were identified.
Feasibility
The systematic review identified 13 studies providing information on the feasibility of implementation of MDA
(Schneider et al unpublished evidence (b)). Ten studies described barriers to implementing MDA due to residents’
absence. Of these, three studies noted that absenteeism was one of the major driving forces of non-adherence to
medicine. One study noted that determining participants’ seasonal mobility prior to the MDA campaign had
contributed to the success of the campaign. Three studies noted difficulties related to determining the optimal
timing of the MDA campaign: weather-related challenges, agricultural activities, overlaps with religious events,
134 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
especially those involving fasting, unpredictable policy changes at the national level and the school year. Feasibility
concerns related to participants’ religion were further noted in one study that attempted to implement directly
observed drug administration but found that some women were unwilling to remove their face coverings in front of
strangers. This issue was resolved by creating sequestered administration sites staffed by accepted local staff.
The GDG judged the feasibility of implementing MDA to vary depending on the size of the population, with
improved feasibility in smaller populations and island communities.
Justification
The GDG judged that the balance of effects probably favoured not implementing MDA to reduce P. falciparum
transmission in moderate to high transmission settings. The GDG judged that the resources required for implementation
of MDA were large and could impact negatively on the implementation of other recommended malaria prevention
strategies. While cost-effectiveness data were limited, the GDG judged that cost-effectiveness probably did not favour
MDA in moderate to high transmission settings. The GDG judged that the acceptability of the intervention was likely to
vary depending on the stakeholder group and previous experience of the population with MDA. The feasibility of
implementing the intervention was judged to vary depending on the size of the population to be covered. The GDG
concluded that there should be a conditional recommendation against the implementation of MDA to reduce
transmission of P. falciparum in moderate to high transmission settings given the lack of evidence for either a short- or
long-term benefit, variability around issues such as acceptability and feasibility and large resource requirements.
In areas with P. vivax transmission, antimalarial medicine can be given as chemoprevention through mass drug
administration (MDA) to reduce transmission.
• MDA may quickly reduce transmission of P. vivax, but the effect wanes within 1–3 months. Therefore, if MDA is
implemented, it should be one of several components of a robust malaria elimination programme (including, at minimum,
good coverage of case-based surveillance with parasitological diagnosis, effective antimalarial treatment including
treatment for hypnozoites, and appropriate prevention tools and strategies) in order to reduce the risk of resurgence after
the MDA programme has ended.
• MDA should be considered only for geographical areas where there is limited risk of importation of malaria either from
adjacent communities or through travel of the population to endemic areas.
• Malaria programmes should consider whether sufficient resources are available to implement MDA without affecting other
components of a robust malaria elimination programme.
• Programmes considering implementing MDA for P. vivax should carefully reflect on how to safely and feasibly administer
treatment to prevent relapses.
Practical Info
MDA without an 8-aminoquinoline medicine may have a short-term (1–3 months) impact on P. vivax transmission. For
MDA to contribute meaningfully towards achievement of malaria elimination, activities must already be in place to
capitalize on the reduction in transmission achieved through the strategy. For that reason, MDA should be implemented
as a component of a robust malaria elimination programme that includes, at minimum, good coverage of case-based
surveillance, quality-assured parasitological diagnosis, effective antimalarial treatment and additional prevention
strategies such as vector control. MDA will have maximal benefit to an elimination programme if the aim is to reduce
transmission to the level that intensive surveillance and follow-up of every case can begin.
MDA is likely to be most effective at reducing transmission in geographical areas where there is limited risk of
importation of malaria either from adjacent communities or through travel of the population to endemic areas.
Additionally, MDA rounds should be scheduled for time periods when populations exhibit low levels of movement in and
135 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
out of the area in order to increase coverage of the intervention and reduce risk of importation. The impact of MDA will
be greater, and last longer, if a large proportion of the population present in the area benefits from the treatment and
prophylaxis provided by the medicine and if the rate of parasite importation is low.
The frequency of rounds and duration of the MDA programme should take into account the local malaria epidemiology,
the half-life of the antimalarial used, and the feasibility and cost of delivering each additional round. Consistent with trial
data, mathematical models predicted that a single round of MDA would lead to an initial decrease in infections, but that
the duration of effect would be short lived. Application of additional rounds is predicted to substantially improve the
impact and duration of effect.
Achieving high coverage of the population and good adherence to the antimalarial medicine are critical aspects of MDA
programmes. MDA programmes ask many asymptomatic, healthy people to take a medicine when they do not feel ill,
with the potential for adverse reactions to occur. Improving coverage and adherence requires development of
understanding and trust in the institutions implementing the programme. Community engagement is thus a key factor in
determining the success of MDA, to improve participation rates and adherence to the full treatment course of the
medicine.
A complete therapeutic course of antimalarial medicine, at doses recommended by the manufacturer, should be given to
all eligible adults and children within the defined geographic area. Drug dosage should be determined by weight
wherever possible, with dosing according to age only in situations where the person’s weight is unknown. The
antimalarial medicines chosen for use in MDA should: a) be WHO recommended and prequalified; b) be efficacious
against local parasites; c) be different from the medicine used as first-line treatment, where possible c) have a superior
safety and tolerability profile; d) provide a longer duration of post-treatment prophylaxis with component medicines that
have closely matched pharmacology to reduce the risk of new infections encountering only a single drug; e) have a
positive public reputation and acceptability and f) be available and low-cost. A drug regimen that can be administered as
a directly-observed single dose is preferred to multi-day regimens.
Depending on the medicine chosen, certain population groups may need to be excluded from MDA, such as: pregnant
women in their first trimester; infants < 6 months of age or weighing <5kgs; people recently treated with the same
medicine; people with a known allergy to the medicine; anyone with severe acute illness or unable to take oral
medication; people taking medication known to interact with the medicine used for MDA; and people with specific
contraindications to the medicine used [141]. MDA should not be given to individuals receiving other forms of malaria
chemoprevention (e.g. seasonal malaria chemoprevention, perennial malaria chemoprevention, or intermittent preventive
treatment during pregnancy).
MDA for P. vivax is complicated because many P. vivax infections are likely to be dormant stages (hypnozoites) in the liver
that will not be cured unless an 8-amnoquinoline, the only type of medicine that treats hypnozoites, is administered.
Without provision of an 8-aminoquinoline, a large proportion of P. vivax cases treated in the MDA programme will
relapse within a few months. However, programmes contemplating providing medicine for radical cure of P. vivax as part
of MDA should carefully consider whether it is feasible to administer this treatment regimen safely, i.e. with testing for
G6PD deficiency prior to treatment, an effective pharmacovigilance system and emergency access to blood transfusion
services. Programmes should also consider whether sufficient coverage and adherence to the full course of radical cure
can be achieved.
Evidence To Decision
• MDA may reduce P. vivax prevalence (RD: -113 per 1000 persons; 95% CI: -119 to -101 per 1000 persons; five
136 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• MDA may result in little to no difference in P. vivax prevalence (RD: 1 per 1000 persons; 95% CI: -12 to 17 per
1000 persons; five cRCTs; low-certainty evidence).
• The evidence is very uncertain about the effect of MDA on the prevalence of P. vivax (RD: -47 per 1000
persons;95% CI: -60 to -16 per 1000 persons; one NRS; very low-certainty evidence).
• The evidence is very uncertain about the effect of MDA on the incidence of P. vivax clinical malaria (RD: -4 per
1000 p-y; 95% CI: -4 to -3 per 100 p-y; one NRS; very low-certainty evidence).
• The evidence is very uncertain about the effect of MDA on the incidence of P. vivax clinical malaria (RD: -44 per
1000 p-y; 95% CI: -50 to -37 per 1000 p-y; one cRCT; very low-certainty evidence).
• MDA may result in little to no difference in P. vivax prevalence (RD: -33 per 1000 persons; 95% CI: -98 to 84
per 1000 persons; one cRCT; low-certainty evidence).
• The evidence is very uncertain about the effect of MDA on the incidence of P. vivax clinical malaria (RD: -150
per 1000 p-y; -153 to -145 per 1000 p-y; one NRS; very low-certainty evidence).
• MDA probably results in little to no difference in serious adverse events within 0–3 months of the last round of
MDA (RD: 1 per 1000 persons; 95% CI: 0 to 11 per 1000 persons; one cRCT; moderate-certainty evidence).
• MDA probably results in little to no difference in serious adverse events 4–12 months after the last round of
MDA (RD: 2 per 1000 persons; 95% CI: -1 to 8 per 1000 persons; one cRCT; moderate-certainty evidence).
• Among people who participated in MDA, the rates of adverse events and serious adverse events were 19.9 per
1000 and 0.3 per 1000 doses of antimalarial medicine, respectively (two cRCTs; not GRADEd because no
information was available from the comparator arm).
Cognizant of the limitations of the available evidence, the GDG judged that the sizes of both the desirable and
137 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
undesirable effects were small, and the balance of effects did not favour either MDA or no MDA for P. vivax.
1The systematic review considered the following as the minimum adult dosage of 8-aminoquinoline medicines to achieve radical cure: 210 mg of
primaquine over eight weeks; 1.25 g of plasmochin over 14 days. One study considered its primaquine adult dosage regimen (40 mg of
primaquine every two weeks for two years) to be radical cure, but as the total dose for an eight-week period (i.e. 160 mg) was less than 210 mg,
the systematic review did not consider this to be radical cure (Schneider et al unpublished evidence (b)).
The GDG judged that there may be important uncertainty or variability in the preferences or values that could not
be determined due to the lack of studies.
Resources
The systematic review identified four studies with information on resource needs for MDA (Schneider et al
unpublished evidence (b)). The cost of MDA varied from ~US$ 1.04 to US$ 19.40 per person per round; one study
estimated that drugs accounted for 70% of the cost of MDA. Compared to reactive drug administration (RDA),
MDA was superior in all cost-effectiveness measures, including cost per infection averted, cost per case averted,
cost per death averted, and cost per disability-adjusted life year (DALY) averted. Furthermore, the cost of MDA per
person reached was substantially lower in an operational setting (US$ 2.90) than in a research setting (US$ 4.71).
The GDG judged the resources required to implement MDA to be large. The GDG found it difficult to judge the cost-
effectiveness of MDA as there were no data on cost or cost-effectiveness identified in the studies of P. vivax. The
GDG judged that the effectiveness and cost of MDA are likely to vary depending on the time period over which they
are measured and whether elimination is achieved.
Equity
The systematic review did not identify any research that addressed the issue of how MDA affects health equity.
The GDG judged the impact of implementing MDA on equity to vary. While MDA had the potential to reach people
who might have difficulty accessing other malaria prevention and treatment services, it also exposes uninfected
people to the potential adverse effects of antimalarials. The GDG felt that MDA could exacerbate inequity if not
implemented appropriately or if implementation resulted only in a small, temporary effect.
Acceptability
The systematic review identified 18 studies with information on acceptability (Schneider et al unpublished evidence
(b)). The most common barrier to acceptability of MDA reported in the literature was fear of adverse events. Two
studies found that participants were concerned that adverse events from MDA might inhibit their economic
productivity, although in another study respondents felt that malaria infection was more likely to limit economic
activity than adverse events.
One study found that, in addition to sensitization on the benefits of MDA, providing healthcare to communities
participating in MDA helped to reduce concerns about adverse effects; however, another study found that the
138 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
presence of expatriate physicians, an ambulance, and the unfamiliar informed consent process elevated rather than
reduced concerns. Previous experience reinforced initial perceptions of MDA: individuals who had been part of
previous MDA trials shared stories in their communities; if those experiences were poor, community members had
negative impressions of MDA. In areas where other malaria interventions had been implemented effectively, MDA
for malaria was viewed more positively. One study found that reported acceptability of MDA increased from 62%
before the intervention to 98% after, while the proportion of respondents who answered that MDA could cause side
effects decreased from 30% to 20% in the same timeframe.
Common themes in analyses of drivers of acceptance were sensitization or education about the intervention,
support from a range of local authority figures, and additional health support. One study reported that “Respondents
who felt that they have received enough information… were more likely to participate in all rounds of MDA,” a theme
that was reiterated in five other studies.
One study found that a lack of engagement with local healthcare providers limited adherence due to conflicting
messages around the efficacy of MDA.
The GDG judged that the acceptability of MDA for P. vivax would vary depending on whether factors that affect
community and individual acceptability have been appropriately addressed in the design of the intervention.
The GDG considered that a country’s previous experience with MDA, whether positive or negative, was likely to
affect their level of acceptance of the intervention. The GDG suggested that a key consideration is whether malaria
programme staff find MDA to be an acceptable intervention, but no surveys of this key stakeholder were identified.
The GDG felt that the inclusion of an 8-aminoquinoline in MDA for radical cure would likely have a negative effect
on the acceptability of the intervention due to safety concerns and the long treatment period.
Feasibility
The systematic review identified 13 studies providing information on the feasibility of implementation of MDA
(Schneider et al unpublished evidence (b)). Ten studies described barriers to implementing MDA due to residents’
absence. Of these, three studies noted that absenteeism was one of the major driving forces of non-adherence to
medicine. One study noted that determining participants’ seasonal mobility prior to the MDA campaign had
contributed to the success of the campaign. Three studies noted difficulties related to determining the optimal
timing of the MDA campaign: weather-related challenges, agricultural activities, overlaps with religious events,
especially those involving fasting, unpredictable policy changes at the national level and the school year. Feasibility
concerns related to participants’ religion were further noted in one study that attempted to implement directly
observed drug administration but found that some women were unwilling to remove their face coverings in front of
strangers. This issue was resolved by creating sequestered administration sites staffed by accepted local staff.
The GDG judged the feasibility of implementing MDA for P. vivax to vary depending on the size of the population,
with improved feasibility in smaller populations and island communities. Feasibility would also vary depending on
whether radical cure using an 8-aminoquinoline medicine was part of the MDA strategy, which would necessitate
testing for G6PD deficiency, an effective pharmacovigilance system and emergency access to blood transfusion
services.
Justification
The GDG concluded that the balance of effects did not favour either MDA or no MDA to reduce P. vivax transmission.
There was a lack of studies evaluating the efficacy and safety of MDA drug regimens that included an 8-aminoquinoline
for radical cure of P. vivax; the GDG expressed concern both for the likely decreased long-term effectiveness of MDA for
P. vivax without use of an 8-aminoquinoline and the increased complexity of safely administering 8-aminoquinolines. The
GDG judged that the resources required for implementation of MDA were large and could impact negatively on the
implementation of other recommended malaria strategies. While cost-effectiveness data were limited, the GDG judged
that cost-effectiveness probably favoured MDA to reduce P. vivax transmission but would depend on the time period
over which it was measured and whether elimination was achieved. The GDG judged that the acceptability of the
intervention was likely to vary depending on the stakeholder group, the population’s previous experience with MDA and
whether radical cure with an 8-aminoquinoline was included. The feasibility of implementing the intervention was
judged to vary depending on the size of the population to be covered and whether radical cure, with the need for G6PD
deficiency testing, an effective pharmacovigilance system and emergency access to blood transfusion services, was
139 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The GDG concluded that MDA could be a useful intervention if it reduced P. vivax transmission quickly to enable the
initiation of intensive surveillance activities. The GDG therefore proposed a conditional recommendation for the use of
MDA for P. vivax.
Research Needs
• Further evidence is needed on the impact (incidence or prevalence of malaria infection at the community level) and
potential harms/ unintended consequences of MDA for P. vivax.
• Evidence is needed on the acceptability, feasibility, impact (incidence or prevalence of malaria infection at the
community level) and potential harms/unintended consequences (death, hospital admission, severe anaemia or any
severe adverse event) of safe provision (including testing for G6PD deficiency and, additionally, an effective
pharmacovigilance system and emergency access to blood transfusion services) of an 8-aminoquinoline as part of
MDA for radical cure of P. vivax.
• Determine the optimal timing and number of MDA rounds to maximize the impact (incidence or prevalence of
malaria infection at the community level) of MDA on P. vivax.
• Determine the minimum effective coverage of MDA in the population to maximize the impact (incidence or
prevalence of malaria infection at the community level) of MDA on P. vivax.
• Determine whether the degree of geographical isolation of communities or mobility of the population modifies the
impact (incidence or prevalence of malaria infection at the community level) of MDA on P. vivax.
Mass treatment with an 8-aminoquinoline medicine alone to reduce the transmission of P. vivax is not recommended.
• Without testing for G6PD deficiency, the GDG noted the potential for severe harm from the use of a therapeutic dose of
an 8-aminoquinoline for radical cure of P. vivax hypnozoites. However, conducting G6PD testing for a large population
would significantly add to the complexity and cost of the intervention.
• The GDG noted that there may be highly exceptional circumstances under which mass relapse prevention (MRP) may be
appropriate, such as during a small focal outbreak of P. vivax in a temperate area. However, under such circumstances the
GDG considered that an MDA programme providing a schizonticide in addition to an 8-aminoquinoline would likely be a
better strategy.
Evidence To Decision
• The evidence is very uncertain about the effect of MRP on the incidence of P. vivax infection. (RD: -102 per
1000 p-y; 95% CI: -103 to -102 per 1000 p-y; two NRSs; very low-certainty evidence).
140 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• The evidence is very uncertain about the effect of MRP on the prevalence of P. vivax infection (RD: -3 per 1000
persons; 95% CI: -4 to -2 per 1000 persons; one NRS; very low-certainty evidence).
• The evidence is very uncertain about the effect of MRP on the incidence of P. vivax infection (RD: -11 per 1000
p-y; 95% CI: -11 to -10 per 1000 p-y; two NRSs; very low-certainty evidence).
Adverse events
• The evidence is very uncertain about the effect of MRP on adverse events (one NRS; very low-certainty
evidence).
The GDG judged that there may be important uncertainty or variability in preferences or values that could not be
determined due to the lack of studies.
Resources
No studies were identified on the costs of implementing MRP.
Equity
No studies were identified addressing the issue of whether MRP increased or decreased health equity.
The GDG judged that equity might be reduced by MRP, given that the undesirable effects were likely to be focalized
in a healthy subgroup of the population with G6PD deficiency.
Acceptability
No studies were identified on the acceptability of MRP.
The GDG was unable to judge whether or not the intervention was acceptable.
Feasibility
The systematic review identified one study on the feasibility of implementing MRP, which provided information on
the size and composition of implementation teams and how adverse events were identified and managed (Shah et al
141 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
unpublished evidence).
The GDG judged that population screening for G6PD deficiency, along with an effective pharmacovigilance system
and emergency access to blood transfusion services, would be needed to implement MRP safely, which would
significantly increase the complexity and cost of the intervention.
Justification
The GDG was disappointed in the very low quality of evidence to judge the impact of MRP on P. vivax transmission. The
GDG judged that the balance of effects probably favoured no MRP while the feasibility of implementing an MRP
programme was very low given the complexity of safely administering radical cure for P. vivax hypnozoites, which would
entail a high cost. Additionally, the GDG was concerned that the MRP strategy does not include an antimalarial medicine
that targets blood-stage parasites (i.e. schizonticide), given evidence for improved efficacy of primaquine against relapses
when co-administered with a schizonticide. The GDG concluded that there should be a conditional recommendation
against implementation of the strategy but considered that there may be highly exceptional circumstances, such as a
small focal outbreak of P. vivax in a temperate area, under which an MRP intervention might be appropriate.
Research Needs
The GDG suggested that the strategy could be reconsidered if a new drug to treat hypnozoites was developed that could
be administered without the need for G6PD testing.
4.3 Vaccine
The use of vaccines for the prevention of malaria partnership, with origins dating back to 1983. Although there are
a handful of P. falciparum malaria vaccine candidates in the
Immunization is a success story for global health and clinical stages of evaluation, RTS,S/AS01 is the first vaccine to
development, saving millions of lives every year. Between 2010 have completed Phase 3 evaluations [143] and the first to be
and 2018, 23 million deaths were averted with the measles provided to children through routine immunization services as
vaccine alone. The number of infants vaccinated annually – more part of phased pilot introductions. In 2015, RTS,S/AS01 received
than 116 million, or 86% of all infants born – has reached the a positive scientific opinion from the European Medicines
highest level ever reported. More than 20 life-threatening Agency [144] and in 2019, it received national regulatory
diseases can now be prevented through immunization. Since authorization for use in the pilot areas of Ghana, Kenya and
2010, 116 countries have introduced vaccines that they did not Malawi for the Malaria Vaccine Implementation Programme. A
previously use, including those against major killers such as separate trial of RTS,S/AS01 took advantage of the vaccine’s
pneumococcal pneumonia, diarrhoea, cervical cancer, typhoid, high initial efficacy by administering a primary series of three
cholera and meningitis [142]. doses at monthly intervals and subsequent annual single doses
just prior to the intense, 4–5 month-long high transmission
A vaccine has the potential to increase the proportion of
season. The vaccine was non-inferior to seasonal malaria
children with access to one or more approaches to malaria
chemoprevention (SMC); the combination of the vaccine and
prevention tools (e.g. ITNs). Introduction of the RTS,S/AS01
SMC was significantly better than either SMC alone or RTS,S/
vaccine in the Malaria Vaccine Implementation Programme
AS01 alone [145].
extended the reach of malaria prevention tools; across the three
pilot countries more than two thirds of children who reportedly Two vaccine candidates are approaching late-stage clinical
did not sleep under an ITN received at least the first dose of evaluation: the R21/MatrixM vaccine candidate targeting PfCSP
RTS,S/AS01. Overall, vaccine introduction increased to over protein [146] and the attenuated whole sporozoite vaccine
90% the proportion of children in each of the three countries PfSPZ [147]. Additional candidates targeting other malaria life-
with access to one or more malaria prevention tool (ITN or cycle stages include the Rh5 blood-stage vaccine
RTS,S/AS01). Vaccine uptake was equitable by sex and candidate [148] and Pfs25 and Pfs230 vaccine candidates
socioeconomic status and had no negative effects on the uptake targeting sexual-stage antigens to prevent human-to-mosquito
of other childhood vaccinations, ITN use, or health-seeking transmission (NCT02942277). New technologies, such as DNA-
behaviour for febrile illness (unpublished evidence). and mRNA-based vaccines [149], the ongoing development of
adjuvants [150], and delivery platforms such as virus-like
Malaria vaccine pipeline
particles (VLPs; the delivery platform used for RTS,S/AS01) and
The RTS,S/AS01 vaccine is the first and currently the only vesicle-based technologies are being explored for use in malaria
malaria vaccine to be recommended for use by WHO. RTS,S/ vaccines. WHO has developed guidelines on the quality, safety,
AS01 is the result of decades of public–private scientific and efficacy of the recombinant malaria vaccines targeting pre-
142 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
erythrocytic and blood stages of P. falciparum [151] and a set of funding support, among other factors. Decision making on
preferred product characteristics (PPCs). The PPCs include whether to adopt and implement the malaria vaccine should be
attributes ranging from safety and efficacy to route of in close collaboration between the NMP and the EPI and other
administration, product stability and storage, in order to help relevant ministry of health departments. In pilot countries, the
support the ongoing development of new malaria vaccines. NMP actively participated in the vaccine introduction and
These PPCs [152] are currently being updated to reflect recent implementation activities in order to ensure that malaria control
advances in malaria vaccine research and development. perspectives were incorporated and to maximize opportunities
for integration. Malaria vaccine technical working groups were
National programmes for immunization and malaria established with joint participation from the EPI and NMP to
provide technical guidance on decision-making and a forum for
The RTS,S/AS01 malaria vaccine should be provided as part of a
alignment. The EPI leads the logistics of vaccine roll-out and
comprehensive malaria control strategy. All malaria control
delivery to relevant health facilities. The EPI manages the
interventions provide partial protection and the highest impact is
planning and activities required for vaccine introduction and
achieved when multiple interventions are used concomitantly.
programme implementation, such as vaccine and supplies
Appropriate mixes of interventions should be identified for
procurement; advocacy; communications and social mobilization;
different subnational strata. These are defined by national
training and supervision of health personnel; logistics and cold
malaria programmes (NMPs) on the basis of the local malaria
chain for vaccine storage; service delivery; and monitoring and
epidemiology (e.g. transmission intensity, age pattern of severe
evaluation. Both fixed sites for vaccination at health care
disease, vector species, insecticide resistance patterns) and
facilities and opportunities for mobile vaccination delivery or
contextual factors (e.g. structure and function of the formal
outreach services should be considered. To increase uptake,
health system).
periodic mass vaccination campaigns or periodic intensified
Where applicable, the malaria vaccine should be integrated into routine immunization activities can be deployed. Monitoring of
relevant immunization guidelines and malaria control strategies, coverage levels occurs through routine health facility data; the
including national strategic plans to define the package of malaria vaccine can be integrated into the District Health
interventions needed to optimize malaria control and elimination Information Software 2 (DHIS2) platform alongside NMP and EPI
in a country. WHO is developing operational guidance on indicators.
principles for the subnational tailoring of malaria interventions.
Please refer to the WHO malaria vaccine position paper for
Country considerations and planning for malaria vaccine more information on the malaria vaccine [153].
introduction should rely on data-driven decision-making in
Please refer to WHO Immunization, Vaccines and Biologicals for
which NMP and Expanded Programme on Immunization (EPI)
more resources and published guidance, including the
staff consider parasite prevalence, disease burden, existing
forthcoming “Guide for introducing a malaria vaccine."
malaria interventions, vaccine delivery, the logistics, strength and
support of the immunization programme, and the availability of
The RTS,S/AS01 malaria vaccine should be used for the prevention of P. falciparum malaria in children living in regions with
moderate to high transmission as defined by WHO.
• The RTS,S/AS01 malaria vaccine should be provided in a four-dose schedule in children from 5 months of age.
• Countries may consider providing the RTS,S/AS01 vaccine seasonally, with a five-dose strategy, in areas with highly seasonal
malaria or with perennial malaria transmission with seasonal peaks.
• Countries that choose to introduce the vaccine in a five-dose seasonal strategy are encouraged to document their experiences,
including adverse events following immunization.
• RTS,S/AS01 malaria vaccine should be provided as part of a comprehensive malaria control strategy.
Practical Info
Vaccine characteristics, content, dosage, administration and storage
RTS,S/AS01 is a pre-erythrocytic recombinant protein vaccine, based on the RTS,S recombinant antigen. It comprises the hybrid
polypeptide RTS, in which regions of the P. falciparum circumsporozoite protein known to induce humoral (R region) and
cellular (T region) immune responses are covalently bound to the hepatitis B virus surface antigen (S). The vaccine is currently
produced as a two-dose RTS,S powder to be reconstituted with a two-dose AS01 adjuvant system suspension. After
reconstitution, the total volume is 1ml (two doses of 0.5 ml). No preservative is included in either the RTS,S formulation or the
AS01 adjuvant system. The vials should therefore be discarded at the end of the vaccination session, or within six hours after
143 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
opening, whichever comes first. The reconstituted 0.5ml vaccine should be administered by injection into the deltoid muscle in
children aged 5 months or older. The shelf life of the RTS,S/AS01 vaccine is three years. A vaccine vial monitor is on the AS01
vial [144].
Schedule
WHO recommends that the first dose of vaccine be administered from 5 months of age. There should be a minimum interval of
four weeks between doses. The vaccine should be administered in a three-dose primary schedule, with a fourth dose provided
12–18 months after the third dose to prolong the duration of protection. However, there can be flexibility in the schedule to
optimize delivery, for example, to align the fourth dose with other vaccines given in the second year of life. Children who begin
their vaccination series should complete the four-dose schedule [153].
Optional schedule for settings with highly seasonal malaria or perennial malaria with seasonal peaks
Countries may consider providing the RTS,S/AS01 vaccine seasonally, with a five-dose strategy in areas with highly seasonal
malaria or with perennial malaria transmission with seasonal peaks. This strategy seeks to maximize vaccine impact by ensuring
that the period of highest vaccine efficacy (just after vaccination) coincides with the period of highest malaria transmission. The
primary series of three doses should be provided at monthly intervals, with additional doses provided annually prior to the peak
transmission season. Countries that choose seasonal deployment of the RTS,S/AS01 vaccine are strongly encouraged to
document their experiences, including the vaccine’s effectiveness, feasibility and occurrence of any adverse events following
immunization—as additional input for future updates to the guidance. WHO also encourages international and national funders
to support relevant learning opportunities [153].
Co-administration
RTS,S/AS01 given in conjunction with routine childhood vaccines has been evaluated in several trials [158][159]. Non-inferiority
criteria were met for all vaccines given with RTS,S/AS01, in comparison with the same vaccines given without RTS,S/AS01.
RTS,S/AS01 can be given concomitantly with any of the following monovalent or combination vaccines: diphtheria, tetanus,
whole cell pertussis, acellular pertussis, hepatitis B, Haemophilus influenzae type b, oral poliovirus, measles, rubella, yellow fever,
rotavirus and pneumococcal conjugate vaccines [144]. No co-administration studies have been conducted with RTS,S/AS01 and
meningococcus A, typhoid conjugate, cholera, Japanese encephalitis, Tick-borne encephalitis, rabies, mumps, influenza or
varicella vaccines [153].
Current WHO guidance defines moderate or high transmission settings as those with an annual incidence greater than about
250 cases per 1000 population or a prevalence of P. falciparum infection in children aged 2—10 years (PfPR2-10) of
approximately 10% or more. These are indicative values and should not be used as strict thresholds.
Vaccine safety
The RTS,S/AS01 vaccine is safe and well tolerated. There is a small risk of febrile seizures within seven days (mainly within 2—3
days) of vaccination. As with any vaccine introduction, proper planning and training of staff to conduct appropriate
pharmacovigilance should take place beforehand.
The only contraindication to use of RTS,S/AS01 vaccine is severe hypersensitivity to any of the vaccine components [144].
The vaccine should be provided to infants and young children aged 5—17 months of age who relocate to an area of moderate to
high transmission, including during emergency situations.
The vaccine has been developed for use in young children living in malaria-endemic settings, and has not undergone full clinical
testing in adults, nor is it recommended for adults. The vaccine is not indicated for travellers, who should use chemoprophylaxis
and vector control methods to prevent malaria when traveling to endemic settings.
Surveillance
As for all new vaccines, the effectiveness and safety of the RTS,S/AS01 vaccine should be monitored post-introduction.
Countries that choose to introduce the vaccine in a five-dose seasonal strategy are encouraged to document their experience,
144 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Research priorities
The WHO-coordinated Malaria Vaccine Implementation Programme will continue through 2023, with continued monitoring of
data on safety, impact, coverage achieved and the added benefit of the fourth dose. In areas with highly seasonal malaria or with
perennial malaria transmission with seasonal peaks, operational research is needed specifically related to the seasonal delivery
of vaccine doses, including annual preseason dosing after a primary series given through the routine health clinics. Further
evaluation will be required to determine how best to deliver the combination of SMC and seasonal malaria vaccination in areas.
Data should be collected on safety, immunogenicity, and effectiveness of annual doses beyond the fifth dose.
Evidence To Decision
• There were significant reductions in clinical malaria (51%); and severe malaria (45%), demonstrated after 12 months'
follow-up of the first three doses in the Phase 3 trial [143].
• There were significant reductions in clinical malaria (39%); severe malaria (29%); severe malaria anaemia (61%);
malaria-related hospitalization (37%); and the need for blood transfusions (30%), demonstrated over 46 months'
follow-up after the first three doses in the Phase 3 trial in children who received a fourth dose 18 months after the
third dose [143].
• There were 1774 clinical malaria cases averted per 1000 children vaccinated with four RTS,S/AS01 doses over 46
months' follow-up in the Phase 3 trial [143].
• There were significant reductions in clinical malaria (24%) demonstrated after 7 years’ follow-up after vaccination
among a subset of children in the Phase 3 trial living in areas of moderate to high transmission; they did not have an
excess risk of clinical or severe malaria [154].
• There were significant reductions in hospitalization with severe malaria (29%) and hospitalization with malarial
parasitemia or antigenemia (21%), demonstrated among children who were age-eligible for three doses of vaccine
delivered through routine systems by the ministries of health in parts of Ghana, Kenya, and Malawi (Milligan et al.
unpublished evidence).
• Median estimates ranged from 200 to 700 deaths averted per 100 000 children vaccinated with a 4-dose schedule in
areas of moderate to high transmission [156].
• There were substantially greater reductions in uncomplicated malaria (63%), hospital admissions with severe malaria
(70%), and death from malaria (73%) among children who received the combination of RTS,S/AS01 seasonal
vaccination and SMC when compared to SMC alone. Seasonal vaccination with RTS,S/AS01 before the peak
transmission season was non-inferior to SMC in preventing clinical malaria [145].
• There is a small risk of febrile seizures within seven days (mainly within 2–3 days) of vaccination [144].
• As with any vaccine introduction, proper planning and training of staff to conduct appropriate pharmacovigilance
should take place beforehand [153].
145 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• As for all new vaccines, the effectiveness and safety of the RTS,S/AS01 vaccine should be monitored post-
introduction [153].
More information can be found in the Full evidence report on the RTS,S/AS01 malaria vaccine background
paper (unpublished evidence) sections 5.3.2 and 6.1 (MVIP safety, methods and results); sections 5.3.3 and 6.2 (MVIP impact,
methods and results); sections 7.2 (Phase 3 results); section 8 (Additional data since Phase 3 completion); section 9
(Modelled public health impact and cost-effectiveness estimates).
Further details on “Benefits and harms” are also included in the SAGE/MPAG Evidence-to-Recommendations framework
(unpublished evidence).
The overall rating of the evidence on RTS,S/AS01 malaria vaccine is considered to be HIGH. The certainty of evidence
ranged from very low to high.
Critical outcomes related to effectiveness of RTS,S/AS01 were mostly rated HIGH in the large-scale Phase 3 clinical trial
and MODERATE (due to large confidence intervals [CIs]) in the pilot implementation study.
Overall the certainty of evidence for the safety outcomes was rated MODERATE. Three safety signals, thought to be chance
findings, were identified in the Phase 3 trial; these rare, unexplained events were graded with LOW and VERY LOW
certainty of evidence:
• An excess of meningitis and cerebral malaria (in the context of overall reduction in severe malaria).
• An excess of deaths among girls who had received RTS,S/AS01 (shown in a post hoc analysis compared to boys).
The Malaria Vaccine Pilot Evaluations were designed to answer the outstanding questions related to safety. Evidence on the
safety outcomes of meningitis, cerebral malaria, and gender-specific mortality is now graded MODERATE certainty
reflecting the wide CIs related to relatively rare events. Multiple WHO advisory committees reviewed the data from the
pilot implementation study and concluded that there was no evidence that the Phase 3 safety signals were causally related
to RTS,S/AS01. Additionally these safety signals were not seen in the Phase 2 trials [157] or subsequent Phase 3
trials [154][145].
More information can be found in the Full evidence report on the RTS,S/AS01 malaria vaccine background
paper (unpublished evidence) Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) Evidence
summary table by the Cochrane Response and the SAGE/MPAG Evidence-to-Recommendations framework (unpublished
evidence).
Malaria remains a primary cause of childhood illness and mortality in much of sub-Saharan Africa.
Preferences and values of the target population have been assessed in several ways:
• Qualitative interviews with caregivers and health providers revealed the perceived value of the vaccine in reducing the
severity and frequency of malaria. Positive attitudes and trust among caregivers increased substantially over time,
driven mainly by their perception of the vaccine’s health benefits in their own children and the broader community.
• Malaria vaccine coverage from cross-sectional household surveys and from routine facility-based administrative data
indicated that the vaccine was acceptable to the target population with relatively rapid scale–up for a new vaccine with
a unique schedule and dropout between doses comparable to other vaccines (see “Feasibility” section).
• Coverage of other interventions from household survey and routine administrative data in areas where the vaccine has
been introduced indicated that the vaccine had no negative effects on the uptake of other childhood vaccinations, on
ITN use, or health–seeking behaviour for febrile illness.
146 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Note: Midline surveys and the second round of the qualitative study were conducted between the provision of the third
dose and the provision of the fourth dose and thus did not capture data on the uptake/coverage/acceptability of the fourth
dose.
More information can be found in the Full evidence report on the RTS,S/AS01 malaria vaccine background
paper (unpublished evidence) sections 5.3.4.2 and 6.3.1 (routine data, methods and results); sections 5.3.4.3 and 6.3.2
(household survey methods and results), and sections 5.3.4.5 and 6.3.4 (qualitative health utilization study methods and
results, unpublished evidence).
Further details on “Values and Preferences" are also included in the SAGE/MPAG Evidence-to-Recommendations
framework (unpublished evidence).
Resources
The resources required are likely to be comparable to other new vaccine introductions.
Mathematical models examined the addition of the vaccine to existing malaria control interventions and treatment [156].
• At an assumed vaccine price of US$5 per dose and PfPR2-10 of 10-65%, the models predicted a median ICER compared
with no vaccine of $25 (95%CI 16–222) per clinical case averted and $87 (95%CI 48–224) per DALY averted for the
four-dose schedule.
• Public health impact and cost-effectiveness tended to be greater at higher levels of transmission.
• Overall, the model estimated that ICERs were only marginally lower for the seasonal vaccination strategies (i.e. more
cost-effective) despite the higher number of overall doses delivered.
• Caution is required in the comparison of cost-effectiveness estimates for different interventions evaluated with
different methods, outcome measures, time intervals and context (e.g. with different concurrent health interventions
and standards of care). Nevertheless, the predictions of RTS,S/AS01 cost per DALY averted are broadly positive and
comparable with other new vaccines, based on mathematical models, and other malaria interventions.
Table 1 is based on the evidence reviewed by the RTS,S/AS01 SAGE/MPAG Working Group on the incremental cost
estimates of introducing and delivering the RTS,S/AS01 malaria vaccine within routine immunization programmes in
subnational areas of the malaria vaccine pilot countries: Ghana, Kenya and Malawi. The line items account for the activities
conducted in the first 1–2 years of vaccine implementation (through December 2020).
More information on the evidence can be found in the Full evidence review on the RTS,S/AS01 malaria vaccine background
paper (unpublished evidence) sections 5.3.4.6 and 6.3.5 (cost of introduction and delivery study methods and results) and
section 9 (unpublished evidence). Further details on “Resource use” and “Cost-effectiveness” (unpublished evidence) are also
included in the SAGE/MPAG Evidence-to-recommendations framework (unpublished evidence).
Table 1: Line items from RTS,S/AS01 cost of delivery and vaccine introduction study
Staff • EPI and NMP, among other ministry of health staff including vaccinators at
health facilities
• District malaria and health information management coordinators for DHIS2
data analysis
147 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
county levels)
Equity
Vaccine uptake was equitable by sex and socioeconomic status.
• Vaccine uptake had no negative effect on the uptake of other childhood vaccinations, ITN use or health-seeking
behaviour for febrile illness.
• Introduction of RTS,S/AS01 extended the reach of malaria prevention tools; across the three pilot countries, more than
two thirds of the children who reportedly did not sleep under an ITN received at least their first dose of the malaria
vaccine.
• Overall, vaccine introduction increased to over 90% the proportion of children in each of the three pilot countries with
access to one or more malaria prevention tools (ITN or RTS,S/AS01).
148 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
More information on the evidence can be found in the Full evidence report on the RTS,S/AS01 malaria vaccine background
paper (unpublished evidence) section 10 (Equity considerations). Further details on “Equity” are also included in the SAGE/
MPAG Evidence-to-Recommendations framework (unpublished evidence).
Acceptability
RTS,S/AS01 malaria vaccine considered acceptable to the following groups:
• Target population (including eligible children and their caregivers): This is based on administrative data and household
surveys that indicate good uptake and coverage, and modest drop-out rates. Continued increases in uptake suggest
that the additional visits needed to receive the vaccine are acceptable to the target populations. Qualitative data
indicate high acceptance and desirability of the vaccine.
• Key stakeholders (including ministries of health and immunization programme managers): This is based on post-
introduction evaluations, the good uptake and coverage of the malaria vaccine, and qualitative study interviews with
health providers. Chief concerns from health providers were around the operational challenges faced in introducing and
delivering RTS,S/AS01 (i.e. increased workload, training, eligibility).
Household surveys found no impact on the use of ITNs in intervention areas following the introduction of RTS,S/AS01,
indicating that both interventions are acceptable and the vaccine has not displaced ITN use. Overall health-seeking
behaviour for febrile illness was also similar between the implementing and comparison groups as well as between the
baseline and midline surveys.
More information on the evidence can be found in the Full evidence report on the RTS,S/AS01 malaria vaccine background
paper (unpublished evidence) sections 5.3.4.2 and 6.3.1 (routine data, methods and results); sections 5.3.4.3 and 6.3.2
(household survey methods and results), sections 5.3.4.4 and 6.3.3 (post-introduction evaluation methods and results) and
sections 5.3.4.5 and 6.3.4 (qualitative health utilization study methods and results). Further details on “Acceptability”
(unpublished evidence) are also included in the SAGE/MPAG Evidence-to-Recommendations framework (unpublished
evidence).
Feasibility
Vaccine introduction is feasible with good and equitable coverage of RTS,S/AS01 seen through routine immunization
systems even in the context of the COVID-19 pandemic.
Administrative data from the start of pilot programme vaccinations in 2019 and April 2021 (24 months in Ghana and
Malawi, and 18 months in Kenya) showed that:
• More than 1.7 million RTS,S/AS01 vaccine doses were administered across the three pilot countries and more than 650
000 children received their first dose.
• All three countries reached more than 70% of their target populations with the first RTS,S/AS01 dose and at least 62%
with the third RTS,S/AS01 dose (unpublished evidence).
More information on the evidence can be found in the Full evidence report on the RTS,S/AS01 malaria vaccine background
paper (unpublished evidence) sections 5.3.4.2 and 6.3.1 (routine data, methods and results). Further details on “Feasibility”
are also included in the SAGE/MPAG Evidence-to-Recommendations framework (unpublished evidence).
Justification
A Framework for WHO recommendation on RTS,S/AS01 malaria vaccine (unpublished evidence), endorsed by SAGE and MPAG
in 2019, provided guidance on how data from the MVIP should inform WHO recommendations, with the aim of ensuring that a
recommendation could be made as soon as the risk–benefit of the vaccine was established with the necessary level of
confidence, such that the vaccine would not be unnecessarily withheld from countries in need if it was found to be safe and
beneficial.
149 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The Framework stated that a WHO recommendation could be made if and when concerns regarding the safety signals were
satisfactorily resolved, and evidence on severe malaria or mortality was assessed as consistent with a beneficial impact of the
vaccine.
The Framework clarified that a recommendation should not be predicated on attaining high coverage, including high coverage
with the fourth vaccine dose, based on: (1) data from the Phase 3 long-term follow up study showing that children living in areas
of perennial moderate to high malaria transmission benefit from three or four doses of the vaccine; and (2) experience that it
usually takes time for new vaccines to attain high coverage, particularly when administered in the second year of life.
The RTS,S/AS01 vaccine is considered safe and well tolerated. There is a small risk of febrile seizures within seven days (mainly
within 2–3 days) of vaccination. As with any vaccine introduction, proper planning and training of staff to conduct appropriate
pharmacovigilance should take place beforehand.
RTS,S/AS01 has a demonstrated ability to quickly achieve high coverage and high impact when delivered through routine
immunization systems, with a 30% reduction in severe malaria observed after the vaccine was introduced in areas where ITNs
are widely used and there is good access to diagnosis and treatment. Modelling shows that the vaccine is cost–effective in areas
of moderate to high malaria transmission.
RTS,S/AS01 increases access to malaria prevention with no negative effect on the uptake other childhood vaccinations, ITN use,
or health–seeking behaviour for febrile illness.
5. CASE MANAGEMENT
Background 1. Early diagnosis and prompt, effective treatment of malaria
Malaria case management, consisting of early diagnosis and Uncomplicated falciparum malaria can progress rapidly to severe
prompt effective treatment, remains a vital component of malaria forms of the disease, especially in people with no or low immunity,
control and elimination strategies. The WHO Guidelines for the and severe falciparum malaria is almost always fatal without
treatment of malaria were first developed in 2006 and have been treatment. Therefore, programmes should ensure access to early
revised periodically, with the most recent edition published in diagnosis and prompt, effective treatment within 24–48 h of the
2015. WHO guidelines contain recommendations on clinical onset of malaria symptoms.
practice or public health policy intended to guide end-users as to
the individual or collective actions that can or should be taken in 2. Rational use of antimalarial agents
specific situations to achieve the best possible health outcomes. To reduce the spread of drug resistance, limit unnecessary use of
Such recommendations are also designed to help the user to select antimalarial drugs and better identify other febrile illnesses in the
and prioritize interventions from a range of potential alternatives. context of changing malaria epidemiology, antimalarial medicines
The third edition of the WHO Guidelines for the treatment of should be administered only to patients who truly have malaria.
malaria consolidated here contains updated recommendations Adherence to a full treatment course must be promoted. Universal
based on new evidence particularly related to dosing in children, access to parasitological diagnosis of malaria is now possible with
and also includes recommendations on the use of drugs to prevent the use of quality-assured rapid diagnostic tests (RDTs), which are
malaria in groups at high risk. also appropriate for use in primary health care and community
settings.
Since publication of the first edition of the Guidelines for the
treatment of malaria in 2006 and the second edition in 2010, all 3. Combination therapy
countries in which P. falciparum malaria is endemic have Preventing or delaying resistance is essential for the success of
progressively updated their treatment policy from use of both national and global strategies for control and eventual
monotherapy with drugs such as chloroquine, amodiaquine and elimination of malaria. To help protect current and future
sulfadoxine–pyrimethamine (SP) to the currently recommended antimalarial medicines, all episodes of malaria should be treated
artemisinin-based combination therapies (ACT). The ACTs are with at least two effective antimalarial medicines with different
generally highly effective and well tolerated. This has contributed mechanisms of action (combination therapy).
substantially to reductions in global morbidity and mortality from
malaria. Unfortunately, resistance to artemisinins has arisen 4. Appropriate weight-based dosing
recently in P. falciparum in South-East Asia, which threatens these To prolong their useful therapeutic life and ensure that all patients
gains. have an equal chance of being cured, the quality of antimalarial
drugs must be ensured, and antimalarial drugs must be given at
Core principles optimal dosages. Treatment should maximize the likelihood of
The following core principles were used by the Guidelines rapid clinical and parasitological cure and minimize transmission
Development Group that drew up the Guidelines for the from the treated infection. To achieve this, dosage regimens
Treatment of Malaria. should be based on the patient’s weight and should provide
effective concentrations of antimalarial drugs for a sufficient time
150 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
to eliminate the infection in all target populations. Please refer to Malaria case management: operations manual [162].
151 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
must be assessed. A skilled microscopist can detect asexual Current tests are based on the detection of histidine-rich protein
parasites at a density of < 10 per µL of blood, but under typical 2 (HRP2), which is specific for P. falciparum, pan-specific or
field conditions, the limit of sensitivity is approximately 100 species-specific Plasmodium lactate dehydrogenase (pLDH) or
parasites per µL [165]. This limit of detection approximates the pan-specific aldolase. The different characteristics of these
lower end of the pyrogenic density range. Thus, microscopy antigens may affect their suitability for use in different
provides good specificity for diagnosing malaria as the cause of a situations, and these should be taken into account in
presenting febrile illness. More sensitive methods allow programmes for RDT implementation. The tests have many
detection of an increasing proportion of cases of incidental potential advantages, including:
parasitaemia in endemic areas, thus reducing the specificity of a
positive test. Light microscopy has other important advantages: • rapid provision of results and extension of diagnostic
services to the lowest-level health facilities and
• low direct costs, if laboratory infrastructure to maintain the communities;
service is available; • fewer requirements for training and skilled personnel (for
• high sensitivity, if the performance of microscopy is high; instance, a general health worker can be trained in 1 day);
• differentiation of Plasmodia species; and
• determination of parasite densities – notably identification • reinforcement of patient confidence in the diagnosis and in
of hyperparasitaemia; the health service in general.
• detection of gametocytaemia;
• allows monitoring of responses to therapy and
They also have potential disadvantages, including:
• can be used to diagnose many other conditions.
• inability, in the case of PfHRP2-based RDTs, to distinguish
Good performance of microscopy can be difficult to maintain, new infections from recently and effectively treated
because of the requirements for adequate training and infections, due to the persistence of PfHRP2 in the blood
supervision of laboratory staff to ensure competence in malaria for 1–5 weeks after effective treatment;
diagnosis, electricity, good quality slides and stains, provision • the presence in countries in the Amazon region of variable
and maintenance of good microscopes and maintenance of frequencies of HRP2 deletions in P. falciparum parasites,
quality assurance [166] and control of laboratory services. making HRP2-based tests not suitable in this region [173];
• poor sensitivity for detecting P. malariae and P. ovale; and
Numerous attempts have been made to improve malaria • the heterogeneous quality of commercially available
microscopy, but none has proven to be superior to the classical products and the existence of lot-to-lot variation.
method of Giemsa staining and oil-immersion microscopy for
performance in typical health care settings [167].
In a systematic review [174], the sensitivity and specificity of
RDTs in detecting P. falciparum in blood samples from patients in
Rapid diagnostic tests
endemic areas attending ambulatory health facilities with
Rapid diagnostic tests (RDTs) are immuno-chromatographic tests
symptoms suggestive of malaria were compared with the
for detecting parasite-specific antigens in a finger-prick blood
sensitivity and specificity of microscopy or polymerase chain
sample. Some tests allow detection of only one species (P.
reaction. The average sensitivity of PfHRP2-detecting RDTs was
falciparum); others allow detection of one or more of the other
95.0% (95% confidence interval [CI], 93.5–96.2%), and the
species of human malaria parasites (P. vivax, P. malariae and P.
specificity was 95.2% (93.4–99.4%). RDTs for detecting pLDH
ovale) [168][169][170]. They are available commercially in various
from P. falciparum are generally less sensitive and more specific
formats, e.g. dipsticks, cassettes and cards. Cassettes and cards
than those for detecting HRP2, with an average sensitivity (95%
are easier to use in difficult conditions outside health facilities.
CI) of 93.2% (88.0–96.2%) and a specificity of 98.5%
RDTs are relatively simple to perform and to interpret, and they
(96.7–99.4%). Several studies have shown that health workers,
do not require electricity or special equipment [171].
volunteers and private sector providers can, with adequate
training and supervision, use RDTs correctly and provide
Since 2012, WHO has recommended that RDTs should be
accurate malaria diagnoses. The criteria for selecting and
selected in accordance with the following criteria, based on the
procuring RDTs can be found on the WHO website.
results of the assessments of the WHO Malaria RDT Product
Testing programme [172]:
Diagnosis with either microscopy or RDTs is expected to reduce
• For detection of P. falciparum in all transmission settings, the overuse of antimalarial medicines by ensuring that treatment is
panel detection score against P. falciparum samples should given only to patients with confirmed malaria infection, as
be at least 75% at 200 parasites/µL. opposed to treating all patients with fever [175]. Although
• For detection of P. vivax in all transmission settings the providers of care may be willing to perform diagnostic tests, they
panel detection score against P. vivax samples should be at do not, however, always respond appropriately to the results.
least 75% at 200 parasites/µL. This is especially true when they are negative. It is therefore
• The false positive rate should be less than 10%. important to ensure the accuracy of parasite- based diagnosis
• The invalid rate should be less than 5%. and also to demonstrate this to users and to provide them with
the resources to manage both positive and negative results
adequately [164].
152 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
All cases of suspected malaria should have a parasitological test (microscopy or RDT) to confirm the diagnosis.
Justification
Prompt, accurate diagnosis of malaria is part of effective disease management. All patients with suspected malaria should be
treated on the basis of a confirmed diagnosis by microscopy examination or RDT testing of a blood sample. Correct diagnosis in
malaria-endemic areas is particularly important for the most vulnerable population groups, such as young children and non-
immune populations, in whom falciparum malaria can be rapidly fatal. High specificity will reduce unnecessary treatment with
antimalarial drugs and improve the diagnosis of other febrile illnesses in all settings.
WHO strongly advocates a policy of “test, treat and track” to improve the quality of care and surveillance.
153 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
with a highly effective ACT is required whether or not the fevers, but, like aspirin and other non-steroidal anti-
patient is considered to be semi-immune. inflammatory drugs, it is no longer recommended because of the
risks of gastrointestinal bleeding, renal impairment and Reye’s
Another potentially dangerous practice is to give only the first syndrome.
dose of a treatment course to patients with suspected but
unconfirmed malaria, with the intention of giving the full Use of anti-emetics
treatment if the diagnosis is confirmed. This practice is unsafe, Vomiting is common in acute malaria and may be severe.
could engender resistance, and is not recommended. Parenteral antimalarial treatment may therefore be required
until oral administration is tolerated. Then a full 3-day course
Additional considerations for clinical management of ACT should be given. Anti-emetics are potentially sedative
and may have neuropsychiatric adverse effects, which could
Can the patient take oral medication?
mask or confound the diagnosis of severe malaria. They should
Some patients cannot tolerate oral treatment and will require
therefore be used with caution.
parenteral or rectal administration for 1–2 days, until they can
swallow and retain oral medication reliably. Although such
Management of seizures
patients do not show other signs of severity, they should
Generalized seizures are more common in children with P.
receive the same initial antimalarial treatments recommended
falciparum malaria than in those with malaria due to other
for severe malaria. Initial rectal or parenteral treatment must
species. This suggests an overlap between the cerebral
always be followed by a full 3-day course of ACT.
pathology resulting from falciparum malaria and febrile
convulsions. As seizures may be a prodrome of cerebral
Use of antipyretics
malaria, patients who have more than two seizures within a 24
In young children, high fevers are often associated with
h period should be treated as for severe malaria. If the seizures
vomiting, regurgitation of medication and seizures. They are
continue, the airways should be maintained and
thus treated with antipyretics and, if necessary, fanning and
anticonvulsants given (parenteral or rectal benzodiazepines or
tepid sponging. Antipyretics should be used if the core
intramuscular paraldehyde). When the seizure has stopped, the
temperature is > 38.5 ºC. Paracetamol (acetaminophen) at a
child should be treated as indicated in section 7.10.5, if his or
dose of 15 mg/kg bw every 4 h is widely used; it is safe and
her core temperature is > 38.5 ºC. There is no evidence that
well tolerated and can be given orally or as a suppository.
prophylactic anticonvulsants are beneficial in otherwise
Ibuprofen (5 mg/kg bw) has been used successfully as an
uncomplicated malaria, and they are not recommended.
alternative in the treatment of malaria and other childhood
Children and adults with uncomplicated P. falciparum malaria should be treated with one of the following ACTs*:
• artemether-lumefantrine (AL)
• artesunate-amodiaquine (AS+AQ)
• artesunate-mefloquine (ASMQ)
• dihydroartemisinin-piperaquine (DHAP)
• artesunate + sulfadoxine-pyrimethamine (AS+SP)
• artesunate-pyronaridine (ASPY) (2022)
*Artesunate + sulfadoxine-pyrimethamine and artesunate-pyronaridine are not recommended for use in the first
trimester of pregnancy. For details of treatment using ACTs in the first trimester of pregnancy, see 5.2.1.4.1 below.
Artesunate-pyronaridine is now included in the list of options for the treatment of uncomplicated malaria (2022). See the full
recommendation and supporting evidence below.
Practical Info
The pipeline for new antimalarial drugs is healthier than ever before, and several new compounds are in various stages of
development. Some novel antimalarial agents are already registered in some countries. The decision to recommend
154 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
antimalarial drugs for general use depends on the strength of the evidence for safety and efficacy and the context of use.
In general, when there are no satisfactory alternatives, newly registered drugs may be recommended; however, for
global or unrestricted recommendations, considerably more evidence than that submitted for registration is usually
required, to provide sufficient confidence for their safety, efficacy and relative merits as compared with currently
recommended treatments.
Several new antimalarial drugs or new combinations have been introduced recently. Some are still in the pre-registration
phase and are not discussed here. Arterolane + piperaquine, artemisinin + piperaquine base and artemisinin +
napththoquine are new ACTs, which are registered and used in some countries. In addition, there are several new generic
formulations of existing drugs. None of these yet has a sufficient evidence base for general recommendation (i.e.
unrestricted use).
Arterolane + piperaquine is a combination of a synthetic ozonide and piperaquine phosphate that is registered in India.
There are currently insufficient data to make general recommendations.
Artemisinin + piperaquine base combines two well-established, well-tolerated compounds. It differs from previous
treatments in that the piperaquine is in the base form, the artemisinin dose is relatively low, and the current
recommendation is for only a 2-day regimen. There are insufficient data from clinical trials for a general
recommendation, and there is concern that the artemisinin dose regimen provides insufficient protection against
resistance to the piperaquine component.
Artemisinin + naphthoquine is also a combination of two relatively old compounds that is currently being promoted as a
single-dose regimen, contrary to WHO advice for 3 days of the artemisinin derivative. There are currently insufficient
data from rigorously conducted randomized controlled trials to make general recommendations.
Many ACTs are generics. The bioavailability of generics of currently recommended drugs must be comparable to that of
the established, originally registered product, and the satisfactory pharmaceutical quality of the product must be
maintained.
Please refer to Good procurement practices for artemisinin-based antimalaria medicines [179].
Evidence To Decision
Desirable effects
• Studies have consistently demonstrated that the six WHO-recommended ACTs result in < 5% PCR-adjusted
treatment failures in settings with no resistance to the partner drug (high- quality evidence).
Undesirable effects
• Increased cost.
Desirable effects:
• A PCR-adjusted treatment failure rate of < 5% has been seen consistently in trials of dihydroartemisinin +
piperaquine (high-quality evidence).
• Dihydroartemisinin + piperaquine has a longer half-life than artemether + lumefantrine, and fewer new
infections occur within 9 weeks of treatment with dihydroartemisinin + piperaquine (high-quality evidence).
• Dihydroartemisinin + piperaquine and artesunate + mefloquine have similar half-lives, and a similar frequency of
new infections is seen within 9 weeks of treatment (moderate-quality evidence).
155 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Undesirable effects:
• A few more patients receiving dihydroartemisinin + piperaquine than those given artesunate + mefloquine had
a prolonged QT interval (low-quality evidence)
• A few more patients receiving dihydroartemisinin + piperaquine than those given artesunate + mefloquine or
artemether + lumefantrine had borderline QT prolongation.
Justification
GRADE
In the absence of resistance to the partner drug, the five recommended ACTs have all been shown to achieve a PCR-
adjusted treatment failure rate of 5% in many trials in several settings in both adults and children (high-quality
evidence) [180][181].
Other considerations
The guideline development group decided to recommend a menu of approved combinations, from which countries can
select first- and second-line treatment.
Remarks
Recommendation: Treat adults and children with uncomplicated P. falciparum malaria (including infants, pregnant women
in their second and third trimesters and breastfeeding women) with ACT.
The WHO-approved first-line ACT options are: artemether + lumefantrine, artesunate + amodiaquine, artesunate +
mefloquine, dihydroartemisinin + piperaquine and artesunate + sulfadoxine–pyrimethamine.
These options are recommended for adults and children, including infants, lactating women and pregnant women in their
second and third trimester.
In deciding which ACTs to adopt in national treatment policies, national policy- makers should take into account: the
pattern of resistance to antimalarial drugs in the country, the relative efficacy and safety of the combinations, their cost,
the availability of paediatric formulations and the availability of co-formulated products.
The Guideline Development Group decided to recommend a “menu” of approved combinations from which countries
can select first- and second- line therapies. Modelling studies suggest that having multiple first-line ACTs available for
use may help to prevent or delay the development of resistance.
A systematic review showed that the dosing regimen of dihydroartemisinin + piperaquine currently recommended by the
manufacturers leads to sub-optimal dosing in young children. The group plans to recommend a revised dosing regimen
based on models of pharmacokinetics.
Further studies of the risk for QT interval prolongation have been requested by the European Medicines Agency.
ACT is a combination of a rapidly acting artemisinin derivative with a longer-acting (more slowly eliminated) partner
drug. The artemisinin component rapidly clears parasites from the blood (reducing parasite numbers by a factor of
approximately 10 000 in each 48 h asexual cycle) and is also active against the sexual stages of the gametocytes that
mediate onward transmission to mosquitos. The longer- acting partner drug clears the remaining parasites and provides
protection against development of resistance to the artemisinin derivative. Partner drugs with longer elimination half-
lives also provide a period of post-treatment prophylaxis.
The GDG recommended dihydroartemisinin + piperaquine for use in 2009 but re-evaluated the evidence in 2013
because additional data on its safety had become available. The group noted the small absolute prolongation of the QT
interval with dihydroartemisinin + piperaquine but was satisfied that the increase was of comparable magnitude to that
156 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Artesunate-pyronaridine (ASPY) is recommended as an artemisinin-based combination therapy option for the treatment
of uncomplicated P. falciparum malaria.
• ASPY should be avoided by individuals with known liver disease (clinically apparent liver disease) because ASPY is
associated with liver transaminitis.
• Pharmacovigilance should be strengthened where ASPY is used for the treatment of malaria.
Practical Info
As with the deployment of any new malaria treatment, pharmacovigilance and resistance surveillance systems should be
strengthened.
Evidence To Decision
The GDG judged the overall certainty of the assessed evidence to be low mostly due to imprecision and
indirectness.
157 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
and preferences, but country-level value judgements are still important, as these could be influenced by the
prevalence of antimalarial partner drug resistance and the prevalence of hepatic diseases.
Resources
Research on formal cost analysis, and cost estimates related to scale are required. However, changing first- or
second-line malaria treatment is quite resource-intensive, requiring staff training and patient information and
introducing supply chain and logistical issues. However, introducing ASPY is not expected to be different from other
ACTs already in use, as any additional cost would be minimal based on the actual cost of the medicine.
Equity
The GDG considered that ASPY is likely to enhance equity, especially in areas of emerging resistance to existing
combinations. The addition of ASPY as a treatment option for malaria will probably increase health equity.
Acceptability
Although in some countries there is limited experience of its use, ASPY is probably acceptable given that some
countries already include ASPY in therapeutic efficacy studies. Aside from the additional resource implications with
the introduction of a new antimalarial regimen, oral treatments are generally well accepted. Some issues might arise
when hepatic risk profiles need to be assessed in the target population.
Feasibility
Policy changes are feasible, since some countries have already started to use ASPY. The medicine is available, and
the treatment regimen is similar to that of other approved ACTs. ASPY has also received a positive scientific opinion
from the European Medicines Agency under Article 58 and is thus included in the WHO list of prequalified
antimalarial medicines. However, the feasibility of strengthening pharmacovigilance will be highly variable from
country to country.
Justification
The GDG reached a consensus on a strong recommendation for the intervention, despite the low certainty of evidence
because of:
• the large magnitude of treatment effect, as well as its non-inferiority and comparability to the other currently
recommended ACTs;
• its tolerability and generally mild, reversible adverse events; and
• the probable increased equity from access to an additional treatment option, specifically in the face of increasing
ACT partner drug resistance.
158 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Research Needs
The GDG highlighted the following evidence gaps requiring further research. These relate to:
• individual patient data meta-analysis comparing hepatic safety and gastrointestinal tolerability (particularly vomiting
in young children and infants within one hour of dosing, as this could alter efficacy) between ASPY and other ACTs;
• continued assessment of efficacy, safety and tolerability of all ACTs, including ASPY, across malaria-endemic regions,
especially in African children;
• further monitoring of efficacy, particularly in children in different settings, and monitoring for adverse events from
inadvertent pregnancy exposures; and
• identification and validation of molecular markers of resistance to pyronaridine.
Evidence To Decision
• Fewer patients taking ACTs containing 3 days of an artemisinin derivative experience treatment failure
within the first 28 days (high-quality evidence).
• Fewer participants taking ACTs containing 3 days of an artemisinin derivative have gametocytaemia at day 7
(high-quality evidence).
Justification
GRADE
In four randomized controlled trials in which the addition of 3 days of artesunate to SP was compared directly with 1
day of artesunate with SP:
Three days of artesunate reduced the PCR-adjusted treatment failure rate within the first 28 days from that with 1
day of artesunate (RR, 0.45; 95% CI, 0.36–0.55, four trials, 1202 participants, high-quality evidence).
Three days of artesunate reduced the number of participants who had gametocytaemia at day 7 from that with 1 day
of artesunate (RR, 0.74; 95% CI, 0.58–0.93, four trials, 1260 participants, high-quality evidence).
Other considerations
The guideline development group considered that 3 days of artemisinin derivative are necessary to provide sufficient
efficacy, promote good adherence and minimize the risk of drug resistance resulting from incomplete treatment.
Remarks
Longer ACT treatment may be required to achieve > 90% cure rate in areas with artemisinin-resistant P. falciparum,
159 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
but there are insufficient trials to make definitive recommendations. A 3-day course of the artemisinin component of
ACTs covers two asexual cycles, ensuring that only a small fraction of parasites remain for clearance by the partner
drug, thus reducing the potential development of resistance to the partner drug. Shorter courses (1–2 days) are
therefore not recommended, as they are less effective, have less effect on gametocytes and provide less protection
for the slowly eliminated partner drug.
160 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
be informed that this ACT should be taken of paediatric tablets containing 25 mg artesunate and 55
immediately after food or a fat containing drink (e.g. mg mefloquine hydrochloride (equivalent to 50 mg
milk), particularly on the second and third days of mefloquine base) and adult tablets containing 100 mg
treatment. artesunate and 220 mg mefloquine hydrochloride
(equivalent to 200 mg mefloquine base)
Artesunate + amodiaquine
Target dose and range: Target doses (ranges) of 4 (2–10)
Formulations currently available: A fixed-dose combination
mg/kg bw per day artesunate and 8.3 (7–11) mg/kg bw per
in tablets containing 25 + 67.5 mg, 50 + 135 mg or 100 +
day mefloquine, given once a day for 3 days
270 mg of artesunate and amodiaquine, respectively
Artesunate + mefloquine dose (mg)
Target dose and range: The target dose (and range) are 4 Body weight (kg)
given daily for 3 days
(2–10) mg/kg bw per day artesunate and 10 (7.5–15) mg/
kg bw per day amodiaquine once a day for 3 days. A total 5 to < 9 25 + 55
therapeutic dose range of 6–30 mg/kg bw per day
9 to < 18 50 + 110
artesunate and 22.5–45 mg/kg bw per dose amodiaquine
is recommended. 18 to < 30 100 + 220
Treatment failure after amodiaquine monotherapy was As concomitant use of rifampicin decreases exposure to
more frequent among children who were underweight for mefloquine, potentially decreasing its efficacy, patients
their age. Therefore, their response to artesunate + taking this drug should be followed up carefully to identify
amodiaquine treatment should be closely monitored. treatment failures.
161 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Factors associated with altered drug exposure and fixed-dose combination. This may compromise adherence
treatment response: The low dose of folic acid (0.4 mg and increase the risk for distribution of loose artesunate
daily) that is required to protect the fetuses of pregnant tablets, despite the WHO ban on artesunate monotherapy.
women from neural tube defects do not reduce the
efficacy of SP, whereas higher doses (5 mg daily) do Resistance is likely to increase with continued widespread
significantly reduce its efficacy and should not be given use of SP, sulfalene– pyrimethamine and cotrimoxazole
concomitantly. (trimethoprim-sulfamethoxazole). Fortunately, molecular
markers of resistance to antifols and sulfonamides
Additional comments: correlate well with therapeutic responses. These should be
monitored in areas in which this drug is used.
The disadvantage of this ACT is that it is not available as a
Children weighing <25kg treated with dihydroartemisinin + piperaquine should receive a minimum of 2.5 mg/kg bw
per day of dihydroartemisinin and 20 mg/ kg bw per day of piperaquine daily for 3 days.
Practical Info
Formulations: Currently available as a fixed-dose combination in tablets containing 40 mg dihydroartemisinin and
320 mg piperaquine and paediatric tablets contain 20 mg dihydroartemisinin and 160 mg piperaquine.
Target dose and range: A target dose (range) of 4 (2–10) mg/kg bw per day dihydroartemisinin and 18 (16–27) mg/kg
bw per day piperaquine given once a day for 3 days for adults and children weighing ≥ 25 kg. The target doses and
ranges for children weighing < 25 kg are 4 (2.5–10) mg/kg bw per day dihydroartemisinin and 24 (20–32) mg/kg bw
per day piperaquine once a day for 3 days.
Recommended dosage regimen: The dose regimen currently recommended by the manufacturer provides adequate
exposure to piperaquine and excellent cure rates (> 95%), except in children < 5 years, who have a threefold
increased risk for treatment failure. Children in this age group have significantly lower plasma piperaquine
concentrations than older children and adults given the same mg/kg bw dose. Children weighing < 25 kg should
receive at least 2.5 mg/kg bw dihydroartemisinin and 20 mg/kg bw piperaquine to achieve the same exposure as
children weighing ≥ 25 kg and adults.
Body weight (kg) Dihydroartemisinin + piperaquine dose (mg) given daily for 3 days
5 to < 8 20 + 160
8 to < 11 30 + 240
11 to < 17 40 + 320
17 to < 25 60 + 480
25 to < 36 80 + 640
High-fat meals should be avoided, as they significantly accelerate the absorption of piperaquine, thereby increasing
162 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
the risk for potentially arrhythmogenic delayed ventricular repolarization (prolongation of the corrected
electrocardiogram QT interval). Normal meals do not alter the absorption of piperaquine.
As malnourished children are at increased risk for treatment failure, their response to treatment should be monitored
closely.
Additional comments: Piperaquine prolongs the QT interval by approximately the same amount as chloroquine but
by less than quinine. It is not necessary to perform an electrocardiogram before prescribing dihydroartemisinin +
piperaquine, but this ACT should not be used in patients with congenital QT prolongation or who have a clinical
condition or are on medications that prolong the QT interval. There has been no evidence of cardiotoxicity in large
randomized trials or in extensive deployment.
Justification
The dosing subgroup reviewed all available dihydroartemisinin-piperaquine pharmacokinetic data (6 published
studies and 10 studies from the WWARN database; total 652 patients) [182][185] and then conducted simulations
of piperaquine exposures for each weight group. These showed lower exposure in younger children with higher risks
of treatment failure. The revised dose regimens are predicted to provide equivalent piperaquine exposures across all
age groups.
Other considerations
This dose adjustment is not predicted to result in higher peak piperaquine concentrations than in older children and
adults, and as there is no evidence of increased toxicity in young children, the GRC concluded that the predicted
benefits of improved antimalarial exposure are not at the expense of increased risk.
163 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
used.
In low-transmission areas, a single dose of 0.25 mg/kg bw primaquine should be given with an ACT to patients with P.
falciparum malaria (except pregnant women, infants aged < 6 months and women breastfeeding infants aged < 6 months)
to reduce transmission. G6PD testing is not required.
Practical Info
In light of concern about the safety of the previously recommended dose of 0.75 mg/kg bw in individuals with G6PD
deficiency, a WHO panel reviewed the safety of primaquine as a P. falciparum gametocytocide and concluded that a
single dose of 0.25 mg/kg bw of primaquine base is unlikely to cause serious toxicity, even in people with G6PD
deficiency [188]. Thus, where indicated a single dose of 0.25mg/kg bw of primaquine base should be given on the first
day of treatment, in addition to an ACT, to all patients with parasitologically confirmed P. falciparum malaria except for
pregnant women, infants < 6 months of age and women breastfeeding infants < 6 months of age, because there are
insufficient data on the safety of its use in these groups.
Dosing table based on the most widely currently available tablet strength (7.5mg base)
Single dose of
Body weight (kg)
primaquine (mg base)
10a to < 25 3.75 3.75
25 to < 50 7.5 7.5
50 to 100 15 15
a Dosing of young children weighing < 10 kg is limited by the tablet sizes currently available.
Please refer to the Policy brief on single-dose primaquine as a gametocytocide in Plasmodium falciparum malaria [189].
Evidence To Decision
• Single doses of primaquine > 0.4 mg/kg bw reduced gametocyte carriage at day 8 by around two thirds
(moderate-quality evidence).
• There are too few trials of doses < 0.4 mg/kg bw to quantify the effect on gametocyte carriage (low-quality
evidence).
• Analysis of observational data from mosquito feeding studies suggests that 0.25 mg/kg bw may rapidly reduce
the infectivity of gametocytes to mosquitoes.
Undesirable effects
164 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• People with severe G6PD deficiency are at risk for haemolysis. At this dose, however, the risk is thought to be
small; there are insufficient data to quantify this risk.
Justification
GRADE
In an analysis of observational studies of single-dose primaquine, data from mosquito feeding studies on 180 people
suggest that adding 0.25 mg/kg primaquine to treatment with an ACT can rapidly reduce the infectivity of gametocytes
to mosquitoes.
In a systematic review of eight randomized controlled trials of the efficacy of adding single-dose primaquine to ACTs for
reducing the transmission of malaria, in comparison with ACTs alone [186]:
• single doses of > 0.4 mg/kg bw primaquine reduced gametocyte carriage at day 8 by about two thirds (RR, 0.34;
95% CI, 0.19–0.59, two trials, 269 participants, high-certainty evidence); and
• single doses of primaquine > 0.6 mg/kg bw reduced gametocyte carriage at day 8 by about two thirds (RR, 0.29;
95% CI, 0.22–0.37, seven trials, 1380 participants, high-certainty evidence).
There have been no randomized controlled trials of the effects on the incidence of malaria or on transmission to
mosquitos.
Other considerations
The guideline development group considered that the evidence of a dose– response relation from observational studies
of mosquito feeding was sufficient to conclude the primaquine dose of 0.25mg/kg bw significantly reduced P. falciparum
transmissibility.
The population benefits of reducing malaria transmission with gametocytocidal drugs such as primaquine require that a
very high proportion of treated patients receive these medicines and that there is no large transmission reservoir of
asymptomatic parasite carriers. This strategy is therefore likely to be effective only in areas of low-intensity malaria
transmission, as a component of elimination programmes.
Remarks
This recommendation excludes high-transmission settings, as symptomatic patients make up only a small proportion of
the total population carrying gametocytes within a community, and primaquine is unlikely to affect transmission.
A major concern of national policy-makers in using primaquine has been the small risk for haemolytic toxicity in G6PD-
deficient people, especially where G6PD testing is not available.
Life-threatening haemolysis is considered unlikely with the 0.25mg/kg bw dose and without G6PD testing [187].
Rationale for the recommendation: The Guideline Development Group considered the evidence on dose–response
relations in the observational mosquito-feeding studies of reduced transmissibility with the dose of 0.25 mg/kg bw and
the judgement of the WHO Evidence Review Group (November 2012). Their view was that the potential public health
benefits of single low-dose (0.25 mg/kg bw) primaquine in addition to an ACT for falciparum malaria, without G6PD
testing, outweigh the potential risk for adverse effects.
165 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
antimalarial drugs. This increases the rate of treatment Large and obese adults
failure with current dosage regimens. The rates of treatment Large adults are at risk for under-dosing when they are
failure are substantially higher in hyperparasitaemic patients dosed by age or in standard pre-packaged adult weight-
and patients in areas with artemisinin-resistant falciparum based treatments. In principle, dosing of large adults should
malaria, and these groups require greater exposure to be based on achieving the target mg/kg bw dose for each
antimalarial drugs (longer duration of therapeutic antimalarial regimen. The practical consequence is that two
concentrations) than is achieved with current ACT dosage packs of an antimalarial drug might have to be opened to
recommendations. It is often uncertain how best to achieve ensure adequate treatment. For obese patients, less drug is
this. Options include increasing individual doses, changing often distributed to fat than to other tissues; therefore, they
the frequency or duration of dosing, or adding an additional should be dosed on the basis of an estimate of lean body
antimalarial drug. Increasing individual doses may not, weight, ideal body weight. Patients who are heavy but not
however, achieve the desired exposure (e.g., lumefantrine obese require the same mg/kg bw doses as lighter patients.
absorption becomes saturated), or the dose may be toxic due
to transiently high plasma concentrations (piperaquine, In the past, maximum doses have been recommended, but
mefloquine, amodiaquine, pyronaridine). An additional there is no evidence or justification for this practice. As the
advantage of lengthening the duration of treatment (by evidence for an association between dose, pharmacokinetics
giving a 5-day regimen) is that it provides additional and treatment outcome in overweight or large adults is
exposure of the asexual cycle to the artemisinin component limited, and alternative dosing options have not been
as well as augmenting exposure to the partner drug. The assessed in treatment trials, it is recommended that this gap
acceptability, tolerability, safety and effectiveness of in knowledge be assessed urgently. In the absence of data,
augmented ACT regimens in these special circumstances treatment providers should attempt to follow up the
should be evaluated urgently. treatment outcomes of large adults whenever possible.
166 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Africa have been treated with amodiaquine alone or pregnancy but that the alterations are insufficient to
combined with SP or artesunate; however, amodiaquine warrant dose modifications at this time. With quinine, no
use for the treatment of malaria in pregnancy has been significant differences in exposure have been seen during
formally documented in only > 1300 pregnancies. Use of pregnancy. Studies of the pharmacokinetics of SP used in
amodiaquine in women in Ghana in the second and third IPTp in many sites show significantly decreased exposure
trimesters of pregnancy was associated with frequent to sulfadoxine, but the findings on exposure to
minor side- effects but not with liver toxicity, bone marrow pyrimethamine are inconsistent. Therefore, no dose
depression or adverse neonatal outcomes. modification is warranted at this time.
Dihydroartemisinin + piperaquine was used successfully in Studies are available of the pharmacokinetics of
the second and third trimesters of pregnancy in > 2000 artemether + lumefantrine, artesunate + mefloquine and
women on the Myanmar–Thailand border for rescue dihydroartemisinin + piperaquine. Most data exist for
therapy and in Indonesia for first-line treatment. SP, artemether + lumefantrine; these suggest decreased
although considered safe, is not appropriate for use as an overall exposure during the second and third trimesters.
artesunate partner drug in many areas because of Simulations suggest that a standard six-dose regimen of
resistance to SP. If artesunate + SP is used for treatment, lumefantrine given over 5 days, rather than 3 days,
co-administration of daily high doses (5 mg) of folate improves exposure, but the data are insufficient to
supplementation should be avoided, as this compromises recommend this alternative regimen at present. Limited
the efficacy of SP. A lower dose of folate (0.4–0.5 mg bw/ data on pregnant women treated with dihydroartemesinin
day) or a treatment other than artesunate + SP should be + piperaquine suggest lower dihydroartemisinin exposure
used. and no overall difference in total piperaquine exposure, but
a shortened piperaquine elimination half-life was noted.
Mefloquine is considered safe for the treatment of malaria The data on artesunate + mefloquine are insufficient to
during the second and third trimesters; however, it should recommend an adjustment of dosage. No data are available
be given only in combination with an artemisinin on the pharmacokinetics of artesunate + amodiaquine in
derivative. pregnant women with falciparum malaria, although drug
exposure was similar in pregnant and non-pregnant women
Quinine is associated with an increased risk for with vivax malaria.
hypoglycaemia in late pregnancy, and it should be used
(with clindamycin) only if effective alternatives are not Lactating women
available. The amounts of antimalarial drugs that enter breast milk
and are consumed by breastfeeding infants are relatively
Primaquine and tetracyclines should not be used in small. Tetracycline is contraindicated in breastfeeding
pregnancy. mothers because of its potential effect on infants’ bones
and teeth. Pending further information on excretion in
Dosing in pregnancy breast milk, primaquine should not be used for nursing
Data on the pharmacokinetics of antimalarial agents used women, unless the breastfed infant has been checked for
during pregnancy are limited. Those available indicate that G6PD deficiency.
pharmacokinetic properties are often altered during
167 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Pregnant women with uncomplicated P. falciparum malaria should be treated with artemether-lumefantrine during
the first trimester.
Practical Info
As with the deployment of any new malaria treatment recommendations, pharmacovigilance and adverse events and
pregnancy outcome surveillance systems should be strengthened.
Evidence To Decision
In various animal studies (including rodents and monkeys), artemisinin has been found to deplete embryonic
erythroblasts at relatively low doses of 1/200–1/400 of the LD50 (equivalent to > 10 mg/kg body weight),
leading to malformation or embryonic death [193][197]. The adverse effects include embryo resorption,
pregnancy loss and congenital anomalies, including shortening of the long bones and heart defects (ventricular
septal and great vessel defects) [198][199]. For this reason, despite its demonstrated lower efficacy in the second
and third trimesters of pregnancy, quinine (in combination with clindamycin) was retained by WHO in 2015 for
treatment in the first trimester until adequate numbers of human exposures to artemisinin could allow for more
safety assessments in humans.
In weighing the risk–benefit ratio, safety risks from antimalarial treatment need to be weighed against the
adverse effects of malaria in the first trimester [200].
A recently updated individual patient data meta-analysis of 34 178 pregnancies included 737 well documented
pregnancies exposed to artemisinin and 1076 exposed to non-artemisinin-based treatments in the first
trimester. Of the exposures to artemisinin, 71% (525) were to artemether-lumefantrine [201]. This meta-analysis
provided the basis for the re-evaluation of the treatment of malaria in the first trimester of pregnancy.
This updated individual patient data review showed that first-trimester treatment with artemether-lumefantrine
was associated with significantly fewer adverse pregnancy outcomes than first-trimester treatment with quinine.
Treatment with artemether-lumefantrine in the first trimester was associated with a statistically significant lower
risk (42%) of adverse pregnancy outcomes compared to treatment with oral quinine (aHR: 0.58; 95% CI:
0.36–0.92) [201]. The numbers of exposures to the other ACTs (excluding artesunate-pyronaridine) were too
small to allow for a subgroup analysis [201]. There is currently no documented record of the use of artesunate-
pyronaridine during the first trimester of pregnancy. Combined with the known better tolerability and
effectiveness and the longer duration of post-treatment prophylaxis, artemether-lumefantrine clearly has a more
168 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
favourable risk–benefit profile than quinine for treating uncomplicated falciparum malaria in the first trimester.
An analysis of all exposures to artemisinin-based treatment (ABT) in the first trimester of pregnancy as a means
of addressing the concerns previously demonstrated in animal studies showed no differences between
pregnancies exposed in the first trimester to artemisinin and those exposed to non-ABT in terms of the
composite adverse pregnancy outcome (ABT=42/736 [5.7%] vs non-ABT=96/1074 [8.9%]; aHR: 0.71; 95% CI:
0.49–1.03). Analysis for adverse pregnancy outcomes against the individual parameters in the composite
analysis, including miscarriage, stillbirth or congenital anomalies, also revealed no statistically significant
difference. There was also no difference in the risk of these adverse pregnancy outcomes when exposures were
restricted to the putative embryo-sensitive period. This meta-analysis had 8126 additional pregnancies with 60
additional artemisinin exposures in the first trimester compared to the review published in 2017 [202]. This
analysis [201] strengthens previous findings of the 2017 review that the potential for artemisinin-based
embryotoxicity observed in animal studies is not reflected in humans treated for malaria. The analysis also
demonstrates how few pregnancy outcomes after ACT exposures in the first trimester of pregnancy can be
documented.
The teratogenic effect of the artemisinin observed in animal studies was not apparent in any of the reviewed
data on human exposure to ACTs in the first trimester of pregnancy. However, there are some reasons to
exercise caution in drawing a definite conclusion on the safety of the artemisinin derivatives as a drug class.
These reasons include: the possibility of immortal time bias, resulting in an inability to detect early fetal losses;
potential bias in observational study designs with exposure to quinine as the main comparator; and current
limited postnatal evaluation, e.g. for cardiovascular and other malformations [203]. In addition, most of the
safety data are on artemether-lumefantrine. Although artemisinin derivatives are rapidly converted to
dihydroartemisinin as their active metabolite, differences between the different derivatives, or differences
caused by the combination with different partner drugs cannot be excluded.
In terms of the safety and tolerability of the currently recommended ACT partner antimalarials, the antifolate
sulfadoxine-pyrimethamine is contraindicated during the first trimester of pregnancy, as it is known to have a
potential teratogenic risk in humans at therapeutic doses [204]. There is currently no documented exposure to
pyronaridine in the first trimester of pregnancy. Among 3428 pregnant women in the second or third trimester
treated with an ACT for P. falciparum malaria (at any parasite density and regardless of symptoms), drug-related
adverse events such as asthenia, poor appetite, dizziness, nausea and vomiting occurred significantly more
frequently in the artesunate-mefloquine group (50.6%) and the artesunate-amodiaquine group (48.5%) than in
the dihydroartemisinin-piperaquine group (20.6%) and the artemether-lumefantrine group (11.5%) (p<0.001 for
comparison among the four groups) [205].
There is a lack of documented exposures very early in gestation (gestational weeks 4–10), which is considered a
critical period for teratogenic risk. Therefore, the potential for any given medicine, including quinine, to cause a
specific teratogenic effect can only be reliably ascertained when it has been administered during this sensitive
period (which is almost impossible to study, given how soon this critical window occurs after the last menstrual
period) [203].
The GDG judged the overall certainty of the assessed evidence across the different outcomes to be low due to
bias inherent in observational studies. It was difficult to generalize across all ACTs because of the limited number
of pregnant women in the first trimester treated with ACTs other than artemether-lumefantrine who were
included in the review.
169 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
to be a more attractive option because of its greater availability and the convenience of a shorter, better
tolerated treatment. Policy-makers and implementers will obviously prefer simplified recommendations on using
artemether-lumefantrine or other ACTs to treat pregnant women with uncomplicated P. falciparum malaria across
all trimesters.
According to a systematic review of sociocultural factors [206], malaria in pregnancy is interpreted in locally
defined categories, despite the higher malaria risk associated with pregnancy. Local context and health workers’
ideas and comments influence concerns about malaria in pregnancy interventions. Factors such as the
understanding of antenatal care, health worker–client interactions, household decision-making, gender relations,
cost and distance to health facilities affect pregnant women’s access to these interventions and their health-
seeking behaviour. It is difficult to ascertain whether any sociocultural factors would result in variability in the
likely preference for artemether-lumefantrine or other ACTs over quinine treatment.
Resources
ACTs, of which artemether-lumefantrine is the most widely used, are the treatment of choice for uncomplicated
falciparum malaria in nearly all malaria-endemic countries and are thus readily available. Conversely, the supply
of quinine has become problematic because of the small proportion of the total population that receive this
antimalarial treatment. Clindamycin, which is recommended in combination with quinine, is commonly
unavailable and unaffordable in most endemic regions. In addition, the quinine + clindamycin regime is
associated with a high pill burden, requiring between 56 and 70 tablets to be ingested over a seven-day period.
In most country programmes, quinine monotherapy is thus currently recommended in the first trimester of
pregnancy. However, in some countries in eastern and southern Africa, quinine is rarely available in public
facilities, and many pregnant women in the first trimester are already being treated with artemether-
lumefantrine, based on reports from the national malaria programmes.
Aligning the first-line treatment of uncomplicated P. falciparum malaria for the first trimester with that currently
recommended for the second and third trimesters would simplify case management, service delivery,
communications and supply chain management. Such an alignment was assessed as likely to result in large
savings. However, research on formal cost analysis and cost estimates regarding the use of artemether-
lumefantrine or other ACTs versus quinine in the first trimester of pregnancy are still lacking.
Equity
Despite the obvious efficiency to be gained by harmonizing the treatment regimens throughout pregnancy, no
studies were found. However, health equity will increase, especially for vulnerable populations, if this more
effective, more accessible and better tolerated treatment is recommended for the management of malaria in all
trimesters of pregnancy.
Acceptability
In considering the acceptability of artemether-lumefantrine versus quinine treatments, the GDG looked to how
quinine is presently being used and accepted.
Adherence to quinine is low because it is frequently associated with adverse effects, including cinchonism,
nausea and hypoglycaemia [195][207][208]. In a review of 35 national guidelines, 66% recommended oral
quinine as first-line treatment for uncomplicated malaria in the first trimester of pregnancy. Of these, only 29%
included the combined use with clindamycin in their guidelines, reflecting the unavailability and/or cost of
clindamycin [209]. Health care reliance on clinical diagnosis and poor adherence to treatment policy, especially in
the first trimester, have been consistently reported. Prescribing practices have been driven by concerns over side
effects and drug safety, patient preferences, drug availability and cost [210].
With poor adherence to the presently recommended quinine-based treatments and better access to ACTs, it
appears that artemether-lumefantrine will be a more acceptable option. A three-day ACT treatment regimen is
170 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Policy-makers and health care workers will likely welcome the evidence-based decision recommending
artemether-lumefantrine for all trimesters of pregnancy. In situations where artemether-lumefantrine is no
longer recommended for the treatment of malaria because of reduced efficacy and/or it is not promptly
available, the use of some of the other ACTs recommended in national guidelines can be considered, given the
demonstrated poorer outcomes of quinine treatment, along with the challenges of adherence to a seven-day
treatment course. However, artesunate plus sulfadoxine-pyrimethamine cannot be recommended in the first
trimester of pregnancy given the potential teratogenicity of antifolates. Furthermore, the lack of documented
outcomes following the use of artesunate-pyronaridine precludes its use in the first trimester.
Feasibility
One consideration in determining the feasibility of the recommendation on treatment of malaria in the first
trimester is that the existing warning against the use of artemisinin in the first trimester implies the need to
consistently screen for pregnancy among all women of childbearing potential prior to treatment for malaria.
However, pregnancy screening is rarely done prior to initiating malaria treatment. As observed by national
programmes, the contraindication of artemisinin in the first trimester has resulted in confusion, most
problematically resulting in pregnant women in the first trimester with severe malaria not receiving the
recommended parenteral artesunate, thereby increasing malaria morbidity and mortality in this particularly
vulnerable subgroup [211][212].
Given that ACTs, particularly artemether-lumefantrine, are already widely used in the treatment of malaria in
pregnancy, although mainly in the second and third trimesters, uptake of artemether-lumefantrine (and other
ACTs) should be feasible in the first trimester of pregnancy. There will also be less confusion once the
recommendations are aligned across all trimesters of pregnancy, implying that artemether-lumefantrine or other
ACTs should be more feasible and adherence to the implementation strategies should improve relative to that
with quinine-based treatment.
Justification
The GDG reached a consensus on a strong recommendation for artemether-lumefantrine as the preferred treatment
of uncomplicated Plasmodium falciparum malaria during the first trimester of pregnancy, despite the low certainty of
evidence because:
• there was a large magnitude of beneficial effect of treatment on efficacy (demonstrated in the second and third
trimesters of pregnancy), specifically a six-fold reduction in treatment failures following artemether-
lumefantrine, compared to the currently recommended quinine-based therapies;
• artemether-lumefantrine was associated with trivial adverse events and significantly lower risk for adverse
pregnancy outcomes in the first trimester of pregnancy;
• artemether-lumefantrine had much better tolerability compared to quinine-based therapies; and
• there is probably increased equity, acceptability and feasibility, resulting from better access to artemether-
lumefantrine and more efficient implementation of ACTs compared to quinine-based treatments.
171 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Research Needs
• Although the safety of ACTs is reassuring and the independent patient data meta-analysis indicated that there is
no apparent effect of gestational age on the risk of PCR-corrected treatment efficacy, collecting further
evidence from clinical studies and close surveillance on the safety of ACT treatment in the first trimester must
be continued and funded. This is particularly the case for artesunate-amodiaquine, artesunate-mefloquine,
dihydroartemisinin-piperaquine and artesunate-pyronaridine, given the relatively few pregnant women in the
first trimester with documented exposures to these drugs compared to artemether-lumefantrine.
• Operational studies including pregnancy registries are needed to strengthen pharmacovigilance among pregnant
women and capture data from mass drug administration programmes and other interventions that may result in
inadvertent exposures to ACTs during the first trimester.
• Continued pharmacovigilance and clinical research, such as controlled experimental studies on the efficacy and
safety of antimalarial medicines, including new antimalarials, for the treatment of malaria in pregnancy, should
be supported and funded given the high burden of malaria in pregnancy globally.
172 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
distinguished between acute and chronic malnutrition. Small studies of the pharmacokinetics of quinine and
Oral absorption of drugs may be reduced if there is chloroquine showed alterations in people with different
diarrhoea or vomiting, or rapid gut transit or atrophy of the degrees of malnutrition. Studies of SP in IPTp and of
small bowel mucosa. Absorption of intramuscular and amodiaquine monotherapy and dihydroartemisinin +
possibly intrarectal drugs may be slower, and diminished piperaquine for treatment suggest reduced efficacy in
muscle mass may make it difficult to administer repeated malnourished children. A pooled analysis of data for
intramuscular injections to malnourished patients. The individual patients showed that the concentrations of
volume of distribution of some drugs may be larger and the lumefantrine on day 7 were lower in children < 3 years
plasma concentrations lower. Hypoalbuminaemia may who were underweight for age than in adequately
reduce protein binding and increase metabolic clearance, nourished children and adults. Although these findings are
but concomitant hepatic dysfunction may reduce the concerning, they are insufficient to warrant dose
metabolism of some drugs; the net result is uncertain. modifications (in mg/kg bw) of any antimalarial drug in
patients with malnutrition.
Infants weighing < 5 kg with uncomplicated P. falciparum malaria should be treated with an ACT at the same mg/kg
bw target dose as for children weighing 5 kg.
Practical Info
The pharmacokinetics properties of many medicines in infants differ markedly from those in adults because of the
physiological changes that occur in the first year of life. Accurate dosing is particularly important for infants. The only
antimalarial agent that is currently contraindicated for infants (< 6 months) is primaquine.
ACT is recommended and should be given according to body weight at the same mg/kg bw dose for all infants,
including those weighing < 5 kg, with close monitoring of treatment response. The lack of infant formulations of
most antimalarial drugs often necessitates division of adult tablets, which can lead to inaccurate dosing. When
available, paediatric formulations and strengths are preferred, as they improve the effectiveness and accuracy of ACT
dosing.
Evidence To Decision
• There is some evidence that artemether + lumefantrine and dihydroartemisinin + piperaquine may achieve
lower plasma concentrations in infants than in older children and adults.
Justification
Evidence supporting the recommendation
Data available were not suitable for evaluation using the GRADE methodology.
In most clinical studies, subgroups of infants and older children were not distinguished, and the evidence for young
infants (< 5 kg) is insufficient for confidence in current treatment recommendations. Nevertheless, despite these
uncertainties, infants need prompt, effective treatment of malaria. There is limited evidence that artemether +
lumefantrine and dihydroartemisinin + piperaquine achieve lower plasma concentrations in infants than in older
children and adults.
Other considerations
The Guideline Development Group considered the currently available evidence too limited to warrant formal
evidence review at this stage, and was unable to recommend any changes beyond the status quo. Further research is
173 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
warranted.
In people who have HIV/AIDS and uncomplicated P. falciparum malaria, artesunate + SP is not recommended if they
are being treated with co-trimoxazole, and artesunate + amodiaquine is not recommended if they are being treated
with efavirenz or zidovudine.
Justification
More data are available on use of artemether + lumefantrine with antiretroviral treatment. A study in children with
uncomplicated malaria in a high-transmission area of Africa showed a decreased risk for recurrent malaria after
treatment with artemether + lumefantrine in children receiving lopinavir–ritonavir-based antiretroviral treatment as
compared with non-nucleoside reverse transcriptase inhibitor-based antiretroviral treatment. Evaluation of
pharmacokinetics in these children and in healthy volunteers showed significantly higher exposure to lumefantrine
and lower exposure to dihydroartemisinin with lopinavir–ritonavir-based antiretroviral treatment, but no adverse
consequences. Conversely, efavirenz-based antiretroviral treatment was associated with a two- to fourfold decrease
in exposure to lumefantrine in healthy volunteers and malaria-infected adults and children, with increased rates of
recurrent malaria after treatment. Close monitoring is required. Increasing artemether + lumefantrine dosing with
efavirenz-based antiretroviral treatment has not yet been studied. Exposure to lumefantrine and other non-
nucleoside reverse transcriptase inhibitor-based antiretroviral treatment, namely nevirapine and etravirine, did not
show consistent changes that would require dose adjustment.
Studies of administration of quinine with lopinavir–ritonavir or ritonavir alone in healthy volunteers gave conflicting
results. The combined data are insufficient to justify dose adjustment. Single-dose atovaquone–proguanil with
efavirenz, lopinavir–ritonavir or atazanavir–ritonavir were all associated with a significantly decreased area under the
concentration–time curve for atovaquone (two- to fourfold) and proguanil (twofold), which could well compromise
treatment or prophylactic efficacy. There is insufficient evidence to change the current mg/kg bw dosing
174 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Travellers with uncomplicated P. falciparum malaria returning to non-endemic settings should be treated with an ACT.
Evidence To Decision
Justification
GRADE
Studies have consistently demonstrated that the five WHO recommended ACTs have less than 5% PCR-adjusted
treatment failure rates in settings without resistance to the partner drug (high quality evidence).
Other considerations
The Guideline Development Group considered the evidence of superiority of ACTs over non-ACTs from endemic
settings to be equally applicable to those travelling from non-endemic settings.
Hyperparasitaemia (2015)
People with P. falciparum hyperparasitaemia are at increased risk for treatment failure, severe malaria and death and
should be closely monitored, in addition to receiving an ACT.
175 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Justification
In falciparum malaria, the risk for progression to severe malaria with vital organ dysfunction increases at higher
parasite densities. In low-transmission settings, mortality begins to increase when the parasite density exceeds 100
000/µL (~2% parasitaemia). On the north-west border of Thailand, before the general introduction of ACT,
parasitaemia > 4% without signs of severity was associated with a 3% mortality rate (about 30-times higher than
from uncomplicated falciparum malaria with lower densities) and a six-times higher risk of treatment failure. The
relationship between parasitaemia and risks depends on the epidemiological context: in higher-transmission settings,
the risk of developing severe malaria in patients with high parasitaemia is lower, but “uncomplicated
hyperparasitaemia” is still associated with a significantly higher rate of treatment failure.
Patients with a parasitaemia of 4–10% and no signs of severity also require close monitoring, and, if feasible,
admission to hospital. They have high rates of treatment failure. Non-immune people such as travellers and
individuals in low-transmission settings with a parasitaemia > 2% are at increased risk and also require close
attention. Parasitaemia > 10% is considered to indicate severe malaria in all settings.
It is difficult to make a general recommendation about treatment of uncomplicated hyperparasitaemia, for several
reasons: recognizing these patients requires an accurate, quantitative parasite count (they will not be identified from
semi-quantitative thick film counts or RDTs), the risks for severe malaria vary considerably, and the risks for
treatment failure also vary. Furthermore, little information is available on therapeutic responses in uncomplicated
hyperparasitaemia. As the artemisinin component of an ACT is essential in preventing progression to severe malaria,
absorption of the first dose must be ensured (atovaquone – proguanil alone should not be used for travellers
presenting with uncomplicated hyperparasitaemia). Longer courses of treatment are more effective; both giving
longer courses of ACT and preceding the standard 3-day ACT regimen with parenteral or oral artesunate have been
used.
176 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
If the malaria species is not known with certainty, adults and children should be treated as for uncomplicated P.
falciparum malaria.
177 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
In areas with chloroquine-susceptible infections, adults and children with uncomplicated P. vivax, P. ovale, P. malariae or P.
knowlesi malaria should be treated with either an ACT or chloroquine.
In areas with chloroquine-resistant infections, adults and children with uncomplicated P. vivax, P. ovale, P. malariae or P.
knowlesi malaria should be treated with an ACT.
* For details of treatment using ACTs in the first trimester of pregnancy, see section 5.2.1.4.1.
Practical Info
In areas with chloroquine-sensitive P. vivax
For chloroquine-sensitive vivax malaria, oral chloroquine at a total dose of 25 mg base/kg bw is effective and well
tolerated. Lower total doses are not recommended, as these encourage the emergence of resistance. Chloroquine is
given at an initial dose of 10 mg base/kg bw, followed by 10 mg/kg bw on the second day and 5 mg/kg bw on the third
day. In the past, the initial 10-mg/kg bw dose was followed by 5 mg/kg bw at 6 h, 24 h and 48 h. As residual chloroquine
suppresses the first relapse of tropical P. vivax (which emerges about 3 weeks after onset of the primary illness), relapses
begin to occur 5–7 weeks after treatment if radical curative treatment with primaquine is not given.
ACTs are highly effective in the treatment of vivax malaria, allowing simplification (unification) of malaria treatment; i.e.
all malaria infections can be treated with an ACT. The exception is artesunate + SP, where resistance significantly
compromises its efficacy. Although good efficacy of artesunate + SP was reported in one study in Afghanistan, in several
other areas (such as South-East Asia) P. vivax has become resistant to SP more rapidly than P. falciparum. The initial
response to all ACTs is rapid in vivax malaria, reflecting the high sensitivity to artemisinin derivatives, but, unless
primaquine is given, relapses commonly follow. The subsequent recurrence patterns differ, reflecting the elimination
kinetics of the partner drugs. Thus, recurrences, presumed to be relapses, occur earlier after artemether + lumefantrine
than after dihydroartemisinin + piperaquine or artesunate + mefloquine because lumefantrine is eliminated more rapidly
than either mefloquine or piperaquine. A similar temporal pattern of recurrence with each of the drugs is seen in the P.
vivax infections that follow up to one third of acute falciparum malaria infections in South-East Asia.
In the systematic review of ACTs for treating P. vivax malaria, dihydroartemisinin + piperaquine provided a longer
prophylactic effect than ACTs with shorter half-lives (artemether + lumefantrine, artesunate + amodiaquine), with
significantly fewer recurrent parasitaemias during 9 weeks of follow-up (RR, 0.57; 95% CI, 0.40–0.82, three trials, 1066
participants). The half-life of mefloquine is similar to that of piperaquine, but use of dihydroartemisinin + piperaquine in
P. vivax mono-infections has not been compared directly in trials with use of artesunate + mefloquine.
The blood stages of P. ovale, P. malariae and P. knowlesi should therefore be treated with the standard regimen of ACT or
chloroquine, as for vivax malaria.
Mixed infections are best detected by nucleic acid-based amplification techniques, such as PCR; they may be
178 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
underestimated with routine microscopy. Cryptic P. falciparum infections in vivax malaria can be revealed in
approximately 75% of cases by RDTs based on the PfHRP2 antigen, but several RDTs cannot detect mixed infection or
have low sensitivity for detecting cryptic vivax malaria. ACTs are effective against all malaria species and so are the
treatment of choice for mixed infections.
Evidence To Decision
Justification
GRADE
In a systematic review of ACTs for the treatment of P. vivax malaria [226], five trials were conducted in Afghanistan,
Cambodia, India, Indonesia and Thailand between 2002 and 2011 with a total of 1622 participants which compared
ACTs directly with chloroquine. In comparison with chloroquine:
ACTs cleared parasites from the peripheral blood more quickly (parasitaemia after 24 h of treatment: RR, 0.42; 95% CI,
0.36–0.50, four trials, 1652 participants, high-quality evidence); and
ACTs were at least as effective in preventing recurrent parasitaemia before day 28 (RR, 0.58; 95% CI, 0.18–1.90, five
trials, 1622 participants, high-quality evidence).
In four of these trials, few cases of recurrent parasitaemia were seen before day 28 with both chloroquine and ACTs. In
the fifth trial, in Thailand in 2011, increased recurrent parasitaemia was seen after treatment with chloroquine (9%), but
was infrequent after ACT (2%) (RR, 0.25; 95% CI, 0.09–0.66, one trial, 437 participants).
ACT combinations with long half-lives provided a longer prophylactic effect after treatment, with significantly fewer
cases of recurrent parasitaemia between day 28 and day 42 or day 63 (RR, 0.57; 95% CI, 0.40–0.82, three trials, 1066
participants, moderate-quality evidence).
Other considerations
The guideline development group recognized that, in the few settings in which P. vivax is the only endemic species and
where chloroquine resistance remains low, the increased cost of ACT may not be worth the small additional benefits.
Countries where chloroquine is used for treatment of vivax malaria should monitor for chloroquine resistance and
change to ACT when the treatment failure rate is > 10% at day 28.
Remarks
Current methods cannot distinguish recrudescence from relapse or relapse from newly acquired infections, but the aim
of treatment is to ensure that the rates of recurrent parasitaemia of any origin are < 10%.
Primaquine has significant asexual stage activity against vivax malaria and augments the therapeutic response to
chloroquine. When primaquine is given routinely for 14 days, it may mask low-level chloroquine resistance and prevent
vivax recurrence within 28 days.
179 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
these settings, chloroquine may still be considered, but countries should monitor chloroquine resistance and change to
ACT when the treatment failure rate is > 10% on day 28.
--
Remarks
Current methods do not distinguish recrudescence from relapse or relapse from newly acquired infection, but the aim of
treatment is to ensure that the rates of recurrent parasitaemia of any origin is < 10% within 28 days.
When primaquine is not given for radical cure, slowly eliminated ACT that prevents recurrent parasitaemia before day 28
should be used (dihydroartemisinin + piperaquine or artesunate + mefloquine).
Primaquine has significant asexual stage activity against vivax malaria and augments the therapeutic response to
chloroquine. When primaquine is given routinely for 14 days, it may mask low-level chloroquine resistance and prevent
vivax recurrence within 28 days.
When primaquine is given routinely for 14 days, ACTs with shorter half-lives (artemether + lumefantrine, or artesunate +
amodiaquine) may be sufficient to keep the rate of recurrent parasitaemia before day 28 below 10%.
The G6PD status of patients should be used to guide administration of primaquine for preventing relapse.
Practical Info
Please refer to Testing for G6PD deficiency for safe use of primaquine in radical cure of P. vivax and P. ovale (Policy
brief) [227] and Guide to G6PD deficiency rapid diagnostic testing to support P. vivax radical cure [172].
To prevent relapse, children and adults (except pregnant women, infants aged < 6 months, women breastfeeding older
infants unless they are known not to be G6PD deficient, and people with G6PD deficiency) should be treated with a
14-day course of primaquine in all transmission settings.
Practical Info
Primaquine for preventing relapse
To achieve radical cure (cure and prevention of relapse), relapses originating from liver hypnozoites must be prevented by
giving primaquine. The frequency and pattern of relapses varies geographically, with relapse rates generally ranging from
8% to 80%. Temperate long-latency P. vivax strains are still prevalent in many areas. Recent evidence suggests that, in
endemic areas where people are inoculated frequently with P. vivax, a significant proportion of the population harbours
dormant but “activatable” hypnozoites. The exact mechanism of activation of dormant hypnozoites is unclear. There is
evidence that systemic parasitic and bacterial infections, but not viral infections, can activate P. vivax hypnozoites, which
explains why P. vivax commonly follows P. falciparum infections in endemic areas where both parasites are prevalent.
Thus, the radical curative efficacy of primaquine must be set against the prevalent relapse frequency and the likely
burden of “activatable” hypnozoites. Experimental studies on vivax malaria and the relapsing simian malaria P. cynomolgi
suggest that the total dose of 8-aminoquinoline given is the main determinant of radical curative efficacy. In most
180 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
therapeutic assessments, primaquine has been given for 14 days. Total doses of 3.5 mg base/kg bw (0.25 mg/kg bw per
day) are required for temperate strains and 7 mg base/kg bw (0.5 mg/kg bw per day) is needed for the tropical, frequent-
relapsing P. vivax prevalent in East Asia and Oceania. Primaquine causes dose-limiting abdominal discomfort when taken
on an empty stomach; it should always be taken with food.
Use of primaquine to prevent relapse in high-transmission settings was not recommended previously, as the risk for new
infections was considered to outweigh any benefits of preventing relapse. This may have been based on underestimates
of the morbidity and mortality associated with multiple relapses, particularly in young children. Given the benefits of
preventing relapse and in the light of changing epidemiology worldwide and more aggressive targets for malaria control
and elimination, the group now recommends that primaquine be used in all settings.
Primaquine formulation: If available, administer scored tablets containing 7.5 or 15 mg of primaquine. Smaller-dose
tablets containing 2.5 and 5 mg base are available in some areas and facilitate accurate dosing in children. When scored
tablets are not available, 5 mg tablets can be used.
Therapeutic dose: 0.25–0.5 mg/kg bw per day primaquine once a day for 14 days.
Use of primaquine to prevent relapse in high-transmission settings was not recommended previously, as the risk for new
infections was considered to outweigh any benefits of preventing relapse. This may have been based on underestimates
of the morbidity and mortality associated with multiple relapses, particularly in young children. Given the benefits of
preventing relapse and in the light of changing epidemiology worldwide and more aggressive targets for malaria control
and elimination, the group now recommends that primaquine be used in all settings.
Evidence To Decision
• 14-day courses of primaquine added to chloroquine reduce relapse rates to a greater extent than chloroquine
alone (high-quality evidence).
• 14-day courses of primaquine added to chloroquine may result in fewer relapses than 7-day courses (low-
quality evidence).
Undesirable effects:
• Primaquine is known to cause haemolysis in people with G6PD deficiency.
• Of the 15 trials included in the Cochrane review, 12 explicitly excluded people with G6PD deficiency; in three
trials, it was unclear whether participants were tested for G6PD deficiency or excluded. None of the trials
reported serious or treatment-limiting adverse events.
Justification
GRADE
In a systematic review of primaquine for radical cure of P. vivax malaria [231], 14 days of primaquine was compared with
placebo or no treatment in 10 trials, and 14 days was compared with 7 days in one trial. The trials were conducted in
Colombia, Ethiopia, India, Pakistan and Thailand between 1992 and 2006.
• 14 days of primaquine (0.25 mg/kg bw per day) reduced relapses during 15 months of follow-up by about 40% (RR,
0.60; 95% CI, 0.48–0.75, 10 trials, 1740 participants, high-quality evidence).
• 14 days of primaquine (0.25 mg/kg bw per day) reduced relapses during 6 months of follow-up by over 50% (RR,
181 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
0.45; 95% CI, 0.25–0.81, one trial, 126 participants, low-quality evidence).
No direct comparison has been made of higher doses (0.5 mg/kg bw for 14 days) with the standard regimen (0.25 mg/kg
bw for 14 days).
Twelve of the 15 trials included in the review explicitly excluded people with G6PD deficiency; the remaining three did
not report on this aspect. No serious adverse events were reported.
Other considerations
In the absence of evidence to recommend alternatives, the guideline development group considers 0.75 mg/kg bw
primaquine given once weekly for 8 weeks to be the safest regimen for people with mild-to-moderate G6PD deficiency.
Remarks
The widely used primaquine regimen of 0.25 mg base/kg bw per day for 14 days is based on studies of long-latency
Korean P. vivax.
In South-East Asia and Oceania, P. vivax relapses at 3-week intervals and is more resistant to primaquine. Consequently,
higher doses of primaquine have been used (0.375–0.5 mg base/kg bw per day), but there are few data from
comparative trials.
Primaquine is contraindicated in pregnancy and lactation < 6 months post-partum, unless the infant has been tested for
G6PD deficiency. It could be given to women who have delivered and ceased breastfeeding.
In the light of changing epidemiology worldwide and more aggressive targets for malaria control and elimination, the
group now recommends primaquine for radical cure of P. vivax in all settings.
To prevent relapse, an additional treatment option of using primaquine 0.5 mg/kg/day for seven days is recommended
to treat P. vivax or P. ovale malaria in children and adults (except pregnant women, infants aged < 6 months, women
breastfeeding infants aged < 6 months, women breastfeeding older infants unless they are known not to be G6PD
deficient, and people with G6PD deficiency).
• As recommended previously, the G6PD status of patients should be used to guide administration of primaquine for
preventing relapse.
• A shorter regimen can lead to better adherence compared to the standard 14-day regimen and thus to fewer relapses.
Practical Info
• Health facilities should continue to monitor for vomiting, anaemia, haemolysis and adherence to treatment.
• Safety monitoring is critical, so strengthening of pharmacovigilance systems is generally needed.
• Surveillance for risk of false-negative results with G6PD tests and for lack of G6PD testing is essential.
Evidence To Decision
182 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
course and may thus decrease the burden of P. vivax relapses. There seems to be no difference in the safety and
tolerability of the standard dose (0.5 mg/kg/day for seven days) compared to 0.25 mg/kg/day for 14 days.
The GDG judged the overall certainty of the evidence to be very low due to the risk of bias, indirectness and serious
imprecision.
Resources
Given that the total dosage will be the same but with a shortened course, differences in costs and savings will be
negligible. However, prescriber training and end-user information to improve rates of prescribing primaquine and
compliance might be needed. As with the current recommendation for the use of primaquine, G6PD testing to guide
the dosing of primaquine needs to be included in the costing for resources.
Equity
Equity may vary depending on the different populations affected. As malaria is a poverty-related disease, improving
treatment efficacy will probably increase equity. Shorter treatment courses with proper health information will likely
lead to fewer relapses. It is assumed that the burden of these relapses falls disproportionately on the poorer
population. However, there is a possibility of reduced equity if poorer populations have more limited access to the
G6PD testing required prior to therapy.
Acceptability
The intervention of 0.5 mg/kg/day for seven days is probably more acceptable and may be preferred by
stakeholders to the current 0.25 mg/kg/day for 14 days.
Feasibility
Feasibility should improve with a shorter treatment course, provided that updated treatment guidelines and training
with the appropriate supporting infrastructure can be delivered.
Justification
The GDG reached a consensus on a strong recommendation for the intervention, despite the very low certainty of
evidence because:
• the moderate magnitude of treatment effect was comparable to that of the standard regimen of 0.25 mg/kg/day for
14 days;
• no significant serious adverse events were detected;
• there is probably increased equity from access to an additional treatment option; and
• there is an expected increase in adherence because of the shorter course.
183 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Research Needs
The GDG highlighted the following evidence gaps requiring further research.
• Studies are needed on the supporting criteria in decision-making, such as patients’ values and preferences relating
to the critical outcomes, costs, equity and acceptability, as well as feasibility studies on the different antimalarial
treatment options.
• Studies are needed on the optimal primaquine dosage by region (high versus low relapse periodicity) and by
subgroup (including young children).
To prevent relapse, an additional treatment option of using primaquine 1.0 mg/kg/day for seven days to treat P. vivax or
P. ovale malaria is not recommended.
• There is a significantly increased risk of serious adverse events (moderate to large undesirable effect) at this daily dosing of
the standard high dose.
Evidence To Decision
The GDG judged the overall certainty of the evidence to be very low due to risk of indirectness, imprecision and risk
of bias in adverse event outcomes.
Resources
Proper instruction and follow-up (especially for complications due to haemolysis) and management of adverse
184 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
events can be an additional cost factor. The costs of G6PD screening should be included in the cost analysis if the 1
mg/kg/day regimen is used.
Equity
Impact on equity may vary depending on the population and risk of serious adverse events. Although a shorter
regimen may lead to better adherence and fewer relapses, the significant increase in adverse events may tilt the
balance. There is the possibility of reduced equity if poorer populations have less access to G6PD testing.
Haemolysis in G6PD heterozygous female patients with intermediate enzyme levels not detected by a quantitative
G6PD test remains a concern.
Acceptability
The shorter regimen is probably more acceptable. At the same time, the significant increase in adverse events is
probably unacceptable in most settings. Gastrointestinal tolerability of the seven-day high-dose regimen seems
worse than in standard 14 day treatment course, although this can potentially be mitigated by intake with food.
Feasibility
Feasibility improves with a shorter treatment course, but the concomitant G6PD screening and monitoring and
management of expected adverse events might not be feasible or affordable in some settings.
Justification
The GDG reached a consensus on a recommendation against this regimen because:
• there were significant serious adverse events at this higher daily dosage.
Research Needs
The GDG highlighted the following evidence gaps requiring further research.
• Larger effectiveness and safety studies are needed, focusing on gastrointestinal tolerability with and without food
and the impact of G6PD testing on haemolysis risk (in particular in heterozygous women).
• Studies are needed on primaquine optimal dosage in areas where the Chesson strain is prevalent, as higher total
doses of primaquine may be required in these settings.
• Implementation research is needed to increase the acceptability and use of G6PD point-of-care testing.
In people with G6PD deficiency, primaquine base at 0.75 mg/kg bw once a week for 8 weeks can be given to prevent
relapse, with close medical supervision for potential primaquine-induced haemolysis.
Practical Info
• In patients known to be G6PD deficient, primaquine may be considered at a dose of 0.75 mg base/kg bw once a
week for 8 weeks. The decision to give or withhold primaquine should depend on the possibility of giving the
treatment under close medical supervision, with ready access to health facilities with blood transfusion services.
• Some heterozygote females who test as normal or not deficient in qualitative G6PD screening tests have
intermediate G6PD activity and can still haemolyse substantially. Intermediate deficiency (30–80% of normal) and
normal enzyme activity (> 80% of normal) can be differentiated only with a quantitative test. In the absence of
185 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
quantitative testing, all females should be considered as potentially having intermediate G6PD activity and given
the 14-day regimen of primaquine, with counselling on how to recognize symptoms and signs of haemolytic
anaemia. They should be advised to stop primaquine and be told where to seek care should these signs develop.
• If G6PD testing is not available, a decision to prescribe or withhold primaquine should be based on the balance of
the probability and benefits of preventing relapse against the risks of primaquine-induced haemolytic anaemia. This
depends on the population prevalence of G6PD deficiency, the severity of the prevalent genotypes and on the
capacity of health services to identify and manage primaquine-induced haemolytic reactions.
Evidence To Decision
• There are no comparative trials of the efficacy or safety of primaquine in people with G6PD deficiency.
Undesirable effects:
• Primaquine is known to cause haemolysis in people with G6PD deficiency.
• Of the 15 trials included in the systematic review, 12 explicitly excluded people with G6PD deficiency; in three
trials, it was unclear whether participants were tested for G6PD deficiency or excluded. None of the trials
reported serious or treatment-limiting adverse events.
Justification
GRADE
In a systematic review of primaquine for radical cure of P. vivax malaria [231], 14 days of primaquine was compared with
placebo or no treatment in 10 trials, and 14 days was compared with 7 days in one trial. The trials were conducted in
Colombia, Ethiopia, India, Pakistan and Thailand between 1992 and 2006.
14 days of primaquine (0.25 mg/kg bw per day) reduced relapses during 15 months of follow-up by about 40% (RR, 0.60;
95% CI, 0.48–0.75, 10 trials, 1740 participants, high-quality evidence).
14 days of primaquine (0.25 mg/kg bw per day) reduced relapses during 6 months of follow-up by over 50% (RR, 0.45;
95% CI, 0.25–0.81, one trial, 126 participants, low-quality evidence).
No direct comparison has been made of higher doses (0.5 mg/kg bw for 14 days) with the standard regimen (0.25 mg/kg
bw for 14 days).
Twelve of the 15 trials included in the review explicitly excluded people with G6PD deficiency; the remaining three did
not report on this aspect. No serious adverse events were reported.
Other considerations
In the absence of evidence to recommend alternatives, the guideline development group considers 0.75 mg/kg bw
primaquine given once weekly for 8 weeks to be the safest regimen for people with mild-to-moderate G6PD deficiency.
186 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
G6PD deficiency varies, but in tropical areas it is typically 3–35%; high frequencies are found only in areas where malaria
is or has been endemic. There are many (> 180) different G6PD deficiency genetic variants; nearly all of which make the
red cells susceptible to oxidant haemolysis, but the severity of haemolysis may vary. Primaquine generates reactive
intermediate metabolites that are oxidant and cause variable haemolysis in G6PD-deficient individuals. It also causes
methemoglobinaemia. The severity of haemolytic anaemia depends on the dose of primaquine and on the variant of the
G6PD enzyme. Fortunately, primaquine is eliminated rapidly so haemolysis is self-limiting once the drug is stopped. In
the absence of exposure to primaquine or another oxidant agent, G6PD deficiency rarely causes clinical manifestations
so, many patients are unaware of their G6PD status. Screening for G6PD deficiency is not widely available outside
hospitals, but rapid screening tests that can be used at points of care have recently become commercially available.
Remarks
Primaquine is contraindicated in pregnancy and lactation, unless the infant has been tested for G6PD deficiency. It could
be given to women once they have delivered and ceased breastfeeding.
When G6PD status is unknown and G6PD testing is not available, a decision to prescribe primaquine should be based
on an assessment of the risks and benefits of adding primaquine.
Justification
If G6PD testing is not available, a decision to prescribe or withhold primaquine should be based on the balance of the
probability and benefits of preventing relapse against the risks of primaquine-induced haemolytic anaemia. This depends
on the population prevalence of G6PD deficiency, the severity of the prevalent genotypes and on the capacity of health
services to identify and manage primaquine-induced haemolytic reactions.
In women who are pregnant or breastfeeding, weekly chemoprophylaxis with chloroquine can be given until delivery
and breastfeeding are completed, then, on the basis of G6PD status, primaquine can be given to prevent future relapse.
Practical Info
Primaquine is contraindicated in pregnant women and in lactating women (unless the infant is known not to be G6PD
deficient).
As an alternative, chloroquine prophylaxis could be given to suppress relapses after acute vivax malaria during
pregnancy. Once the infant has been delivered and the mother has completed breastfeeding, primaquine could then be
given to achieve radical cure.
Few data are available on the safety of primaquine in infancy, and in the past primaquine was not recommended for
infants. There is, however, no specific reason why primaquine should not be given to children aged 6 months to 1 year
(provided they do not have G6PD deficiency), as this age group may suffer multiple relapses from vivax malaria. The
guideline development group therefore recommended lowering the age restriction to 6 months.
187 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Evidence To Decision
• Chloroquine prophylaxis reduced recurrent P. vivax malaria in pregnant women (moderate-quality evidence).
Justification
GRADE
In a systematic review of malaria chemoprophylaxis in pregnant women [232], chloroquine prophylaxis against P. vivax
during pregnancy was directly evaluated in one trial conducted in Thailand in 2001. In comparison with no
chemoprophylaxis:
Chloroquine prophylaxis substantially reduced recurrent P. vivax malaria (RR, 0.02; 95% CI, 0.00–0.26, one trial, 951
participants, moderate- quality evidence).
Recommendation
Primaquine is contraindicated in pregnant or breastfeeding women with P. vivax malaria. Therefore, consider weekly
chemoprophylaxis with chloroquine until delivery and breastfeeding are completed, then treat with 14 days of
primaquine to prevent future relapse.
188 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
saturation < 92% on room air with a respiratory rate > 30/ in shock. Blood should be taken for cross-matching, a full
min, often with chest indrawing and crepitations on blood count, a platelet count, clotting studies, blood culture
auscultation and full biochemistry (if possible). Careful attention should be
• Significant bleeding: Including recurrent or prolonged paid to the patient’s fluid balance in severe malaria in order to
bleeding from the nose, gums or venepuncture sites; avoid over- or under-hydration. Individual requirements vary
haematemesis or melaena widely and depend on fluid losses before admission.
• Shock: Compensated shock is defined as capillary refill ≥ 3
s or temperature gradient on leg (mid to proximal limb), The differential diagnosis of fever in a severely ill patient is
but no hypotension. Decompensated shock is defined as broad. Coma and fever may be due to meningoencephalitis or
systolic blood pressure < 70 mm Hg in children or < 80 malaria. Cerebral malaria is not associated with signs of
mmHg in adults, with evidence of impaired perfusion (cool meningeal irritation (neck stiffness, photophobia or Kernig’s
peripheries or prolonged capillary refill). sign), but the patient may be opisthotonic. As untreated
• Hyperparasitaemia: P. falciparum parasitaemia > 10% bacterial meningitis is almost invariably fatal, a diagnostic
lumbar puncture should be performed to exclude this
condition. There is also considerable clinical overlap between
Severe vivax and knowlesi malaria: defined as for falciparum septicaemia, pneumonia and severe malaria, and these
malaria but with no parasite density thresholds. conditions may coexist. When possible, blood should always
be taken on admission for bacterial culture. In malaria-endemic
Severe knowlesi malaria is defined as for falciparum malaria
areas, particularly where parasitaemia is common in young age
but with two differences:
groups, it is difficult to rule out septicaemia immediately in a
• P. knowlesi hyperparasitaemia: parasite density > 100 000/ shocked or severely ill obtunded child. In all such cases,
µL empirical parenteral broad-spectrum antibiotics should be
• Jaundice and parasite density > 20 000/µL. started immediately, together with antimalarial treatment.
189 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
190 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
adults, there is a very thin dividing line between over- overlap,particularly in children in areas of moderate and high
hydration, which may produce pulmonary oedema, and under- transmission.Thus broad- spectrum antibiotic treatment should
hydration, which contributes to shock, worsening acidosis and be given with antimalarial drugs to all children with suspected
renal impairment. Careful, frequent evaluation of jugular severe malaria in areas of moderate and high transmission until
venous pressure, peripheral perfusion, venous filling, skin a bacterial infection is excluded. After the start of antimalarial
turgor and urine output should be made. treatment, unexplained deterioration may result from a
supervening bacterial infection.Enteric bacteria (notably
Blood transfusion Salmonella) predominated in many trial series in Africa, but a
Severe malaria is associated with rapid development of variety of bacteria have been cultured from the blood of
anaemia, as infected, once infected and uninfected patients with a diagnosis of severe malaria.
erythrocytes are haemolysed and/or removed from the
circulation by the spleen. Ideally, fresh, cross-matched blood Patients with secondary pneumonia or with clear evidence of
should be transfused; however, in most settings, cross- aspiration should be given empirical treatment with an
matched virus-free blood is in short supply. As for fluid appropriate broad-spectrum antibiotic. In children with
resuscitation, there are not enough studies to make strong persistent fever despite parasite clearance, other possible
evidence-based recommendations on the indications for causes of fever should be excluded, such as systemic
transfusion; the recommendations given here are based on Salmonella infections and urinary tract infections, especially in
expert opinion. In high-transmission settings, blood transfusion catheterized patients. In the majority of cases of persistent
is generally recommended for children with a haemoglobin fever, however, no other pathogen is identified after parasite
level of < 5 g/100 mL (haematocrit < 15%). In low-transmission clearance. Antibiotic treatment should be based on culture and
settings, a threshold of 20% (haemoglobin, 7 g/100 mL) is sensitivity results or,if not available, local antibiotic sensitivity
recommended. These general recommendations must, patterns.
however, be adapted to the individual, as the pathological
consequences of rapid development of anaemia are worse Use of anticonvulsants
than those of chronic or acute anaemia when there has been The treatment of convulsions in cerebral malaria with
adaptation and a compensatory right shift in the oxygen intravenous (or, if this is not possible, rectal) benzodiazepines
dissociation curve. or intramuscular paraldehyde is similar to that for repeated
seizures from any cause. In a large, double-blind, placebo-
Exchange blood transfusion controlled evaluation of a single prophylactic intramuscular
Many anecdotal reports and several series have claimed the injection of 20 mg/kg bw of phenobarbital to children with
benefit of exchange blood transfusion in severe malaria, but cerebral malaria, the frequency of seizures was reduced but
there have been no comparative trials, and there is no the mortality rate was increased significantly. This resulted
consensus on whether it reduces mortality or how it might from respiratory arrest and was associated with additional use
work. Various rationales have been proposed: of benzodiazepine.
• removing infected red blood cells from the circulation and A 20 mg/kg bw dose of phenobarbital should not be given
therefore lowering the parasite burden (although only the without respiratory support. It is not known whether a lower
circulating, relatively non-pathogenic stages are removed, dose would be effective and safer or whether mortality would
and this is also achieved rapidly with artemisinin not increase if ventilation were given. In the absence of further
derivatives); information, prophylactic anticonvulsants are not
• rapidly reducing both the antigen load and the burden of recommended.
parasite-derived toxins, metabolites and toxic mediators
produced by the host; and Treatments that are not recommended
• replacing the rigid unparasitized red cells by more easily In an attempt to reduce the high mortality from severe malaria,
deformable cells, therefore alleviating microcirculatory various adjunctive treatments have been evaluated, but none
obstruction. has proved effective and many have been shown to be
harmful. Heparin, prostacyclin, desferroxamine, pentoxifylline,
low- molecular-mass dextran, urea, high-dose corticosteroids,
aspirin anti-TNF antibody, cyclosporine A,dichloroacetate,
Exchange blood transfusion requires intensive nursing care and
adrenaline, hyperimmune serum,N-acetylcysteine and bolus
a relatively large volume of blood, and it carries significant
administration of albumin are not recommended.In
risks. There is no consensus on the indications, benefits and
addition,use of corticosteroids increases the risk for
dangers involved or on practical details such as the volume of
gastrointestinal bleeding and seizures and has been associated
blood that should be exchanged. It is, therefore, not possible to
with prolonged coma resolution times when compared with
make any recommendation regarding the use of exchange
placebo.
blood transfusion.
Treatment of severe malaria during pregnancy
Concomitant use of antibiotics
Women in the second and third trimesters of pregnancy are
The threshold for administering antibiotic treatment should be
more likely to have severe malaria than other adults, and, in
low in severe malaria. Septicaemia and severe malaria are
low-transmission settings, this is often complicated by
associated, and there is substantial diagnostic
pulmonary oedema and hypoglycaemia. Maternal mortality is
191 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
approximately 50%, which is higher than in non-pregnant Although P. vivax malaria is considered to be benign, with a low
adults. Fetal death and premature labour are common. case-fatality rate, it may cause a debilitating febrile illness with
Parenteral antimalarial drugs should be given to pregnant progressive anaemia and can also occasionally cause severe
women with severe malaria in full doses without delay. disease, as in P. falciparum malaria. Reported manifestations of
Parenteral artesunate is the treatment of choice in all severe P. vivax malaria include severe anaemia,
trimesters. Treatment must not be delayed. If artesunate is thrombocytopenia, acute pulmonary oedema and, less
unavailable, intramuscular artemether should be given, and if commonly, cerebral malaria, pancytopenia, jaundice, splenic
this is unavailable then parenteral quinine should be started rupture, haemoglobinuria, acute renal failure and shock.
immediately until artesunate is obtained.
Prompt effective treatment and case management should be
Obstetric advice should be sought at an early stage, a the same as for severe P. falciparum malaria (see section 5.5.1).
paediatrician alerted and blood glucose checked frequently. Following parenteral artesunate, treatment can be completed
Hypoglycaemia should be expected, and it is often recurrent if with a full treatment course of oral ACT or chloroquine (in
the patient is receiving quinine. Severe malaria may also countries where chloroquine is the treatment of choice). A full
present immediately after delivery. Postpartum bacterial course of radical treatment with primaquine should be given
infection is a common complication and should be managed after recovery.
appropriately.
Please refer to Management of severe malaria - A practical
Treatment of severe P. vivax malaria handbook, 3rd edition [233].
5.2.2.1 Artesunate
Adults and children with severe malaria (including infants, pregnant women in all trimesters and lactating women) should
be treated with intravenous or intramuscular artesunate for at least 24 h and until they can tolerate oral medication.
Once a patient has received at least 24 h of parenteral therapy and can tolerate oral therapy, treatment should be
completed with 3 days of an ACT.
Practical Info
Artesunate is dispensed as a powder of artesunic acid, which is dissolved in sodium bicarbonate (5%) to form sodium
artesunate. The solution is then diluted in approximately 5 mL of 5% dextrose and given by intravenous injection or by
intramuscular injection into the anterior thigh.
The solution should be prepared freshly for each administration and should not be stored. Artesunate is rapidly
hydrolysed in-vivo to dihydroartemisinin, which provides the main antimalarial effect. Studies of the pharmacokinetics of
parenteral artesunate in children with severe malaria suggest that they have less exposure than older children and adults
to both artesunate and the biologically active metabolite dihydroartemisinin. Body weight has been identified as a
significant covariate in studies of the pharmacokinetics of orally and rectally administered artesunate, which suggests
that young children have a larger apparent volume of distribution for both compounds and should therefore receive a
slightly higher dose of parenteral artesunate to achieve exposure comparable to that of older children and adults.
Please refer to the Information note on delayed haemolytic anaemia following treatment with artesunate [235].
192 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Evidence To Decision
• In both adults and children, parenteral artesunate prevented more deaths than parenteral quinine (high-quality
evidence).
• For intravenous administration, artesunate is given as a bolus, whereas quinine requires slow infusion.
• For intramuscular administration, artesunate is given in a smaller volume than quinine.
Undesirable effects:
• Artesunate is associated with a small increase in neurological sequelae at the time of hospital discharge
(moderate-quality evidence). The difference is no longer evident on day 28 after discharge (moderate-quality
evidence).
Justification
GRADE
In a systematic review of artesunate for severe malaria [234], eight randomized controlled trials with a total of 1664
adults and 5765 children, directly compared parenteral artesunate with parenteral quinine. The trials were conducted in
various African and Asian countries between 1989 and 2010.
• reduced mortality from severe malaria by about 40% in adults (RR, 0.61; 95% CI, 0.50–0.75, five trials, 1664
participants, high-quality evidence);
• reduced mortality from severe malaria by about 25% in children (RR, 0.76; 95% CI, 0.65–0.90, four trials, 5765
participants, high-quality evidence); and
• was associated with a small increase in neurological sequelae in children at the time of hospital discharge (RR, 1.36;
95% CI, 1.01–1.83, three trials, 5163 participants, moderate-quality evidence), most of which, however, slowly
resolved, with little or no difference between artesunate and quinine 28 days later (moderate-quality evidence).
Other considerations
The guideline development group considered that the small increase in neurological sequelae at discharge after
treatment with artesunate was due to the delayed recovery of the severely ill patients, who would have died had they
received quinine. This should not be interpreted as a sign of neurotoxicity. Although the safety of artesunate given in the
first trimester of pregnancy has not been firmly established, the guideline development group considered that the
proven benefits to the mother outweigh any potential harm to the developing fetus.
Remarks
Parenteral artesunate is recommended as first-line treatment for adults, children, infants and pregnant women in all
trimesters of pregnancy.
Although the safety of artesunate in the first trimester of pregnancy has not been firmly established, the group
considered that the proven benefits to the mother outweigh the potential harms to the developing fetus.
193 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Children weighing < 20 kg should receive a higher dose of artesunate (3 mg/kg bw per dose) than larger children and
adults (2.4 mg/kg bw per dose) to ensure equivalent exposure to the drug.
*Not evaluated using the GRADE framework; recommendation based on pharmacokinetic modelling
Practical Info
Artesunate is dispensed as a powder of artesunic acid, which is dissolved in sodium bicarbonate (5%) to form sodium
artesunate. The solution is then diluted in approximately 5 mL of 5% dextrose and given by intravenous injection or by
intramuscular injection into the anterior thigh.
The solution should be prepared freshly for each administration and should not be stored. Artesunate is rapidly
hydrolysed in-vivo to dihydroartemisinin, which provides the main antimalarial effect. Studies of the pharmacokinetics of
parenteral artesunate in children with severe malaria suggest that they have less exposure than older children and adults
to both artesunate and the biologically active metabolite dihydroartemisinin. Body weight has been identified as a
significant covariate in studies of the pharmacokinetics of orally and rectally administered artesunate, which suggests
that young children have a larger apparent volume of distribution for both compounds and should therefore receive a
slightly higher dose of parenteral artesunate to achieve exposure comparable to that of older children and adults.
Justification
The dosing subgroup reviewed all available pharmacokinetic data on artesunate and the main biologically active
metabolite dihydroartemisinin following administration of artesunate in severe malaria (published pharmacokinetic
studies from 71 adults and 265 children) [236][237]. Simulations of artesunate and dihydroartemisinin exposures were
conducted for each age group. These showed underexposure in younger children. The revised parenteral dose regimens
are predicted to provide equivalent artesunate and dihydroartemisinin exposures across all age groups.
Other considerations
Individual parenteral artesunate doses between 1.75 and 4 mg/kg have been studied and no toxicity has been observed.
The GRC concluded that the predicted benefits of improved antimalarial exposure in children are not at the expense of
increased risk.
If artesunate is not available, artemether should be used in preference to quinine for treating children and adults with
severe malaria.
194 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Practical Info
Artemether
Artemether is two to three times less active than its main metabolite dihydroartemisinin. Artemether can be given as an
oil-based intramuscular injection or orally. In severe falciparum malaria, the concentration of the parent compound
predominates after intramuscular injection, whereas parenteral artesunate is hydrolysed rapidly and almost completely
to dihydroartemisinin. Given intramuscularly, artemether may be absorbed more slowly and more erratically than water-
soluble artesunate, which is absorbed rapidly and reliably after intramuscular injection. These pharmacological
advantages may explain the clinical superiority of parenteral artesunate over artemether in severe malaria.
Artemether is dispensed dissolved in oil (groundnut, sesame seed) and given by intramuscular injection into the anterior
thigh.
Therapeutic dose: The initial dose of artemether is 3.2 mg/kg bw intramuscularly (to the anterior thigh). The
maintenance dose is 1.6 mg/kg bw intramuscularly daily.
Quinine
Quinine treatment for severe malaria was established before the methods for modern clinical trials were developed.
Several salts of quinine have been formulated for parenteral use, but the dihydrochloride is the most widely used. The
peak concentrations after intramuscular quinine in severe malaria are similar to those after intravenous infusion. Studies
of pharmacokinetics show that a loading dose of quinine (20 mg salt/kg bw, twice the maintenance dose) provides
therapeutic plasma concentrations within 4 h. The maintenance dose of quinine (10 mg salt/ kg bw) is administered at
8-h intervals, starting 8 h after the first dose. If there is no improvement in the patient’s condition within 48 h, the dose
should be reduced by one third, i.e. to 10 mg salt/kg bw every 12 h.
Rapid intravenous administration of quinine is dangerous. Each dose of parenteral quinine must be administered as a
slow, rate-controlled infusion (usually diluted in 5% dextrose and infused over 4 h). The infusion rate should not exceed 5
mg salt/kg bw per h.
Whereas many antimalarial drugs are prescribed in terms of base, for historical reasons quinine doses are usually
recommended in terms of salt (usually sulphate for oral use and dihydrochloride for parenteral use). Recommendations
for the doses of this and other antimalarial agents should state clearly whether the salt or the base is being referred to;
doses with different salts must have the same base equivalents. Quinine must never be given by intravenous bolus
injection, as lethal hypotension may result.
Quinine dihydrochloride should be given by rate-controlled infusion in saline or dextrose solution. If this is not possible,
it should be given by intramuscular injection to the anterior thigh; quinine should not be injected into the buttock in
order to avoid sciatic nerve injury. The first dose should be split, with 10 mg/kg bw into each thigh. Undiluted quinine
dihydrochloride at a concentration of 300 mg/ mL is acidic (pH 2) and painful when given by intramuscular injection, so it
is best to administer it either in a buffered formulation or diluted to a concentration of 60–100 mg/mL for intramuscular
injection. Gluconate salts are less acidic and better tolerated than the dihydrochloride salt when given by the
intramuscular and rectal routes.
As the first (loading) dose is the most important in the treatment of severe malaria, it should be reduced only if there is
clear evidence of adequate pre-treatment before presentation. Although quinine can cause hypotension if administered
rapidly, and overdose is associated with blindness and deafness, these adverse effects are rare in the treatment of severe
malaria. The dangers of insufficient treatment (i.e. death from malaria) exceed those of excessive initial treatment.
Evidence To Decision
Desirable effects:
• In children > 12 years and adults, parenteral artesunate probably prevents more deaths than intramuscular
artemether (moderate-quality evidence).
• No randomized controlled trials have been conducted in children aged ≤ 12 years.
--
195 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Is intramuscular artemether superior to parenteral quinine in preventing death from severe malaria?
Desirable effects:
• In children, artemether is probably equivalent to quinine in preventing death (moderate-quality evidence).
• In children > 5 years and adults, artemether may be superior to quinine (moderate-quality evidence).
• Artemether is easier to administer, requiring a smaller fluid volume for intramuscular injection.
Is parenteral artesunate superior to parenteral quinine in preventing death from severe malaria?
--
Is intramuscular artemether superior to parenteral quinine in preventing death from severe malaria?
Justification
GRADE
A systematic review of intramuscular artemether for severe malaria comprised two randomized controlled trials in Viet
Nam in which artemether was compared with artesunate in 494 adults, and 16 trials in Africa and Asia in which
artemether was compared with quinine in 716 adults and 1447 children [238]. The trials were conducted between 1991
and 2009.
In comparison with artesunate, intramuscular artemether was not as effective at preventing deaths in adults in Asia (RR,
1.80; 95% CI, 1.09–2.97; two trials, 494 participants, moderate-quality evidence).
Artemether and artesunate have not been directly compared in randomized trials in African children.
• Intramuscular artemether prevented a similar number of deaths in children in Africa (RR, 0.96; 95% CI, 0.76–1.20;
12 trials, 1447 participants, moderate-quality evidence).
• Intramuscular artemether prevented more deaths in adults in Asia (RR, 0.59; 95% CI, 0.42–0.83; four trials, 716
participants, moderate-quality evidence).
Other considerations
Indirect comparisons of parenteral artesunate and quinine and of artemether and quinine were considered by the
guideline development group with what is known about the pharmacokinetics of the two drugs. They judged the
accumulated indirect evidence to be sufficient to recommend parenteral artesunate rather than intramuscular
artemether for use in all age groups.
--
Is parenteral artesunate superior to parenteral quinine in preventing death from severe malaria?
Remarks
Intramuscular artemether should be considered only when parenteral artesunate is not available.
Recommendation
Treat children and adults with severe malaria with parenteral artesunate for at least 24 h.
196 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
--
Is intramuscular artemether superior to parenteral quinine in preventing death from severe malaria?
Remarks
Quinine is retained as an option for treating severe malaria when artesunate or artemether is not available or is
contraindicated.
Recommendation
If parenteral artesunate is not available, use artemether in preference to quinine for treating children and adults with
severe malaria.
Where complete treatment of severe malaria is not possible, but injections are available, adults and children should be
given a single intramuscular dose of artesunate, and referred to an appropriate facility for further care. Where
intramuscular artesunate is not available, intramuscular artemether or, if that is not available, intramuscular quinine
should be used.
Where intramuscular injection of artesunate is not available, children < 6 years should be treated with a single rectal
dose (10mg/kg bw) of artesunate, and referred immediately to an appropriate facility for further care. Rectal artesunate
should not be used in older children and adults.
197 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Practical Info
Adjustment of parenteral dosing in renal failure of hepatic dysfunction
The dosage of artemisinin derivatives does not have to be adjusted for patients with vital organ dysfunction. However,
quinine accumulates in severe vital organ dysfunction. If a patient with severe malaria has persisting acute kidney injury
or there is no clinical improvement by 48 h, the dose of quinine should be reduced by one third, to 10 mg salt/kg bw
every 12 h. Dosage adjustments are not necessary if patients are receiving either haemodialysis or haemofiltration.
Follow-on treatment
The current recommendation of experts is to give parenteral antimalarial drugs for the treatment of severe malaria for a
minimum of 24 h ounce started (irrespective of the patient’s ability to tolerate oral medication earlier) or until the patient
can tolerate oral medication, before giving the oral follow-up treatment.
After initial parenteral treatment, once the patient can tolerate oral therapy, it is essential to continue and complete
treatment with an effective oral antimalarial drug by giving a full course of effective ACT (artesunate + amodiaquine,
artemether + lumefantrine or dihydroartemisinin + piperaquine). If the patient presented initially with impaired
consciousness, ACTs containing mefloquine should be avoided because of an increased incidence of neuropsychiatric
complications. When an ACT is not available, artesunate + clindamycin, artesunate + doxycycline, quinine + clindamycin
or quinine + doxycycline can be used for follow-on treatment. Doxycycline is preferred to other tetracyclines because it
can be given once daily and does not accumulate in cases of renal failure, but it should not be given to children < 8 years
or pregnant women. As treatment with doxycycline is begun only when the patient has recovered sufficiently, the 7-day
doxycycline course finishes after the artesunate, artemether or quinine course. When available, clindamycin may be
substituted in children and pregnant women.
Please refer to Rectal artesunate for pre-referral treatment of severe malaria [240].
Evidence To Decision
• No studies of direct comparison of rectal artesunate with parenteral antimalarial drugs for pre-referral
treatment.
• In hospital care, parenteral artesunate reduces the number of deaths to a greater extent than parenteral quinine
(high-quality evidence) and probably reduces the number of deaths from that with intramuscular artemether
(moderate-quality evidence).
Justification
GRADE
In a systematic review of pre-referral treatment for suspected severe malaria, in a single large randomized controlled trial
of 17 826 children and adults in Bangladesh, Ghana and the United Republic of Tanzania, pre-referral rectal artesunate
was compared with placebo [239].
• Rectal artesunate reduced mortality by about 25% in children < 6 years (RR, 0.74; 95% CI, 0.59–0.93; one trial,
8050 participants, moderate- quality evidence).
• Rectal artesunate was associated with more deaths in older children and adults (RR, 2.21; 95% CI, 1.18–4.15; one
trial 4018 participants, low- quality evidence).
198 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Other considerations
The guideline development group could find no plausible explanation for the finding of increased mortality among older
children and adults in Asia who received rectal artesunate, which may be due to chance. Further trials would provide
clarification but are unlikely to be done. The group was therefore unable to recommend its use in older children and
adults.
In the absence of direct evaluations of parenteral antimalarial drugs for pre- referral treatment, the guideline
development group considered the known benefits of artesunate in hospitalized patients and downgraded the quality of
evidence for pre-referral situations. When intramuscular injections can be given, the group recommends intramuscular
artesunate in preference to rectal artesunate.
Remarks
This recommendation applies to all people with suspected severe malaria, including infants, lactating women and
pregnant women in all trimesters.
Where intramuscular artesunate is not available, use rectal artesunate (in children < 6 years), intramuscular artemether or
intramuscular quinine.
Parasitological diagnosis during epidemics Epidemics of mixed falciparum and vivax or vivax malaria
In the acute phase of epidemics and complex emergency ACTs (except artesunate + SP) should be used to treat
situations, facilities for laboratory diagnosis with good- uncomplicated malaria in mixed-infection epidemics, as they
quality equipment and reagents and skilled technicians are are highly effective against all malaria species. In areas with
often not available or are overwhelmed. Attempts should be pure P. vivax epidemics, ACTs or chloroquine (if prevalent
made to improve diagnostic capacity rapidly, including strains are sensitive) should be used.
provision of RDTs. If diagnostic testing is not feasible, the
most practical approach is to treat all febrile patients as Anti-relapse therapy for P. vivax malaria
suspected malaria cases, with the inevitable consequences of
Administration of 14-day primaquine anti-relapse therapy for
over-treatment of malaria and potentially poor management
vivax malaria may be impractical in epidemic situations
of other febrile conditions. If this approach is used, it is
because of the duration of treatment and the difficulty of
imperative to monitor intermittently the prevalence of
199 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
ensuring adherence. If adequate records are kept, therapy drugs is that they kill circulating ring-stage parasites and thus
can be given in the post-epidemic period to patients who accelerate therapeutic responses. This is lost in resistance to
have been treated with blood schizontocides. artemisinin. As a consequence, parasite clearance is slowed,
and ACT failure rates and gametocytaemia both increase.
Malaria elimination settings The reduced efficacy of artemisinin places greater selective
pressure on the partner drugs, to which resistance is also
Use of gametocytocidal drugs to reduce transmission
increasing. This situation poses a grave threat. In the past
ACT reduces P. falciparum gametocyte carriage and
chloroquine resistant parasites emerged near the
transmission markedly, but this effect is incomplete, and
Cambodia–Thailand border and then spread throughout Asia
patients presenting with gametocytaemia may be infectious
and Africa at a cost of millions of lives. In Cambodia, where
for days or occasionally weeks, despite ACT. The strategy
artemisinin resistance is worst, none of the currently
of using a single dose of primaquine to reduce infectivity and
recommended treatment regimens provides acceptable cure
thus P. falciparum transmission has been widely used in low
rates (> 90%), and continued use of ineffective drug
transmission settings.
regimens fuels the spread of resistance. In Cambodia use of
Use of primaquine as a P. falciparum gametocytocide has a atovaquone–proguanil instead of ACT resulted in very rapid
particular role in programmes to eliminate P. falciparum emergence of resistance to atovaquone.
malaria. The population benefits of reducing malaria
In this dangerous, rapidly changing situation, local treatment
transmission by
guidelines cannot be based on a solid evidence base;
gametocytocidal drugs require that a high proportion of
however, the risks associated with continued use of
patients receive these medicines. WHO recommends the
ineffective regimens are likely to exceed the risks of new,
addition of a single dose of primaquine (0.25 mg base/kg
untried regimens with generally safe antimalarial drugs. At
bw) to ACT for uncomplicated falciparum malaria as a
the current levels of resistance, the artemisinin derivatives
gametocytocidal medicine, particularly as a component of
still provide significant antimalarial activity; therefore, longer
elimination programmes. A recent review of the evidence on
courses of treatment with existing or new augmented
the safety and effectiveness of primaquine as a
combinations or treatment with new partner medicines (e.g.
gametocytocide of P. falciparum indicates that a single dose
artesunate + pyronaridine) may be effective. Studies to
of 0.25 mg base/kg bw is effective in blocking infectivity to
determine the best treatments for artemisinin-resistant
mosquitos and is unlikely to cause serious toxicity in people
malaria are needed urgently.
with any of the G6PD variants. Thus, the G6PD status of the
patient does not have to be known before primaquine is It is strongly recommended that single-dose primaquine (as a
used for this indication. gametocytocide) be added to all falciparum malaria
treatment regimens as described in section 5.2.5. For the
Artemisinin-resistant falciparum malaria
treatment of severe malaria in areas with established
Artemisinin resistance in P. falciparum is now prevalent in
artemisinin resistance, it is recommended that parenteral
parts of Cambodia, the Lao People’s Democratic Republic,
artesunate and parenteral quinine be given together in full
Myanmar, Thailand and Viet Nam. There is currently no
doses, as described in section 5.5.
evidence for artemisinin resistance outside these areas. The
particular advantage of artemisinins over other antimalarial
200 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
inadequately absorbed. Antimalarial medicines must be agencies, customs and excise authorities and medicines
manufactured according to good manufacturing practice, regulatory agencies to deal more effectively with falsified
have the correct drug and excipient contents, be proved to medicines. Private sector drug distribution outlets should
have bioavailability that is similar to that of the reference have more information and active engagement with
product, have been stored under appropriate conditions and regulatory agencies. WHO, in collaboration with other
be dispensed before their expiry date. United Nations agencies, has established an international
mechanism to prequalify manufacturers of ACTs on the basis
Tools to assess drug quality at points of sale are being of their compliance with internationally recommended
developed, but the capacity of medicines regulatory agencies standards of manufacture and quality. Manufacturers of
in most countries to monitor drug quality is still limited. Legal antimalarial medicines with prequalified status are listed on
and regulatory frameworks must be strengthened, and there the prequalification web site [241].
should be greater collaboration between law enforcement
National drug and regulatory authorities should ensure that the antimalarial medicines provided in both the public and
the private sectors are of acceptable quality, through regulation, inspection and law enforcement.
201 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• confirmation of the quality of the antimalarial medicines safety profiles of the currently recommended antimalarial
tested; drugs are reasonably well described and supported by an
• molecular genotyping to distinguish between re- evidence base of several thousand participants (mainly from
infections and recrudescence and to identify genetic clinical trials); however, rare but serious adverse drug
markers of drug resistance; reactions will not be detected in clinical trials of this size,
• studies of parasite susceptibility to antimalarial drugs in particularly if they occur primarily in young children,
culture; and pregnant women or people with concurrent illness, who are
• measurement of antimalarial drug levels to assess usually under-represented in clinical trials. Rare but serious
exposure in cases of slow therapeutic response or adverse drug reactions are therefore detected only in
treatment failure prospective phase IV post-marketing studies or population-
based pharmacovigilance systems. In particular, more data
are urgently needed on the safety of ACTs during the first
Pharmacovigilance
trimester of pregnancy and on potential interactions
Governments should have effective pharmacovigilance
between antimalarial and other commonly used medicines.
systems (such as the WHO pregnancy registry) to monitor
the safety of all drugs, including antimalarial medicines. The
All malaria programmes should regularly monitor the therapeutic efficacy of antimalarial drugs using the standard WHO
protocols.
Practical Info
Routine monitoring of antimalarial drug efficacy is necessary to ensure effective case management and for early
detection of resistance. WHO recommends that the efficacy of first- and second-line antimalarial treatments be tested
at least once every 24 months at all sentinel sites. Data collected from studies conducted according to the standard
protocol inform national treatment policies.
Please refer to the tools for monitoring antimalarial drug efficacy and Methods for surveillance of antimalarial drug
efficacy [242] which includes tools and materials to conduct routine therapeutic efficacy studies (TES). It is a reference
for national programmes and investigators conducting routine surveillance studies to assess the efficacy of medicines
that have already been registered.
• Methods and techniques for clinical trials on antimalarial drug efficacy: Genotyping to identify parasite
populations [243]
• Report on antimalarial drug efficacy, resistance and response: 10 years of surveillance (2010-2019) [244]
202 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
assessment. Decisions about coverage, feasibility, acceptability patients (such as parasite count and stage of parasite
and cost may require input from various health professionals, development and intra-leukocyte pigment). In severe malaria, an
community representatives, health economists, academics and RDT can be used to confirm malaria rapidly so that parenteral
health system managers. antimalarial treatment can be started immediately. Where
possible, however, blood smears should be examined by
Opportunities and risks microscopy, with frequent monitoring of parasitaemia (e.g. every
The recommendations made in these guidelines provide an 12 h) during the first 2–3 days of treatment in order to monitor
opportunity to improve malaria case management further, to the response.
reduce unnecessary morbidity and mortality and to contribute to
continued efforts towards elimination. Failure to implement the Choosing ACT
basic principles of combination therapy and rational use of In the absence of resistance, all the recommended ACTs have
antimalarial medicines will risk promoting the emergence and been shown to result in parasitological cure rates of > 95%.
spread of drug resistance, which could undo all the recent gains Although there are minor differences in the oral absorption,
in malaria control and elimination. bioavailability and tolerability of the different artemisinin
derivatives, there is no evidence that these differences are
General guiding principles for choosing a case management clinically significant in currently available formulations. It is the
strategy and tools properties of the partner medicine and the level of resistance to
it that determine the efficacy of a formulation.
Choosing a diagnostic strategy
The two methods currently considered suitable for routine Policy-makers should also consider:
patient management are light microscopy and RDTs. Different
strategies may be adopted in different health care settings. The • local data on the therapeutic efficacy of the ACT,
choice between RDTs and microscopy depends on local • local data on drug resistance,
circumstances, including the skills available, the patient case- • the adverse effect profiles of ACT partner drugs,
load, the epidemiology of malaria and use of microscopy for the • the availability of appropriate formulations to ensure
diagnosis of other diseases. When the case-load of patients with adherence,
fever is high, the cost of each microscopy test is likely to be less • cost.
than that of an RDT; however, high-throughput, high-quality
microscopy may be less operationally feasible. Although several
In parts of South-East Asia, artemisinin resistance is
RDTs allow diagnosis of both P. falciparum and P. vivax infections,
compromising the efficacy of ACTs and placing greater selection
microscopy has further advantages, including accurate parasite
pressure on resistance to the partner medicines. Elsewhere,
counting (and thus identification of high parasite density),
there is no convincing evidence for reduced susceptibility to the
prognostication in severe malaria, speciation of other malaria
artemisinins; therefore, the performance of the partner drugs is
parasites and sequential assessment of the response to
the determining factor in the choice of ACT, and the following
antimalarial treatment. Microscopy may help to identify other
principles apply:
causes of fever. High-quality light microscopy requires well-
trained, skilled staff, good staining reagents, clean slides and, • Resistance to mefloquine has been found in parts of
often, electricity to power the microscope. It requires a quality mainland South-East Asia where this drug has been used
assurance system, which is often not well implemented in intensively. Nevertheless, the combination with artesunate
malaria-endemic countries. is very effective, unless there is also resistance to
artemisinin. Resistance to both components has
In many areas, malaria patients are treated outside the formal
compromised the efficacy of artesunate + mefloquine in
health services, e.g. in the community, at home or by private
western Cambodia, eastern Myanmar and eastern Thailand.
providers. Microscopy is generally not feasible in the community,
• Lumefantrine shares some cross-resistance with
but RDTs might be available, allowing access to confirmatory
mefloquine, but this has not compromised its efficacy in any
diagnosis of malaria and the correct management of febrile
of the areas in which artemether + lumefantrine has been
illnesses. The average sensitivity of HRP2-detecting RDTs is
used outside South-East Asia.
generally greater than that of RDTs for detecting pLDH of P.
• Until recently, there was no evidence of resistance to
falciparum, but the latter are slightly more specific because the
piperaquine anywhere, but there is now reduced
HRP2 antigen may persist in blood for days or weeks after
susceptibility in western Cambodia. Elsewhere, the
effective treatment. HRP2-detecting RDTs are not suitable for
dihydroartemisinin + piperaquine combination is highly
detecting treatment failure. RDTs are slightly less sensitive for
effective.
detecting P. malariae and P. ovale. The WHO Malaria RDT
• Resistance to SP limits its use in combination with
Product Testing programme provides comparative data on the
artesunate to the few areas in which susceptibility is
performance of RDT products to guide procurement. Since
retained.
2008, 210 products have been evaluated in five rounds of
• Amodiaquine remains effective in combination with
product testing [172].
artesunate in parts of Africa and the Americas, although
For the diagnosis of severe malaria, microscopy is preferred, as it elsewhere resistance to this drug was prevalent before its
provides a diagnosis of malaria and assessment of other introduction in an ACT.
important parameters of prognostic relevance in severely ill
203 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Considerations in use of artemisinin-based combination therapy Other operational issues in managing effective treatment
Oral artemisinin and its derivatives (e.g. artesunate, artemether, Individual patients derive the maximum benefit from an ACT if
dihydroartemisinin) should not be used alone. In order to simplify they can access it within 24–48 h of the onset of malaria
use, improve adherence and minimize the availability of oral symptoms. The impact in reducing transmission at a population
artemisinin monotherapy, fixed-dose combination ACTs are level depends on high coverage rates and the transmission
strongly preferred to co-blistered or co-dispensed loose tablets intensity. Thus, to optimize the benefits of deploying ACTs, they
and should be used when they are readily available. Fixed-dose should be available in the public health delivery system, the
combinations of all recommended ACT are now available, except private sector and the community, with no financial or physical
artesunate + SP. Fixed-dose artesunate + amodiaquine performs barrier to access. A strategy for ensuring full access (including
better than loose tablets, presumably by ensuring adequate community management of malaria in the context of integrated
dosing. Unfortunately, paediatric formulations are not yet case management) must be based on analyses of national and
available for all ACTs. local health systems and may require legislative changes and
regulatory approval, with additional local adjustment as indicated
The choice of ACT in a country or region should be based on by programme monitoring and operational research. To optimize
optimal efficacy and adherence, which can be achieved by: the benefits of effective treatment, wide dissemination of
national treatment guidelines, clear recommendations,
• minimizing the number of formulations available for each
appropriate information, education and communication
recommended treatment regimen
materials, monitoring of the deployment process, access and
• using, where available, solid formulations instead of liquid
coverage, and provision of adequately packaged antimalarial
formulations, even for young patients.
drugs are needed.
204 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The choice of ACTs in a country or region should be based on optimal efficacy, safety and adherence.
Practical Info
Pharmacovigilance is the practice of monitoring the effects of medical drugs after they have been licensed for use, especially to
identify and evaluate previously unreported adverse reactions. A practical handbook on the pharmacovigilance of antimalarial
medicines [246] provides a step-by-step approach for antimalarial pharmacovigilance. Designed for health officials, planners, and
other health workers, it focuses on active and passive pharmacovigilance, reporting, event monitoring and other key factors.
Drugs used as first line treatment should not be used in IPTp, PMC, SMC, IPTsc or MDA.
When possible:
fixed-dose combinations should be used rather than co-blistered or loose, single-agent formulations; andfor young children and
infants, paediatric formulations, with a preference for solid formulations (e.g. dispersible tablets) should be used rather than
liquid formulations.
205 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
intervention on transmission, key outcomes are measured at the activities during the post-elimination period will depend on the
community level rather than only among those who actually health system and the malariogenic potential of the area, that is,
receive or participate in the intervention. the degree of receptivity to transmission and the risk or rate of
importation of malaria infections. Strategies targeted to specific
In post-elimination settings, malaria programmes must continue to higher-risk areas or groups, or in response to the identification of
actively intervene in order to prevent re-establishment of an imported or introduced infection, are required in post-
transmission. Countries will need to ensure that diagnosis and elimination settings working to prevent re-establishment of
treatment services are available everywhere as part of universal transmission.
health coverage as imported cases can be identified anywhere and
at any time. However, the extent and intensity of additional
Mass testing and treatment (MTaT) to reduce the transmission of malaria is not recommended.
The GDG noted that there may be exceptional circumstances under which MTaT might be appropriate, such as a transmission focus
in a very low transmission or post-elimination setting where MDA is not an acceptable or feasible strategy.
Evidence To Decision
Beneficial outcomes
• MTaT does not reduce the prevalence of malaria two months after the last round (RD: -26 per 1000 population;
95% CI [CI] -68 to 15 per 100 persons; one cRCT; high-certainty evidence).
• MTaT does not reduce the incidence of malaria 0–12 months after the start of the intervention (RD: -117 per 1000
p-y (p-y); 95% CI: -303 to 93 per 1000 p-y; one cRCT; high-certainty evidence).
• MTaT probably results in little to no difference in the incidence of malaria (measured only in children) 6–12 months
206 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
after the start of the intervention (RD: 4 per 1000 p-y; 95% CI: -2 to 8 per 1000 p-y; two cRCTs; moderate-
certainty evidence).
• MTaT reduces the incidence of clinical malaria 0–12 months after the start of the intervention (RD: -44 per 1000 p-
y; 95% CI: -70 to -12 per 1000 p-y; two cRCTs; high-certainty evidence).
Adverse events
• Among people treated as part of MTaT, the most common adverse events were fever (0.023/person-day), headache
(0.008/person-day, vomiting (0.006/person-day), cough (0.004/person-day), shivering (0.003/person-day) and
nasal congestion (0.002/person-day) (one cRCT, not GRADEd because no information was available from the
comparator arm).
The GDG judged that the beneficial impact of MTaT on malaria incidence and prevalence at the community level was
trivial, as were the potential adverse events.
The GDG judged that there may be important uncertainty or variability in preferences or values that could not be
determined due to the lack of studies.
Resources
The systematic review identified two studies on the cost and cost effectiveness of MTaT in southern Zambia (Bhamani et
al unpublished evidence). The overall cost per test administered was US$ 4.39, whereas the overall cost for treatment
with artemether-lumefantrine (AL) was US$ 34.74. Personnel and vehicles were the largest cost drivers, followed by
trainings and rapid diagnostic tests. The estimated cost per DALYs averted was US$ 804, which in the context of Zambia
was considered a highly cost-effective health intervention.
The GDG judged the resources required to implement MTaT to be large. Although one study found MTaT to be a cost-
effective intervention in the context of southern Zambia, the GDG judged the impact of the intervention in general to
be likely trivial. Therefore, with high costs, the cost-effectiveness would probably favour not conducting MTaT.
Equity
No studies were identified that addressed the issue of whether MTaT increased or decreased health equity.
The GDG felt that MTaT may favour disadvantaged segments of the population who otherwise might have limited or no
access to the health system for diagnostic testing and treatment for malaria. Therefore, the GDG judged that MTaT
would probably increase health equity.
207 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Acceptability
The acceptability of MTaT was reported in three qualitative studies identified by the systematic review (Bhamani et al
unpublished evidence). One study in western Kenya found that the community engaged in an MTaT intervention reported
concerns over testing in the absence of symptoms. These concerns were mostly related to the fear of covert HIV testing
and some lack of understanding of the possibility of asymptomatic malaria. Other issues related to acceptability were
failure to adhere to the full treatment course, treatment effectiveness and the need for intense sensitization activities. In
the post-implementation round, although many participants appreciated the intervention and expressed an overall
positive experience, some concerns remained, including fear of covert HIV testing and failure to adhere to treatment.
One study in Zambia aimed to understand perceptions of community health workers and community members on MTaT.
In general, MTaT was perceived very positively by most community health workers and community members. However,
some barriers identified by community health workers included difficult transportation to hard-to-reach areas; difficulty
charging personal digital assistants for data collection due to unavailability of charging sources; and commodity
shortages. Among community participants, most barriers were related to the perceived fears around covert HIV testing
and use of blood samples for “Satanism”. Lack of community health worker skills and training to conduct testing and
treatment was also a perceived barrier among some community members. Lastly, this study also identified the perceived
feeling of wellness once symptoms subsided as a barrier to adherence to treatment. One study in Ghana assessed the
perception of health workers and community members on MTaT. Overall, the health workers and community
participants perceived MTaT as a feasible intervention with many benefits, including reducing incidence in children,
increasing sensitization of the community on malaria, reducing hospital admissions, increasing work productivity,
reducing expenditure for treatment, providing timely access to treatment at home, and reducing travel to health
facilities. However, health care workers were concerned about revenue lost from internally-generated funds at the
health facility. Some of the challenges experienced during MTaT were misconceptions and rumours (e.g. fear of being
infected with epilepsy by health workers), concerns over the safety of drugs, and a lack of trust in health workers’ skills
and knowledge.
The GDG judged that MTaT was probably acceptable to key stakeholders.
Feasibility
The systematic review identified two studies reporting on the feasibility of MTaT campaigns in Kenya and Ghana
(Bhamani et al unpublished evidence). However, one MTaT campaign was implemented within a well developed and well
maintained health and demographic surveillance system in Kenya. The other study from Ghana reported on the
perception of MTaT as feasible by health workers and community members.
The GDG noted that the type of parasitological test used (rapid diagnostic test, microscopy or nucleic acid based test)
would affect the feasibility of implementing the strategy as tests that are not conducted at the point-of-contact would
be more difficult to implement, require more staff with more technical training and likely delay identification and
treatment of positive cases.
The feasibility of implementing MTaT would also depend on whether radical cure of P. vivax using an 8-aminoquinoline
medicine was part of the MTaT strategy, which would necessitate testing for G6PD deficiency, an effective
pharmacovigilance system and emergency access to blood transfusion services.
The GDG judged that MTaT was probably a feasible intervention to implement.
Justification
The GDG judged that there was moderate certainty evidence that MTaT had a trivial impact on malaria prevalence and
incidence. Although there may be some benefit to health equity by reaching people who may otherwise have difficulty
accessing malaria diagnostic and testing services, and the intervention was found to be acceptable to stakeholders and
feasible to implement, the resources required to implement MTaT were considered to be large. The GDG felt that there may
be transmission foci in very low transmission settings where an MTaT intervention could be beneficial but decided to provide
a conditional recommendation against implementing MTaT to reduce the transmission of malaria.
Research Needs
Further evidence is needed on the impact (prevalence and incidence of malaria infection at the community level) of MTaT
when rounds are conducted at more frequent intervals (at least once per month while there is transmission of malaria). This
research should include evaluation of the feasibility of implementation and acceptability of the strategy to health care
workers and community members. Data on the cost of the strategy and the cost-effectiveness compared to passive
208 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
209 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
In areas with very low to low transmission or post-elimination settings preventing re-establishment of transmission,
antimalarial medicine can be given as chemoprevention to people with increased risk of infection relative to the general
population to reduce transmission.
• Persons given antimalarials should be those with increased risk of infection compared to the general population and their
infections should constitute a large proportion of the parasite reservoir in the area.
• The factors identifying individuals or groups at increased risk of infection should be easy to recognise, thereby improving the
acceptability and feasibility of the intervention.
• Programmes considering implementing targeted drug administration for P. vivax should carefully consider how to safely and
feasibly administer treatment to prevent relapses.
• Care should be taken to avoid stigmatizing groups at increased risk of infection.
• Additional complementary strategies to eliminate or prevent re-establishment of malaria transmission should be in place.
Practical Info
TDA depends on detailed, recent knowledge of the epidemiology and ecology of malaria in an area. This knowledge is
generally based on a strong passive surveillance system that can detect all suspected cases, diagnose infections, collect and
analyse case-based data and characterize cases according to potential risk factors. (The ability to conduct case investigations
at the home of the person diagnosed with malaria is not a requirement for a TDA programme but could potentially improve
the quality of the data collected.)
The persons given antimalarials in a TDA programme should be those with an increased risk of infection compared to the
general population. This could include individuals in key demographic groups or with certain occupations or behaviours that
are known to be associated with increased infection rates. Additionally, data from the surveillance system should
demonstrate that infections in these individuals are likely to comprise a large proportion of the infectious reservoir in the
area. Finally, the characteristics or risk factors that define the group at increased risk of infection should be easily
recognizable or identifiable; if not, the TDA programme will be more challenging to implement and possibly less acceptable
to stakeholders.
Malaria elimination programmes implementing TDA should recognize that, as areas approach elimination, malaria infections
become more concentrated in certain geographies and populations that may already be socially disadvantaged. This includes
migrants, displaced persons, ethnic minorities and poor rural communities. A TDA programme should actively seek to
prevent further adverse social impact on these groups. Language choices can frame the way that groups are perceived, and
TDA programmes should avoid labelling groups of people as “reservoirs" of infection or “hot” populations. Referring to
chemoprevention for malaria in higher-risk “situations” rather than higher-risk “groups” can shift the focus away from
scapegoating certain populations. By engaging communities affected by malaria in elimination settings, including those that
may be socially marginalized, malaria elimination programmes can improve their understanding of local social dynamics and
identify strategies to provide better services to people at risk of malaria infection. TDA programmes should monitor the
social impact of their interventions to determine if stigma is occurring to any malaria-affected populations and to determine
whether their efforts to avoid stigma are working.
Achieving high coverage of the affected population and good adherence to the antimalarial medicine are critical aspects of
TDA programmes. TDA programmes ask many asymptomatic, healthy people to take a medicine when they do not feel ill,
with the potential for adverse reactions to occur. Improving coverage and adherence requires development of understanding
and trust in the institutions implementing the programme. Community engagement is thus a key factor in determining the
success of TDA, to improve participation rates and adherence to the full treatment course of the medicine.
A complete therapeutic course of antimalarial medicine, at doses recommended by the manufacturer, should be given to all
eligible adults and children. Drug dosage should be determined by weight wherever possible, with dosing according to age
only in situations where the person’s weight is unknown. The antimalarial medicines chosen for use in TDA should: a) be
WHO recommended and prequalified; b) be efficacious against local parasites; c) be different from the medicine used as
first-line treatment, where possible c) have a superior safety and tolerability profile; d) provide a longer duration of post-
treatment prophylaxis with component medicines that have closely matched pharmacology to reduce the risk of new
infections encountering only a single drug; e) have a positive public reputation and acceptability and f) be available and low-
cost. Programmes in areas with P. falciparum may consider including a single, low-dose of primaquine in TDA programmes in
210 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
order to increase the gametocytocidal effect, although no evidence of an additional benefit from provision of single low-
dose primaquine in a TDA programme was reviewed. A drug regimen that can be administered as a directly-observed single
dose is preferred to multi-day regimens.
Depending on the medicine chosen, certain population groups may need to be excluded from TDA, such as: pregnant
women in their first trimester; infants < 6 months of age or weighing <5kgs; people recently treated with the same medicine;
people with a known allergy to the medicine; anyone with severe acute illness or unable to take oral medication; people
taking medication known to interact with the medicine used for TDA; and people with specific contraindications to the
medicine used. Although rarely implemented in the same area, TDA should not be given to individuals receiving other forms
of malaria chemoprevention (e.g. seasonal malaria chemoprevention, perennial malaria chemoprevention, or intermittent
preventive treatment during pregnancy) [141].
Programmes contemplating providing medicine for radical cure of P. vivax hypnozoites as part of TDA programme should
carefully consider whether it is feasible to administer this treatment regimen safely, i.e. with testing for G6PD deficiency
prior to treatment, an effective pharmacovigilance system and emergency access to blood transfusion services. Programmes
should consider whether sufficient coverage and adherence to the full course of radical cure can be achieved.
Evidence To Decision
The GDG determined that TDA would be most appropriate in very low to low transmission or post-elimination settings.
The GDG decided that the PICO question should be modified accordingly (i.e. limited to such settings) and that only the
two NRSs conducted in post-elimination settings should be considered as direct evidence of the impact of TDA.
Beneficial outcomes
• The evidence is very uncertain about the effect of TDA on the prevalence of malaria. (Both NRSs found no malaria
cases in either the targeted group or the community after use of TDA in migrant workers among whom malaria had
been detected prior to the intervention; very low-certainty evidence).
Adverse events
• One NRS monitored adverse events 1–5 months post-intervention and no serious adverse events were reported
during or after the treatment.
• One NRS recorded adverse events in 397 out of the 1094 treated individuals; the majority were classified as minor:
predominantly dizziness and headache for chloroquine and abdominal pain for primaquine. A single case of
primaquine-induced haemolysis was recorded in a person with an incorrect G6PD test result.
The GDG judged the potential benefits of the TDA strategy in some settings to be large, particularly if TDA contributes
to the prevention of re-establishment of transmission. The potential undesirable effects of TDA were judged to be small.
The GDG determined that the balance of effects probably favoured TDA in settings of very low to low transmission or
post-elimination of malaria.
211 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The GDG judged that there may be important uncertainty or variability in preferences or values that could not be
determined due to the lack of studies.
Resources
No studies on the cost or cost-effectiveness of TDA in areas of very low to low transmission or post-elimination settings
were found.
The GDG judged that the costs required to implement TDA were moderate, and that cost-effectiveness probably
favoured implementation of TDA.
Equity
No studies were identified addressing the issue of whether TDA increased or decreased health equity. However, one
article was identified that discussed the potential for strategies such as TDA to lead to social stigmatization if care was
not taken to choose terms and descriptions carefully and focus on higher risk situations rather than specific
groups [249].
The GDG judged that a targeted strategy that intervenes in a small group of people more affected by malaria than the
population surrounding them would likely improve health equity. However, the GDG recognized that, although malaria is
not itself a stigmatizing disease, targeting specific groups might raise fears that they were sources of contagion and
could lead to social isolation and stigmatization.
Acceptability
No studies were identified addressing the issue of acceptability of TDA in areas of very low to low transmission or post-
elimination of malaria.
Feasibility
No studies were identified that addressed the issue of feasibility of implementing TDA in areas of very low to low
transmission or post-elimination of malaria.
The GDG identified several factors likely to affect the feasibility of implementing the strategy, including the choice of
drug and the size of the area to be covered. The feasibility of implementing TDA would also vary depending on whether
radical cure for P. vivax using an 8-aminoquinoline medicine was part of the TDA strategy, which would necessitate
testing for G6PD deficiency, an effective pharmacovigilance system and emergency access to blood transfusion services.
The GDG judged that implementation of the strategy was feasible with significant planning and agreement of the local
authorities.
Justification
Although the quality of evidence was very low, the GDG concluded that the balance of effects probably favoured
implementing TDA, particularly in post-elimination settings to prevent re-establishment of transmission. As long as it is
relatively simple to identify individuals or groups at increased risk of infection, and care is taken to avoid stigmatizing these
212 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
groups, TDA is likely to be more equitable, acceptable and feasible than mass strategies involving the entire population of an
area.
Research Needs
• Further evidence is needed on the impact (prevalence and incidence of malaria infection at the community level ) and
potential harms/unintended consequences of TDA for malaria in very low to low transmission or post-elimination
settings.
• Evidence is needed on the acceptability, feasibility, impact (prevalence and incidence of malaria infection at the
community level) and potential harms/unintended consequences (death, hospital admission, severe anaemia or any
severe adverse events) of safe provision (including testing for G6PD deficiency and, additionally, an effective
pharmacovigilance system and emergency access to blood transfusion services) of an 8-aminoquinoline as part of TDA
for radical cure of P. vivax infections.
• Investigate approaches to characterizing higher-risk situations with respect to their contribution to the overall human
infectious reservoir.
• Evidence is needed to optimize the delivery of TDA with respect to the synchronicity of treatments, time intervals
between rounds of treatment, number of rounds needed per year and number of years needed to sustainably reduce
malaria transmission.
• Evidence is needed on whether TDA stigmatizes groups that might already be socially isolated, such as migrants or
refugee populations.
Testing and treatment of people with an increased risk of infection relative to the general population to reduce the
transmission of malaria is not recommended.
The GDG noted that there may be limited circumstances under which targeted testing and treatment (TTaT) could be beneficial. For
example, TTaT could be used when people at a higher risk of infection can be easily identified and chemoprevention is not
acceptable to the population. Additionally, TTaT could be used if safe and effective implementation of radical cure to prevent P.
vivax relapses is only feasible for those with confirmed infections.
Evidence To Decision
213 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
community level. No studies were conducted in very low to low transmission or post-elimination settings.
The GDG determined that the TTaT strategy would be most relevant in very low to low transmission or post-elimination
settings and, therefore, decided that the PICO question should be modified accordingly (i.e. limited to such settings). As
a result, the GDG did not consider evidence on benefits from the studies included in the review.
The potential harms (i.e. adverse events) from the intervention were considered likely to be trivial, as people who
received treatment would be infected with malaria and, therefore, would receive treatment according to national
guidelines.
The judgements of the GDG related to the balance of effects was based on its expert opinions and indirect information
from related interventions, such as MTaT and TDA. The GDG judged that the balance of effects probably favoured not
implementing TTaT.
The GDG judged that there may be important uncertainty or variability in preferences or values that could not be
determined due to the lack of studies.
Resources
No studies on the cost or cost-effectiveness of TTaT in very low to low transmission or post-elimination settings were
found.
The GDG judged that the costs required to implement TTaT were moderate, and that the cost-effectiveness probably
favoured not implementing TTaT.
Equity
No studies were identified that addressed the issue of whether TTaT increased or decreased health equity.
The GDG judged that a targeted strategy that intervenes in a small group of people more affected by malaria than the
population surrounding them would improve health equity.
Acceptability
No studies were identified that addressed the issue of acceptability of TTaT in areas of very low to low transmission or
post-elimination of malaria.
The GDG judged that TTaT was probably acceptable to stakeholders, as it is a type of active case detection.
Feasibility
No studies were identified that addressed the issue of feasibility of TTaT in areas of very low to low transmission or
post-elimination of malaria.
The GDG noted that the type of parasitological test used (rapid diagnostic test, microscopy or nucleic acid-based test)
214 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
would affect the feasibility of implementing the strategy as tests that are not point-of-contact would be more difficult to
implement, require more technical staff and delay identification and treatment of positive cases.
The feasibility of implementing TTaT would also depend on whether radical cure of P. vivax using an 8-aminoquinoline
medicine was part of the TTaT strategy, which would necessitate testing for G6PD deficiency, an effective
pharmacovigilance system and emergency access to blood transfusion services.
Justification
The GDG judged that the likely impact of TTaT on malaria transmission in very low to low or post-elimination settings would
be trivial, based on experiences with MTaT, challenges with detecting very low parasite densities and a lack of diagnostics for
hypnozoites. The GDG felt that there may be specific situations where TTaT could be beneficial, for example, when the
parasite reservoir is very clearly limited to a small group of people and infections are detectable. Additionally, TTaT could be
used if chemoprevention is either not acceptable to the population or safe and effective implementation of radical cure to
prevent P. vivax is only feasible for those with confirmed infections, but in most settings, TTaT is not likely to reduce malaria
transmission.
Research Needs
While further evidence of the impact (prevalence and incidence of malaria infection at the community level) of TTaT could
change the direction or strength of the recommendation given the lack of published studies on the impact of TTaT, the GDG
did not judge that this research gap was a priority.
Routine malaria testing and treatment of people arriving at points of entry (land, sea or air) to reduce importation is not
recommended.
No studies of the impact of testing and treatment at points of entry on the rate of malaria importation were found by the
systematic review. Routine testing and treatment for malaria at points of entry is unlikely to be acceptable or feasible to implement.
215 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Evidence To Decision
Malaria testing and treatment of organized or identifiable groups arriving or returning from malaria-endemic areas (2022)
Relatively easy access to these groups within a short time after entry is required for this strategy to be feasible and
acceptable. This strategy may be particularly critical to areas in post-elimination that are working to prevent re-
establishment of transmission.
Evidence To Decision
The GDG noted that border screening may take two forms: the traditional approach of testing and treatment of
individuals at the time of entry through land crossings, seaports or airports; and the testing and treatment of organized
or identifiable groups (e.g. military, migrant workers or religious pilgrims) recently arriving or returning from malaria-
endemic areas. Because there are clear differences in the feasibility and acceptability of these two approaches, the GDG
developed two separate recommendations.
The benefits of testing and treatment at points of entry could not be assessed as no studies reporting on critical
outcomes were identified by the review.
The potential harms (i.e. adverse events) from the intervention were considered likely to be trivial, as people who
received treatment would be infected with malaria and, therefore, treated according to national guidelines.
The GDG judged that the balance of effects probably varied depending on the source population, strictness of entry into
the area, coverage of the intervention, species of parasite, type of parasitological test and the area’s epidemiological
profile with respect to malaria.
The GDG judged that there may be important uncertainty or variability in preferences or values that could not be
determined due to the lack of studies.
216 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Resources
The systematic review identified one study with data on the cost of testing and treatment at points of entry (Coma-Cros
et al unpublished evidence). One NRS study of testing and treatment of recently arrived migrant workers estimated that
the programme cost US$ 226 080 annually between 2013 and 2017. No studies of the cost-effectiveness of testing and
treatment at points of entry were identified.
The GDG judged the costs required to implement testing and treatment at points of entry to be moderate, and the cost-
effectiveness probably varied depending on whether the intervention was applied to individuals at the point of entry or
to organized or identifiable groups immediately after arrival. In the latter case, the GDG judged that the intervention was
probably cost-effective compared to not testing and treating organized or identifiable groups.
Equity
No studies were identified that addressed the issue of whether testing and treatment at points of entry increased or
decreased health equity.
The GDG judged that the testing and treatment of organized or identifiable groups was likely to be more equitable than
the routine testing and treatment of individuals at the point of entry.
Acceptability
The systematic review identified two studies of acceptability (Coma-Cros et al unpublished evidence). One study assessed
the number of refusals for testing at border crossing points between Cambodia and Thailand, Viet Nam and Lao People’s
Democratic Republic. Out of 4110 people approached, 904 (22%) refused to be tested for malaria. The main reasons for
refusal included: not having enough time (51.6%); not perceiving themselves to be at risk of malaria and thus not
requiring testing (40.6%); being scared to give blood (34.2%); and having an apparent language or cultural barrier
(23.9%) (multiple answers possible). A qualitative study conducted in 2010 in the Solomon Islands on the feasibility and
acceptability of testing all travellers using rapid diagnostic tests and offering treatment to those found positive
suggested that there was acceptance and support for such an approach.
The GDG judged that acceptance of testing and treatment at points of entry was likely to vary by stakeholder (less
acceptable to travellers at the time of entry but potentially more acceptable to organized groups immediately after
entry). Among travellers, acceptability is likely to vary considerably depending on factors such as the location of the
screening, the time required and the travellers’ level of concern regarding possible malaria infection.
Feasibility
No studies on the feasibility of implementing testing and treatment at points of entry were identified.
The GDG noted that the type of parasitological test used (rapid diagnostic test, microscopy or nucleic acid-based test)
would affect the feasibility of implementing the strategy as tests that are not administered at point-of-contact would be
more difficult to implement, require more technical staff and delay identification and treatment of positive cases.
The GDG judged that the feasibility of implementing routine testing and treatment at points of entry would likely vary.
Implementing such an intervention at airports or seaports was considered unlikely to be feasible due to the high volume
of travellers and the time required to test and treat. The feasibility of implementing testing and treatment at land
crossings was considered to be more feasible but would depend on the volume of travellers. Additionally, the feasibility
of covering a high proportion of people crossing into the country through land crossings would depend on the strictness
with which entry into the country was controlled and the porosity of the border. In most areas with porous borders, the
GDG judged that the feasibility of implementing a testing and treatment with sufficient coverage at land crossings would
be low.
However, the feasibility of implementing testing and treatment among organized or identifiable groups arriving or
recently returned from malaria-endemic areas was considered to be high. The GDG knew of many reports of military
groups, labour migrants and religious groups in countries eliminating malaria or preventing re-establishment who were
tested and treated for malaria after returning from periods in malaria-endemic areas.
217 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Justification
With respect to routine testing and treatment at points of entry (land, sea or air), the GDG, in the absence of direct evidence,
judged that the impact on importation of malaria was likely to be small except in areas nearing elimination or post-
elimination. The GDG felt that the acceptability and feasibility of testing and treating for malaria at points of entry would be
low given the likely disruption to travel.
When considering organized or identifiable groups of people (e.g. military, migrant laborers or religious pilgrims) arriving or
returning from malaria -endemic areas, the GDG judged that testing these groups for malaria and treating those who are
positive could help countries nearing elimination or preventing re-establishment by reducing importation. The acceptability
and feasibility of this strategy was considered higher than routine testing and treatment at points of entry but would depend
upon the local circumstances.
Research Needs
• Evidence is needed on the efficiency (number of imported cases identified as a proportion of all imported cases
identified during the same period) of testing and treating organized or identifiable groups of people arriving or returning
from malaria-endemic areas in terms of the importation of malaria.
• Investigate novel approaches to improving the efficiency of identifying and implementing testing and treatment among
organized or identifiable groups, such as the plantation ambassador programme in Malaysia.
218 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
be extended to neighbours. If the infection was imported from have had the same exposure as the index case, such as co-
elsewhere, RDA can be administered to individuals who may travellers and co-workers.
• Programmes implementing reactive drug administration (RDA) should have the capacity to conduct case investigations at the
residence to determine the likely location of infection and to identify those individuals co-exposed with the index case.
• Programmes implementing RDA should have the capacity to enumerate and provide antimalarials to the people residing with
or near a confirmed malaria case and others that share the same risk of infection.
• The people given antimalarial medicine in an RDA intervention should share the same risk of having acquired infection as the
index case or be at risk of acquiring infection from the index case. This includes residents in the same household or
neighborhood, co-travellers and co-workers. However, if the infection was imported and the residence is not located in a
receptive area, there may be no benefit from RDA.
• Programmes contemplating implementation of RDA for P. vivax should carefully consider how to safely and feasibly administer
treatment to prevent relapses.
Practical Info
When used, RDA should be one of several components of a programme to eliminate or prevent re-establishment of malaria,
including intensive follow-up of every case as described in the Framework for malaria elimination [10].
RDA depends on a strong passive surveillance system that detects suspected cases, tests all suspected cases for malaria
with a quality-assured parasitological test and investigates all cases at their residence. If these elements are not in place, it is
unlikely that an RDA intervention will have any effect on transmission.
It is essential to determine the likely location of infection through a case investigation that identifies the location of the
person during the likely period of infection in order to understand where or in what group of people the RDA intervention
should take place. RDA should be administered to other residents of the same house if the person is determined to have
been infected locally. Programmes may consider extending the radius of RDA to neighbours depending on the local
epidemiology and ecology of malaria. If the index infection is not likely to have been acquired at the residence, programmes
should administer RDA to all people identified as having the same exposure to infection as the index case. People with the
same risk of infection are likely to be those who travelled, worked or engaged in leisure activities with the index case. If the
infection was classified as imported from elsewhere and the household is not located in a receptive area, there may be no
benefit to RDA.
Countries that are at very low or low transmission but not yet close to achieving zero indigenous cases should prioritize
implementation of RDA and reactive IRS over RACDT. However, RACDT should be added on top of RDA when countries are
closer to elimination to strengthen the sensitivity of the surveillance system to monitor progress towards elimination and,
post-elimination, to provide additional evidence of a malaria-free status.
RDA should be implemented according to standardized operating procedures (SOPs). A household listing of all people
residing within the limits of RDA as specified by the SOPs should be developed and verified, along with a list of all people
who may have been co-exposed. The RDA programme should seek to provide antimalarial medicine to everyone listed, using
different approaches as needed to reach everyone at risk.
Achieving high coverage of the targeted population and good adherence to the antimalarial medicine are critical aspects of
RDA programmes. RDA programmes ask many asymptomatic, healthy people to take a medicine when they do not feel ill,
with the potential for adverse reactions to occur. Improving coverage and adherence requires development of understanding
and trust in the institutions implementing the programme. Community engagement is thus a key factor in determining the
success of RDA, to improve participation rates and adherence to the full treatment course of the medicine.
A complete therapeutic course of antimalarial medicine, at doses recommended by the manufacturer, should be given to all
eligible adults and children. Drug dosage should be determined by weight wherever possible, with dosing according to age
219 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
only in situations where the person’s weight is unknown. The antimalarial medicines chosen for use in RDA should: a) be
WHO recommended and prequalified; b) be efficacious against local parasites; c) be different from the medicine used as
first-line treatment, where possible c) have a superior safety and tolerability profile; d) provide a longer duration of post-
treatment prophylaxis with component medicines that have closely matched pharmacology to reduce the risk of new
infections encountering only a single drug; e) have a positive public reputation and acceptability and f) be available and low-
cost. Programmes in areas with P. falciparum may consider including a single, low-dose of primaquine in an RDA programmes
in order to increase the gametocytocidal effect, although there is no evidence of additional benefit from provision of of
single low-dose primaquine in an RDA programme. A drug regimen that can be administered as a directly-observed single
dose is preferred to multi-day regimens.
Depending on the medicine chosen, certain population groups may need to be excluded from RDA, such as: pregnant
women in their first trimester; infants < 6 months of age or weighing < 5kgs; people recently treated with the same
medicine; people with a known allergy to the medicine; anyone with severe acute illness or unable to take oral medication;
people taking medication known to interact with the medicine used for RDA; and people with specific contraindications to
the medicine used [141]. Although rarely implemented in the same area, RDA should not be given to individuals receiving
other forms of malaria chemoprevention (e.g. seasonal malaria chemoprevention, perennial malaria chemoprevention, or
intermittent preventive treatment during pregnancy).
Programmes contemplating providing medicine for radical cure of P. vivax hypnozoites as part of RDA should carefully
consider whether it is feasible to administer this treatment regimen safely, i.e. with testing for G6PD deficiency prior to
treatment, an effective pharmacovigilance system and emergency access to blood transfusion services. Programmes should
consider whether sufficient coverage and adherence to the full course of radical cure can be achieved.
Evidence To Decision
Beneficial outcomes
• RDA may reduce malaria prevalence (RD: -5 per 1000 persons; 95% CI: -9 to 2 per 1000 persons; four cRCTs; low-
certainty evidence).
• RDA probably reduces the incidence of parasitaemia (RD: -7 per 1000 p-y; 95% CI: -17 to 13 per 1000 p-y; two
cRCTs; moderate-certainty evidence).
• RDA probably results in little to no difference in the incidence of clinical malaria (RD: -2 per 1000 p-y; -4 to 1 per
1000 p-y; six cRCTs; moderate-certainty evidence).
• The evidence is very uncertain about the effect of RDA on the incidence of clinical malaria. (RD: -2 per 1000 p-y; -3
to -1 per 1000 p-y; one NRS; very low-certainty evidence).
Adverse events
Four cRCTs reported on adverse events; however, only two studies reported adverse events from the RDA arm and the
comparator arm. In RDA arms with DP:
• 123 (6.9%) mild adverse events were reported from 1775 participants receiving DP; all were resolved.
• 75 (7.6%) adverse events were reported from 979 participants receiving DP; 69 were rated as mild and six as
moderate.
• 68 (3.8%) adverse events reported from 1776 participants receiving DP; 54 were rated as mild and 14 as moderate.
220 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The NRS in Peru that used chloroquine plus seven days of primaquine for radical cure of P. vivax hypnozoites reported
no adverse events but there was no active pharmacovigilance system.
The GDG judged that there may be important uncertainty or variability in preferences or values that could not be
determined due to the lack of studies.
Resources
The systematic review identified one study from Zambia with data on the financial and economic costs of RDA
(Steinhardt et al unpublished evidence (c)). The study identified index cases through active rather than passive
surveillance. The total cost of two rounds of RDA (with DP) conducted between 2014 and 2015, covering a total
population of 132 393 was US$ 912 767 (all figures in 2015 US$). The mean cost per person reached was US$ 85.69
(interquartile range [IQR] US$39.92).
The overall incremental costs per infection and case averted (vs. standard of care) for RDA were US$ 810 and US$ 6
353, respectively. In high transmission settings, the incremental costs per infection and case averted were US$ 429 and
US$ 5951, respectively; in low transmission settings, they were US$ 1119 and US$ 6755, respectively. Incremental cost
per DALY averted for infections and cases were US$ 4889 and US$ 38 344, respectively.
The GDG judged the resources required for RDA to be large but dependent on the number of index cases.
Equity
No studies were identified that addressed the issue of whether RDA increased or decreased health equity.
Acceptability
The systematic review identified six studies in four countries (Eswatini, Gambia, Namibia and Zambia) with information
on acceptability (Steinhardt et al unpublished evidence (c)). Community acceptance of RDA was high (refusal rate of 2% or
lower) in Namibia and Zambia. However, in Eswatini, the overall refusal rate was about 4%, with refusal rates of 1.4%
(11/776) and 5.3% (65/1232) in seasons 1 and 2, respectively. In Namibia, participants expressed concern over having
“to take medicine without feeling sick”. Similarly, participants in Gambia “generally considered it unnecessary to take
medicine without having any symptoms”. Continued community sensitization has been recommended to mitigate these
stigmas. In the systematic review, no studies reporting on the acceptability of RDA to health care workers or
policymakers were found.
221 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The GDG judged that RDA was probably acceptable to key stakeholders given the high rate of participation in RDA
programmes.
Feasibility
Data on the feasibility of implementing RDA were summarized from five studies in four countries (Eswatini, Gambia,
Namibia and Zambia) (Steinhardt et al unpublished evidence (c)). All countries used a three-day regimen of an ACT. RDA
coverage, defined as the proportion of index cases followed up, varied between countries with a low of 62.4% in
Eswatini to about 97% in Gambia.
RDA adherence data were abstracted from three studies in three countries (Eswatini, Gambia and Zambia). Full
adherence, defined as taking all three doses of an ACT and verifying that no tablets remained in the blister pack, was
above 90% in all the countries.
The feasibility of implementing RDA would also depend on whether radical cure of P. vivax using an 8-aminoquinoline
medicine was part of the RDA strategy, which would necessitate testing for G6PD deficiency, an effective
pharmacovigilance system and emergency access to blood transfusion services.
Justification
While the GDG concluded that the balance of effects favoured neither RDA nor the comparison, the panel judged that the
intervention would likely have been more effective if studies had compared RDA to no RDA rather than to RACDT. The GDG
judged that RDA was probably an acceptable, feasible and potentially cost-effective strategy when numbers of cases are low
enough to permit programmes to conduct case investigations, including in post-elimination settings working to prevent re-
establishment of infection. The GDG concluded that a conditional recommendation for RDA as a component of an
elimination programme should be issued.
Research Needs
• Further evidence is needed on the impact (prevalence and incidence of malaria infection at the community level) and
potential harms/unintended consequences of RDA.
• Evidence is needed on the acceptability, feasibility, impact (prevalence and incidence of malaria infection at the
community level) and potential harms/unintended consequences (death, hospital admission, severe anaemia or any
severe adverse event) of safe provision (including testing for G6PD deficiency and, additionally, an effective
pharmacovigilance system and emergency access to blood transfusion services) of an 8-aminoquinoline as part of RDA
for radical cure of P. vivax infections.
• Investigate the optimal approach to delimiting the target area for implementation of RDA around an index case in order
to maximize reductions in transmission of malaria.
• Determine the optimal time interval between index case detection and RDA to maximize reductions in transmission of
malaria.
• Determine whether additional rounds of RDA should be repeated in the same residences or neighborhood to prevent
subsequent generations of transmission.
222 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
antimalarial medicine only to those who are known to be malaria in the area. If the index infection was imported from
infected may improve adherence to treatment, population elsewhere, RACDT should be conducted among individuals
acceptance of the intervention and equity while decreasing the who may have the same exposure as the index case, such as
risk of unintended consequences and depleting stocks of co-travellers and co-workers.
medicines.
As an active case detection strategy, RACDT is an essential
Reactive interventions that are applied geographically should component of the final phase of elimination as it improves the
target the likely location of infection of the index case. The sensitivity of the surveillance system while maintaining
likely location of infection is determined through a case specificity; RACDT accomplishes this by increasing testing in
investigation, using the date of symptom onset and knowledge areas more likely to experience transmission of malaria.
of the incubation period for the specific parasite species to RACDT provides important information to countries close to
determine the location of the person during the likely period of elimination by identifying any additional cases around the
infection. If the likely location of infection is a residence, index case that could suggest gaps in the surveillance system.
RACDT can be conducted at least in the household of the Once countries have reached zero indigenous cases, RACDT
person with the confirmed case, but could also be extended to provides additional evidence to the Malaria Elimination
neighbours. The radius of the intervention should be Certification Panel that the country has interrupted indigenous
determined based on an understanding of the epidemiology of transmission.
In areas approaching elimination or post-elimination settings preventing re-establishment of transmission, all people residing
with or near a confirmed malaria case and all people who share the same risk of infection (e.g. co-travellers and co-workers)
can be tested for malaria and treated if positive.
Until an area is nearing elimination or is post-elimination, it is unlikely that reactive case detection and treatment (RACDT) will have
any effect on malaria transmission. However, RACDT becomes an essential component of surveillance when countries are nearing
interruption of transmission to monitor progress towards elimination. When countries are post-elimination and working towards
certification, RACDT can strengthen a country’s claim that it has reached and maintained zero indigenous cases. RACDT is an
essential part of surveillance and response to prevent re-establishment of malaria.
Practical Info
RACDT should be implemented when areas are nearing interruption of transmission and malaria cases are rare. When used,
RACDT should be one of several components of a programme to eliminate or prevent re-establishment of malaria, including
intensive surveillance as described in the Framework for malaria elimination [10].
RACDT depends on a strong passive surveillance system that detects suspected cases, tests all suspected cases for malaria
with a parasitological test and investigates all cases at their place of residence. RACDT complements this surveillance system
through active case finding around index cases.
It is essential to determine the likely location of infection through a case investigation that identifies the location of the
person during the likely period of infection in order to understand where or in what group of people an RACDT intervention
should take place. RACDT should be administered to other residents of the same house if the person is determined to have
been infected locally. Programmes may consider extending the radius of RACDT to neighbours depending on the local
epidemiology and ecology of malaria. If a person was not likely to have been infected at the residence, programmes should
administer RACDT to all people identified as having the same risk of acquiring infection as the index case. People with the
same risk of infection are likely to be those who travelled, worked or engaged in leisure activities with the index case.
Countries that are at very low or low transmission but not yet close to achieving zero indigenous cases should prioritize
implementation of RDA and reactive IRS over RACDT. However, RACDT may be added on top of RDA when countries are
closer to elimination to strengthen the sensitivity of the surveillance system to monitor progress towards elimination and,
post-elimination, to provide additional evidence of a malaria-free status. When RACDT and RDA are jointly implemented,
chemoprevention is provided to everyone, irrespective of the results of the parasitological test. However, testing results are
used to monitor progress towards elimination or demonstrate that the country has reached zero indigenous cases.
RACDT should be implemented according to SOPs. A household listing of all people residing within the limits of RDA as
223 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
specified by the SOPs should be developed and verified, along with a list of all individuals who may have been co-exposed.
The RACDT programme should seek to test everyone listed, using different approaches as needed to reach everyone at risk.
Malaria cases detected during RACDT should be treated with antimalarial medicine according to the national treatment
protocol if not already provided chemoprevention through RDA.
Evidence To Decision
Beneficial outcomes
• The evidence is very uncertain about the effect of RACDT on the prevalence of malaria (RD: 25 per 1000 persons;
95% CI [95% CI] -1 to 72 per 1000 persons; one cRCT; very low-certainty evidence).
• The evidence is very uncertain about the effect of RACDT on the incidence of clinical malaria (RD: 3 per 1000 p-y;
95% CI: -1 to 7 per 1000 p-y; three cRCTs; very low-certainty evidence).
• The evidence is very uncertain about the effect of RACDT on parasite prevalence among people who participate in
RACDT (two NRSs; very low-certainty evidence).
Adverse events
Three cRCTs reported on adverse events. All trials used AL in the RACDT arms while DP was provided for the RDA arms.
• In Zambia, no events were reported from the RACDT arm compared to 123 (6.9%) adverse events reported from 1
775 persons administered DP in the RDA arm;
• In Namibia, 1 (1.0%) participant out of 96 reported an adverse event compared to 17 (0.4%) out of 4 247 in the
RDA arm using DP;
• In Eswatini, no adverse events were reported from the RACDT arm while 68 (3.8%) of 1 776 participants reported
adverse events in the RDA arm provided with DP.
The GDG judged that there may be important uncertainty or variability in preferences or values that could not be
determined due to the lack of studies
224 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Resources
The systematic review identified four studies with information on the costs of RACDT (Steinhardt et al unpublished
evidence (d)). The average cost of RACDT varied across different regions – from US$ 5.21 in Thailand to US$ 27.60 in
Indonesia. In Senegal, the cost per person screened by RACDT was US$ 14.00. Costing models developed based on the
experience of implementing partners, operational documents and costing studies from Ethiopia, Senegal and Zambia
found that the average annual financial cost per capita (total population of 360 000 based on one region, three districts,
20 health facility catchment areas [HFCAs] each, and 6000 population per HFCA) were US$ 1.07 for the first year of
RACDT, and US$ 0.65 per year for the subsequent five years (2014 US$) and the per capita economic cost was US$
1.27 in first year of RACDT, and US$ 0.75 per year for the subsequent five years (2014 US$).
Total costs for RACDT varied between study areas ranging from US$ 3469 in Indonesia to US$ 10 486 in Thailand for
total personnel and US$ 257 (Indonesia) to US$13 969 (Thailand) for commodities, services and other costs. The
variations in personnel, commodity, service and other costs specific to case investigation and RACDT activities are likely
due to differences in programme structure and the level of integration of malaria-related activities into the broader
healthcare system.
In Zambia, the mean annual cost of RACDT per HFCA was US$ 1177 (median = US$ 923, IQR US$ 651–1417). The
variation in costs was driven by the number of community health workers and index cases detected. Costs related to
community health workers and data review meetings accounted for the largest share of total costs. Rapid diagnostic
tests and medicines accounted for less than 10% of total costs.
Cost models based on studies from Ethiopia, Senegal, and Zambia showed that targeted search radius and per diems
paid to community health workers dominated intervention parameters. In Indonesia, at 0.4% prevalence of infection, the
cost per infection detected was US$ 7070, which declined to US$ 1767 when the prevalence was 1.6%. Cost declines
began to plateau thereafter.
The GDG judged the resources required for RACDT to be moderate, depending on the number of index cases.
Equity
No studies were identified that addressed the issue of whether RACDT increased or decreased health equity.
Acceptability
The systematic review identified community acceptability data from three studies conducted in Namibia, Senegal and
Zambia (Steinhardt et al unpublished evidence (d)). Community acceptance of RACDT was high (refusal rate 2% or lower).
In Namibia, some “hesitation/resistance” during pre-trial was reported but community engagement and sensitization
appear to have helped participation. Similarly, in Senegal, the high RACDT participation has been attributed to advanced
cascade sensitization, making follow-up appointments to follow up absent members, and conducting return visits to the
compound on the same or next day. Lack of community confidence in community health workers’ ability to address
diseases other than malaria and community unwillingness to visit community health workers for malaria testing were
reported in Zambia.
There were no studies reporting data directly on health care workers’ acceptance of RACDT. Related information was
abstracted from two studies. In Zambia, community health workers reported lack of motivation to conduct RACDT,
which was in part linked to community health workers feeling their community service went unrecognized. The lack of
stipend or financial support was the biggest problem noted by community health workers, who were volunteers.
The GDG judged that RACDT was probably acceptable to key stakeholders.
Feasibility
The systematic review identified feasibility and health systems considerations data from 17 studies, of which seven were
from sub-Saharan Africa and eight from the Asia-Pacific region (Steinhardt et al unpublished evidence (d)). The proportion
225 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
of households reached by RACDT varied across different geographical locations – from 49% of index case households
investigated in Zanzibar to 100% in Jiangsu, China. Similarly, the proportion of households reached in a timely manner
also varied across different locations – from about 20% in Zanzibar to 100% in China. Barriers and challenges to RACDT
implementation were identified along all three steps of RACDT.
First, index case detection and notification from private health facilities was low and these cases were largely reported
to be missed by RACDT in Cambodia and Zanzibar. Collaborating and engaging the private sector in malaria surveillance
systems has been identified as critical, particularly in areas where many patients resort to private providers, facilities
including drug shops, and pharmacies. Within the public health sector, delayed presentation of malaria patients to health
facilities, poor preparation of village clinics to participate in surveillance programmes, and the lack of adequate human
resources and malaria rapid diagnostic tests haves been reported as barriers and challenges to effective implementation
of RACDT. Second, the complexity of case investigation procedures and lack of standard operating procedures have
been identified as barriers to effective case investigation. Difficulty with case classification (imported vs. local) due to
incomplete travel histories has also been reported. During peak malaria transmission seasons, the proportions of case
investigations conducted were lower than in other times mainly because community health workers were overwhelmed
by patient volumes and there were insufficient numbers of malaria rapid diagnostic tests. To overcome these barriers,
authors from a study in Zambia suggested that the programme would benefit from additional community health workers
or the suspension of RACDT during the high-transmission season. Third, difficulty accessing mountainous terrains,
flooded areas, and border areas with highly mobile populations were reported as barriers to timely follow up during the
RACDT intervention. To overcome the barriers posed by flooding during the rainy season, study authors from Zambia
recommended that community health workers, particularly those serving flood-prone areas, be provided with rain gear
and access to boats.
Another barrier to effective implementation of RACDT was identified as the large numbers of households to screen,
particularly in high-density areas of the Asia-Pacific region. Incomplete case investigation forms also limited follow-up
and the lack of household-level listings of all individuals in the RACDT area meant that those conducting RACDT did not
always know which households to include in the RACDT. Imported cases posed a major challenge for RACDT
interventions. District-level responses alone were unlikely to be effective in interrupting transmission when most malaria
cases were imported from outside the district. Communication and surveillance linkages with other operational districts
and their malaria response teams were considered necessary. In the case of Bhutan, RACDT buffer zones sometimes
extended beyond international borders, limiting implementation of adequate RACDT activities. Strengthened cross-
border collaborations are needed to ensure adequate coverage of households across borders, as well as migrant and
mobile populations. Other barriers to conducting effective RACDT were stockouts of malaria rapid diagnostic tests,
which prevented testing around index cases, the limit of detection of most rapid diagnostic tests, and the inability of P.
falciparum-only rapid diagnostic tests to detect other species and low-density infections. In Botswana, malaria
microscopy was used as the gold standard for malaria diagnosis, so all RDT-positive malaria cases were re-examined by
microscopy; however, it was challenging to ensure a high quality of malaria microscopy slides prepared by health centre
staff in these settings. A lack of health care workers to conduct malaria activities and lack of surveillance officers at the
district level were reported to result in inadequate supervision, case investigation and follow-up. Lack of motivation
among health care workers to pursue case investigation and contact testing, particularly on weekends and public
holidays, was also reported. Maintaining workforce motivation and providing consistent support, supervision and
incentives were recommended to overcome these challenges.
The feasibility of implementing RACDT would also depend on whether radical cure of P. vivax using an 8-aminoquinoline
medicine was part of the RACDT strategy, which would necessitate testing for G6PD deficiency, an effective
pharmacovigilance system and emergency access to blood transfusion services.
Justification
Although the GDG was not presented with any relevant evidence for the benefit of RACDT in reducing transmission of
malaria, RACDT is considered an essential surveillance strategy for countries nearing elimination in order to ensure that
there are no cases remaining around or associated with a confirmed case. The GDG concluded that a conditional
recommendation for RACDT as a component of the end-stage of an elimination programme should be issued.
Research Needs
No research needs were identified by the GDG.
226 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
In areas approaching elimination or post-elimination settings preventing re-establishment of transmission, indoor residual
spraying of insecticide can be conducted in in the houses of confirmed cases and neighbours to prevent or reduce
transmission of malaria.
• In areas approaching elimination or post-elimination settings where proactive indoor residual spraying (IRS) is occurring,
programmes can consider switching to reactive IRS only, depending on the receptivity of the area.
• Programmes considering adding reactive IRS on top of proactive IRS should balance the potential added benefit with
increasing cost and the risk of insecticide resistance.
• In areas approaching elimination or post-elimination settings where no IRS is occurring, initiating reactive IRS may be
beneficial, depending on whether IRS is a suitable vector control strategy. IRS is most effective where the vector population is
susceptible to the insecticide(s) being applied, the majority of mosquitoes feed and rest indoors and where most structures are
suitable for spraying.
• If the index infection was imported and the residence is not located in a receptive area, there may be no benefit from reactive
IRS.
Practical Info
Please refer to the Practical Info section for IRS (4.1.1) for more information on operational issues related to IRS.
When used, reactive IRS should be one among several components of a programme to eliminate or prevent re-establishment
of malaria, including intensive surveillance as described in the Framework for malaria elimination [10].
Reactive IRS depends on a strong passive surveillance system that detects suspected cases, tests all suspected cases for
malaria with a parasitological test and investigates all cases at their place of residence. If these elements are not in place, it is
unlikely that an reactive IRS intervention can be effectively implemented.
It is essential to determine the likely location of infection through a case investigation that identifies the location of the
person during the likely period of infection in order to understand where the reactive IRS intervention should take place.
Reactive IRS should be applied to the residence if the person is determined to have been infected locally. Programmes
should extend RIRS to neighbours, with the radius of implementation depending on the local epidemiology and ecology of
malaria. If the index infection is not likely to have been acquired at the residence, reactive IRS might still reduce the chances
of onward transmission. However, if the infection was classified as imported and the household is not located in a receptive
area, there may be no benefit to reactive IRS.
In very low to low transmission settings where standard IRS is occurring (proactive spraying), there may be advantages to
programmes from switching to reactive IRS. Decisions to switch from standard IRS to reactive IRS should be based on
assessments that include:
• the potential risk of increasing malaria transmission by scaling back proactive IRS;
227 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
In settings where no standard IRS is occurring, reactive IRS may be beneficial, depending on the factors listed below.
• The programme has the capacity to conduct case investigations at the residences of cases to determine whether the
case is imported or local.
• The capacity of the vector control programme to respond quickly to conduct reactive IRS after identification of a
confirmed case.
• The population living in the houses where RIRS is applied are at risk of infection.
• The majority of the vector population feeds and rests indoors.
• The vectors are susceptible to the insecticide that is being deployed.
• People mainly sleep indoors at night.
• The majority of structures are suitable for spraying.
Programmes considering adding reactive IRS on top of proactive IRS should balance the potential added benefit with the risk
of insecticide resistance and increased cost, and develop protocols that take into account the time since the dwelling was
last sprayed. Reactive IRS depends upon a strong passive surveillance system that detects suspected cases, tests all
suspected cases for malaria with a parasitological test and investigates all cases at the residence. If these elements are not in
place, it is unlikely that an reactive IRS intervention will have an impact on malaria transmission.
Evidence To Decision
Beneficial outcomes
• Reactive IRS reduces the prevalence of malaria (RD: -27 per 1000 persons; 95% CI: -35 to -8 per 1000 persons;
one cRCT [superiority design]; high-certainty evidence).
• Reactive IRS may have little to no effect on the incidence of clinical malaria. (RD: -14 per 1000 p-y; 95% CI: -32 to
4 per 1000 p-y; one cRCT [superiority design]; moderate-certainty evidence).
• Reactive IRS probably results in little to no difference in incidence of clinical malaria compared with proactive IRS
(mean difference: 0.1 per 1000 p-y; 95% CI: -0.38 to 0.59 per 1000 p-y; one cRCT [non-inferiority design];
moderate-certainty evidence).
Adverse events
• Reactive IRS results in little to no difference in reported adverse events (RD: 2 per 1000 persons; 95% CI: -2 to 1
per 1000 persons; one cRCT [superiority design]; high-certainty evidence).
• Reactive IRS results in little to no difference in serious adverse events (deaths) compared with proactive IRS (one
cRCT [non-inferiority design]; high-certainty evidence).
228 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
The GDG judged that there may be important uncertainty or variability in preferences or values that could not be
determined due to the lack of studies.
Resources
The systematic review identified one study from South Africa with data on cost and cost-effectiveness of reactive IRS
compared to proactive IRS (non-inferiority trial) (Gimnig et al unpublished evidence). Over the two-year study, the average
annual economic cost was US$ 184 319 per 100 000 population in the proactive IRS arm compared to US$ 88 258 per
100 000 population in the reactive IRS arm, a 52% cost savings. Using the cost per DALY, the incremental cost-
effectiveness ratios were estimated overall and for each year of the study. It was estimated that per additional DALY
averted, reactive IRS saved US$ 7845 (95% CI: US$ 2902–64 907) over proactive IRS. During year 1, when the
incidence of malaria was low, the savings per additional DALY averted in the RIRS arm was estimated at US$ 35 149.
The lower bound of the 95% CI was US$ 6481, while at the higher bound, RIRS was both less expensive and more
effective. In year 2, when incidence was higher, the savings per additional DALY averted in the reactive IRS arm was US$
3869 (95% CI: US$ 1371–50 689). The cost-effectiveness thresholds were set at US$ 2637 (43% of GDP per capita)
and US$ 3557 (58% of GDP per capita). At the incidence observed during the trial, reactive IRS would have a 94–98%
probability of being the cost-effective choice at either threshold. It was estimated that reactive IRS would remain the
preferred strategy up to an incidence of 2.0–2.7 cases per 1000 person-years using the higher and lower cost-
effectiveness thresholds.
The GDG judged that the resources required for reactive IRS are likely to vary depending on whether the programme is
moving from proactive IRS to reactive IRS or starting an RIRS programme from scratch. The resource requirements are
also likely to vary depending on the number of index cases. However, the GDG judged that cost-effectiveness probably
favours reactive IRS.
Equity
No studies were identified that addressed the issue of whether reactive IRS increased or decreased health equity.
Because reactive IRS focuses resources where they are needed, the GDG judged that reactive IRS probably increased
health equity.
Acceptability
The systematic review identified one study from Namibia with information on the acceptability of reactive IRS (Gimnig
et al unpublished evidence). Refusals of households to participate in reactive IRS were due to lack of notification before
arrival and reluctance to move furniture at short notice. In year two of the study, advance notification was provided to
households and < 1% refused reactive IRS. Community participants generally considered reactive IRS to be a useful tool
for malaria prevention, and participants in the study arms that did and did not receive reactive IRS indicated a desire to
have their houses sprayed. Participants specifically referenced IRS’s effectiveness, noting reductions in both flies and
mosquitoes. In the endline survey, 616 of 624 respondent (98.7%) from the reactive IRS arm indicated that they would
participate in the same intervention again.
The GDG noted that reactive IRS would likely be more accepted by households than proactive IRS because residents
would know that a malaria case had been detected in or near their home. The GDG judged that reactive IRS was
probably acceptable to key stakeholders.
229 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Feasibility
The systematic review identified two case studies from China that reported on their implementation of reactive IRS
(Gimnig et al unpublished evidence).
The GDG judged that reactive IRS was likely feasible to implement.
Justification
Proactive IRS applied campaign-style across a geographical area has long been a staple of malaria vector control and is
currently recommended by WHO for large-scale deployment in areas of ongoing transmission. Reactive IRS uses the same
intervention (application of a residual insecticide to the interior surfaces of a dwelling) as does proactive IRS; however,
reactive IRS is triggered by a single case of malaria and applied in a limited geographical area around the likely location of
infection of the index case. When transmission is low and cases are clustered, the GDG noted that RIRS might be more cost-
effective than proactive IRS as the area at risk of transmission is more limited. However, the benefits gained by introducing
RIRS are likely to depend on whether the programme already has a proactive IRS programme or not; whether the
programme intends to scale back proactive IRS to reactive IRS or add reactive IRS on top of proactive IRS; and the
characteristics of the vector and human populations. As a result, the GDG provided a conditional recommendation for
reactive IRS.
Research Needs
• Further evidence is needed on the impact (prevalence and incidence of malaria infection at the community level) and
potential harms/unintended consequences of reactive IRS.
• Determine the impact (prevalence and incidence of malaria infection at the community level ) of reactive IRS in areas
with different mosquito behaviours.
• Determine the impact (prevalence and incidence of malaria infection at the community level ) of reactive IRS in areas
where P. vivax is transmitted.
• Investigate the optimal approach to delimiting the target area for implementation of reactive IRS around an index case.
• Determine the optimal time interval between case detection and reactive IRS.
• Determine whether additional rounds of reactive IRS should be repeated in the same households to prevent
subsequent generations of transmission.
• Determine the benefit and acceptability of switching from IRS to reactive IRS or adding reactive IRS on top of proactive
IRS.
7. SURVEILLANCE
Surveillance is “the continuous and systematic collection, analysis planning of resource allocation. In settings where malaria is being
and interpretation of disease-specific data, and the use of that eliminated, the objectives of surveillance are to identify,
data in the planning, implementation and evaluation of public investigate and eliminate foci of continuing transmission, prevent
health practice” [251]. and cure infections, and confirm elimination. After elimination has
been achieved, the role of surveillance becomes that of preventing
Pillar 3 of the Global technical strategy for malaria 2016–2030 [5] is re-establishment of malaria.
to transform malaria surveillance into a key intervention in all
malaria-endemic countries and in those countries that have A malaria surveillance system comprises the people, procedures,
eliminated malaria but remain susceptible to re-establishment of tools and structures necessary to generate information on malaria
transmission. cases and deaths. The information is used for planning,
implementing, monitoring and evaluating malaria programmes. An
Although surveillance guidance is not evaluated using the GRADE effective malaria surveillance system enables programme
framework, surveillance forms is the basis of operational activities managers to:
in settings at any level of transmission and is therefore included in
these Guidelines for reference. The objective of surveillance is to • identify and target areas and population groups most severely
support reduction of the burden of malaria, eliminate the disease affected by malaria, to deliver the necessary interventions
and prevent its re-establishment. In settings where transmission effectively and to advocate for resources;
remains relatively high and the aim of national programmes is to • regularly assess the impact of intervention measures and
reduce the burden of morbidity and mortality, malaria surveillance progress in reducing the disease burden and help countries to
is often integrated into broader routine health information systems decide whether adjustments or combinations of interventions
to provide data for overall analysis of trends, stratification and are required to further reduce transmission;
230 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
◦ detect and respond to epidemics in a timely way; specific packages of interventions to target different sub-
◦ provide relevant information for certification of populations. For example, case management should be accessible
elimination; and wherever there is a possibility of malaria cases seeking treatment.
◦ monitor whether the re-establishment of transmission has How case management is delivered will vary according to factors
occurred and, if so, guide the response. such as health-seeking behaviour, the accessibility and functioning
of the public health infrastructure, availability of the private retail
sector and the potential for community services. Local data are
Please refer to the WHO Malaria surveillance, monitoring &
essential to complete the malaria stratification and select the
evaluation: a reference manual [30].
optimal mixes of interventions. The guidance explains how to
undertake a comprehensive multi-indicator stratification process
Subnational stratification
to define sub-national intervention mixes that are optimized to
WHO has made guidance available on the strategic use of data to
achieve strategic goals. As countries will rarely have all the
inform subnational stratification (see chapter 2 of WHO technical
resources they need to fully implement their ideal plan, a careful
brief for countries preparing malaria funding requests for the Global
resource prioritization process is then required to maximize the
Fund (2020-2022)) [252]. This guidance was developed in
impact of available resources. Prioritization should be based on the
recognition of the increasing heterogeneity of malaria risk within
expected impact of interventions and consider value for money
countries as malaria control improves and the need to use
across the whole country, driven by local evidence.
problem-solving approaches to identify appropriate, context-
8. METHODS
The consolidated WHO Guidelines for malaria were prepared in proposal, including formulating key questions, as well as
accordance with WHO standards and methods for guideline suggesting potential members for the GDGs. Technical leads then
development and originally published as the Guidelines for the obtained declarations of interest from GDG members, assessed
treatment of malaria (3rd edition, 2015) and the Guidelines for these and oversaw the management of any potential conflicts of
interest, as well as the finalization and submission of a planning
malaria vector control (1st edition, 2019). Details of the approach
proposal to the Guidelines Review Committee (GRC) for review
can be found in the WHO Handbook for guideline development [1].
and approval.
Here we provide an overview of the standards, methods,
processes and platforms applied by the Global Malaria Programme
The GDGs - external bodies of experts and stakeholders - were
across the topics covered in this guideline [103][253][254] and a
responsible for the development of the evidence-based
description of the joint process (with WHO Immunization,
recommendations contained in the Guidelines. As well as providing
Vaccination and Biologicals department) used to develop the
expert opinion, the specific tasks of the GDGs included:
malaria vaccine recommendation.
• providing inputs on the scope of the Guidelines;
Organization and process
• building on the work of the Guidelines Steering Groups to
The WHO guideline development process involved planning;
finalize the key recommendation questions in PICO format;
conducting a “scoping” and needs assessment; establishing an
• choosing and ranking priority outcomes to guide the evidence
internal WHO Guidelines Steering Groups and external Guidelines
reviews and focus the recommendations;
Development Groups (GDGs); formulating key recommendation
• reviewing eligibility criteria for the inclusion of studies in the
questions using the PICO (Population, Intervention, Comparison,
evidence reviews;
Outcome) format; commissioning evidence reviews or where a
• providing input on appropriate measures of outcomes of
recent review was already available, commissioning an
interest to be included in the evidence reviews;
independent assessment of the review using the AMSTAR
• validating the list of included and excluded studies;
checklist [114]; applying GRADE (Grading of Recommendations
• reviewing the meta-analyses, GRADE evidence profiles or
Assessment, Development and Evaluation) methodology to assess
other assessments of the certainty of evidence used to inform
the certainty of evidence; and using evidence-to-decision (EtD)
the recommendations;
frameworks to take the GRADE results and contextual factors into
• interpreting the evidence, considering different factors
account in developing recommendations. This methodology
included in the EtD framework and judging how these factors
ensures that the link between the evidence base and the
may impact the direction and strength of a recommendation,
recommendations is transparent. The Guidelines have been
particularly in terms of the overall balance of benefits and
consolidated and will be continuously updated in the online
harms;
MAGICapp publication platform (www.magicapp.org) as new
• formulating recommendations, taking into account benefits,
evidence becomes available and published in user-friendly formats
harms, values and preferences, feasibility, equity,
available on all electronic devices.
acceptability, resource requirements, cost and cost-
effectiveness and other factors, as appropriate;
Technical leads in the Global Malaria Programme established
• identifying methodological shortcomings and evidence gaps
Guidelines Steering Groups for each technical area to support
in the available body of evidence, and providing guidance on
drafting the scope of the Guidelines and preparing the planning
231 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
how to address these as part of future research; of evidence into recommendations, and guideline development
• reviewing and approving the final recommendations prior to processes. The methodologists supported the planning, scoping
submission to the GRC; and and development of key questions and assisted the GDG in
• contributing to the dissemination of the final formulating evidence-informed recommendations in a transparent
recommendations. and explicit manner. The methodologists served as the
methodological co-chairs of some GDG meetings.
232 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
233 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
234 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
procedures and summarized at the beginning of each meeting to prior to their appointment in advance of each meeting. Review of
all participants. Where necessary, GDG members were excluded DOI forms revealed no relevant conflicts and all members
from the discussion and/or decision-making on topics for which participated in all discussions. Support for the closed sessions of
they had declared interests. The members of the GDGs and a the SAGE/MPAG Working Group’s full evidence review was
summary of their declarations of interest are listed in Section 10: provided by a restricted WHO Secretariat - known as the SAGE/
Contributors and Interests. MPAG Working Group Secretariat - composed of the IVB and
GMP Directors, and other staff who were not involved in the
Link to WHO prequalification generation or synthesis of evidence being reviewed by the MVIP
When a recommendation is linked to the introduction of a new Programme Advisory Group (see Section 10.2 Contributors –
tool or product, there is a parallel process managed by the WHO malaria vaccine).
Prequalification Team to ensure that diagnostics, medicines,
vaccines and vector control products meet global standards of The SAGE/MPAG Working Group performed the following
quality, safety and efficacy, in order to optimize use of health functions: developed relevant and answerable question(s) in PICO
resources and improve health outcomes. The prequalification format, reviewed and interpreted the evidence, with explicit
process consists of a transparent, scientifically sound assessment consideration of the overall balance of benefits and harms;
that, includes dossier review, consistency testing or performance examined and provided input to the GRADE evidence profiles
evaluation, and site visits to manufacturers. This information, in developed by the Cochrane Response; and formulated the
conjunction with other procurement criteria, is used by United proposed recommendations for SAGE/MPAG in alignment with
Nations and other procurement agencies to make purchasing the 2019 RTS,S Framework for WHO
decisions regarding these health products. This parallel process recommendation (unpublished evidence), taking into
aims to ensure that recommendations are linked to prequalified account benefits, harms, values and preferences, feasibility, equity,
products and that prequalified products are linked to a acceptability, resource requirements and other factors, as
recommendation for their use. appropriate.
Joint process for developing the malaria vaccine SAGE and MPAG were jointly convened on 6 October 2021 to
recommendation review the work of the SAGE/MPAG Working Group, to consider
the malaria vaccine evidence and to reach consensus
In order to enable joint decision-making on a malaria vaccine, the on their vaccine recommendations to the Director-General of
different guideline development processes of the Global Malaria WHO [257][258].
Programme and the WHO Department for Immunization, Vaccines
and Biologicals (IVB) were harmonized following discussion with Following the Director General's endorsement of the SAGE/MPAG
the WHO Department of Quality, Norms and Standards. The recommendations, the evidence and deliberations that informed
standard process for the development of WHO vaccine the WHO malaria vaccine position paper were put into the format
recommendations was used as the basis for developing the malaria required for the Weekly Epidemiological Record by the WHO
vaccine recommendation. The process employed by the Strategic Secretariat and reviewed by the a WHO Editorial Board as per the
Group of Experts (SAGE) on Immunization, described here, standard SAGE process. The draft was subject to broad peer
complies with the principles and requirements of the standard review. Reviewers included members of SAGE, WHO Regional
GRC process which is described above and used for the Offices, external subject matter experts, selected national
development of the WHO Guidelines for malaria. MPAG members immunization and malaria control programme managers, other
exceptionally participated in the guideline development process interested parties (who had not been involved in the process to
given their previous role in developing the malaria vaccine that point) and industry. Request for peer review from industry
recommendation in 2015 and because both advisory groups had was coordinated through the International Federation of
been kept up to date with the progress of the Malaria Vaccine Pharmaceutical Manufacturers Association and the Developing
Implementation Programme (MVIP). Country Vaccine Manufacturer Network.
A SAGE/MPAG Working Group was established with Terms of The final recommendation, GRADE and evidence-to-
Reference and an open call for members. The SAGE/MPAG decision frameworks, and other relevant components were
Working Group members (biographies are publicly accessible on included in the WHO Guidelines for malaria and submitted for GRC
the WHO Malaria Vaccine Implementation Programme website) review in parallel with the development of the WHO position
were required to complete a Declaration of Interest (DOI) form paper in the Weekly Epidemiologic Record.
9. GLOSSARY
The Glossary lists the terms contained in the WHO malaria contained here.
terminology 2021 update [259] which is reviewed and agreed by
the Drafting Committee on Malaria Terminology first convened in
2015. Please refer to that document for additional information on Compliance with a regimen
adherence
the Drafting Committee and the process to review and update (chemoprophylaxis or treatment) or with
malaria terminology and for more detailed notes on the glossary
235 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
236 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
A case contracted locally, with strong Compulsory reporting of all malaria cases by
epidemiological evidence linking it directly medical units and medical practitioners to
case, introduced
to a known imported case (first-generation case notification either the health department or the malaria
local transmission) control programme, as prescribed by
national laws or regulations
case, locally A case acquired locally by mosquito-borne
acquired transmission A geographical area defined and served by a
health programme or institution, such as a
Occurrence of malaria infection in a person
hospital or community health centre, which
in whom the presence of malaria parasites catchment area
case, malaria is delineated on the basis of population
in the blood has been confirmed by a
distribution, natural boundaries and
diagnostic test
accessibility by transport
Case suspected of being malaria that is not
case, presumed Severe P. falciparum malaria with impaired
confirmed by a diagnostic test
consciousness (Glasgow coma scale < 11,
cerebral malaria
Malaria case attributed to the recurrence of Blantyre coma scale < 3) persisting for > 1
asexual parasitaemia after antimalarial hour after a seizure
treatment, due to incomplete clearance of
Certification granted by WHO after it has
case, asexual parasitaemia of the same
been proved beyond reasonable doubt that
recrudescent genotype(s) that caused the original
local human malaria transmission by
illness. A recrudescent case must be
certification of Anopheles mosquitoes has been
distinguished from reinfection and relapse,
malaria-free interrupted in an entire country for at least
in the case of P. vivax and P. ovale
status three consecutive years and a national
Malaria case attributed to activation of surveillance system and a programme for
case, relapsing hypnozoites of P. vivax or P. ovale acquired the prevention of reintroduction are in
previously place
237 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Diagnosis of malaria by detection of malaria Portion of the life cycle of the malaria
diagnosis,
parasites or Plasmodium-specific antigens or parasite from merozoite invasion of red
parasitological
genes in the blood of an infected individual blood cells to schizont rupture. The
erythrocytic cycle
duration is approximately 24 h in P. knowlesi,
Condition of suspended animation or 48 h in P. falciparum, P. ovale and P. vivax,
diapause temporary arrest in the development of and 72 h in P. malariae.
immature and adult mosquitoes
exophagy Tendency of mosquitoes to feed outdoors
dosage regimen Prescribed formulation, route of
(or treatment administration, dose, dosing interval and exophily Tendency of mosquitoes to rest outdoors
regimen) duration of treatment with a medicine
For vector investigations, simulated house
Quantity of a medicine to be taken at one with entry and exit traps for sampling
dose
time or within a given period mosquitoes entering and exiting, blood-
experimental huts
feeding indoors (when a host is present),
One or a series of doses that may be given and surviving or dying in each sub-sample,
dose, loading at the start of therapy with the aim of per day or night
achieving the target concentration rapidly
A combination in which two antimalarial
Capacity of an antimalarial medicine to fixed-dose medicines are formulated together in the
achieve the therapeutic objective when combination same tablet, capsule, powder, suspension or
drug efficacy administered at a recommended dose, granule
which is well tolerated and has minimal
toxicity A defined circumscribed area situated in a
focus, malaria
currently or formerly malarious area that
238 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
contains the epidemiological and ecological Number of newly diagnosed malaria cases
factors necessary for malaria transmission incidence, malaria during a defined period in a specified
population
Sexual stage of malaria parasites that can
gametocyte potentially infect anopheline mosquitoes Period between inoculation of malaria
incubation period
when ingested during a blood meal parasites and onset of clinical symptoms
Censuses and mapping to determine the index, human Proportion of mosquito blood meals from
geographical distribution of the human population and blood humans
reconnaissance other features relevant for malaria
Mean parasite density on slides examined
transmission in order to guide interventions
index, parasite- and found positive for a sample of the
The period of reproductive development in density population; calculated as the geometric
the female mosquito, including host- mean of individual parasite density counts
gonotrophic cycle,
seeking, blood feeding, digestion of a blood
mosquito Operational procedure and strategy for
meal, ovarian development, search for a
malaria vector control involving spraying
breeding site and oviposition. indoor residual
interior surfaces of dwellings with a residual
spraying
gonotrophic insecticide to kill or repel endophilic
Female mosquitoes that take more than one
discordance mosquitoes
blood meal per gonotrophic cycle
(dissociation)
Inside any shelter likely to be used by
Process in which mosquitoes at one or indoors humans or animals, where mosquitoes may
several stages (eggs, larvae, pupae, adults) feed or rest
hibernation
survive by means of behavioural or
Long-term presence of parasitaemia that is
physiological changes during cold periods
infection, chronic not causing acute or obvious illness but
Any structure other than a tent or mobile could potentially be transmitted
house
shelter in which humans sleep
Malaria infection with more than one
infection, mixed
The ecosystem, including people and species of Plasmodium
household animals occupying the same house and the
Any person or animal in which Plasmodium
accompanying vectors infection,
species live and multiply, such that they can
reservoir of
Application of liquid insecticide formulation be transmitted to a susceptible host
house-spraying to specified (mostly interior) surfaces of
Low-density blood-stage malaria infections
buildings infection,
that are not detected by conventional
submicroscopic
human landing A method for collecting vectors as they land microscopy
catch on individuals
Capable of transmitting infection, a term
infectious
A high density of parasites in the blood, commonly applied to human hosts
which increases the risk that a patient’s
hyperparasitaemia Capable of producing infection, a term
condition will deteriorate and become
commonly applied to parasites (e.g.,
severe malaria infective
gametocytes, sporozoites) or to the vector
Persistent liver stage of P. vivax and P. ovale (mosquito)
malaria that remains dormant in host
Ability of sporozoites of a specific strain of
hepatocytes for variable periods, from three
Plasmodium to be injected by Anopheles
hypnozoite weeks to one year (exceptionally even
mosquitoes into susceptible humans and
longer), before activation and development
develop through the liver stage to infect red
into a pre-erythrocytic schizont, which then
blood cells ("infectivity to humans") and the
causes a blood-stage infection (relapse) infectivity
ability of competent Anopheles mosquitoes
Rate of influx of parasites via infected to ingest human Plasmodium gametocytes
importation rate individuals or infected Anopheles spp. which undergo development until the
mosquitoes mosquito has infective sporozoites in its
salivary glands ("infectivity to mosquitoes").
Probability of influx of infected individuals
importation risk
and/or infective anopheline mosquitoes insecticide Chemical product (natural or synthetic) that
239 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
kills insects. Ovicides kill eggs; larvicides a surface, maintains its insecticidal activity
(larvacides) kill larvae; pupacides kill pupae; residual for a considerable time by either contact or
adulticides kill adult mosquitoes. Residual fumigant action
insecticides remain active for an extended
integrated vector
period Rational decision-making for optimal use of
management
resources for vector control
Resistance to one insecticide by a (IVM)
mechanism that also confers resistance to
insecticide, cross- intermittent Administration of a full treatment course of
another insecticide, even when the insect
resistance preventive an antimalarial medicine at predefined
population has not been selected by
treatment of intervals to school children, in order to
exposure to the latter
malaria in school- prevent illness in areas with moderate to
Amount of an insecticide (usually expressed aged children high malaria transmission
insecticide as the concentration per standard period of
intermittent
discriminating exposure), which, in a sample of
preventive Please see ‘perennial malaria
dose, or mosquitoes containing resistant individuals,
treatment in chemoprevention’
diagnostic dose distinguishes between susceptible and
infants (IPTi)
for resistance resistant phenotypes and determines their
respective proportions intermittent A full therapeutic course of antimalarial
preventive medicine given to pregnant women at
Amount of active ingredient of insecticide
treatment in routine prenatal visits, regardless of
applied per unit area of treatment (mg/m2)
pregnancy (IPTp) whether the woman is infected with malaria
for indoor residual spraying and treated
insecticide, dose mosquito nets, or per unit of space (mg/m3) A non-native species that establishes in a
for space spraying and per unit area of new ecosystem, and causes, or has the
invasive species
potential to cause, harm to the
application (g/ha or mg/m2) or per volume
environment, economy, or human health
of water (mg/L) for larvicides
Management of aquatic habitats (water
Insecticide product consisting of two or
bodies) that are potential habitats for
insecticide, more active ingredients mixed as one larval source
mosquito larvae, in order to prevent
mixture formulation so that, when applied, the management
completion of development of the
mosquito will contact both simultaneously
immature stages
Strategy for mitigating resistance, whereby
larvicide Substance used to kill mosquito larvae
insecticides with different modes of action
are applied in different parts of an area For P. vivax and P. ovale infections, the
insecticide mosaic under coverage (usually in a grid pattern), so period between the primary infection and
that parts of the mosquito populations are subsequent relapses. This stage is
latent period
exposed to one insecticide and others to asymptomatic; parasites are absent from
another the bloodstream but present in
hepatocytes.
Property of mosquitoes to survive exposure
insecticide to a standard dose of insecticide; may be A factory-treated mosquito net made of
resistance the result of physiological or behavioural material into which insecticide is
adaptation incorporated or bound around the fibres.
long-lasting
The net must retain its effective biological
Strategy involving sequential applications of insecticidal net
insecticide activity for at least 20 WHO standard
insecticides with different modes of action (LLIN)
rotation washes under laboratory conditions and
to delay or mitigate resistance
three years of recommended use under
insecticide Less-than-average susceptibility to field conditions.
tolerance insecticide but not inherited as resistance
malaria case (See Case, malaria)
Insecticide that exerts a toxic action on
malaria, cerebral (See Cerebral malaria)
insecticide, mosquitoes when they rest on a treated
contact surface; the insecticide is absorbed via the Reduction of disease incidence, prevalence,
tarsi (feet). morbidity or mortality to a locally
malaria control acceptable level as a result of deliberate
insecticide, Insecticide that acts by releasing vapour
efforts. Continued interventions are
fumigant from a volatile substance
required to sustain control.
insecticide, Insecticide that, when suitably applied onto
malaria Interruption of local transmission (reduction
240 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
241 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Antimalarial treatment with a single active and P. vivax cause malaria in humans.
compound or a synergistic combination of Human infection with the monkey malaria
monotherapy
two compounds with related mechanisms of parasite P. knowlesi and very occasionally
action with other simian malaria species may occur
in tropical forest areas.
Centralized database of all foci of malaria
national focus infection in a country, which includes Population living in a geographical area
register relevant data on physical geography, population at risk where locally acquired malaria cases have
parasites, hosts and vectors for each focus occurred in the past three years
national malaria Centralized database with individual records An implementation unit targeted for
case register of all malaria cases registered in a country population, target activities or services (e.g., prevention,
treatment)
Mosquito net that repels, disables or kills
mosquitoes that come into contact with the Administration of a full treatment course of
insecticide on the netting material. post-discharge an antimalarial medicine to children
net, insecticide- Insecticide treated nets (ITNs) include those malaria hospitalized for severe anemia, starting at
treated (ITN) that require treatment and retreatment chemoprevention time of discharge from hospital and
(often referred to as conventional nets) and continuing at predefined intervals.
those that are “long-lasting” (see definition
Development of the malaria parasite from
of long-lasting insecticidal net). pre-erythrocytic
the time it first enters the host and invades
development
The stage of malaria parasite that develops liver cells until the hepatic schizont ruptures
from the ookinete; the oocyst grows on the
oocyst Period between inoculation of parasites and
outer wall of the midgut of the female pre-patent period
the first appearance of parasitaemia
mosquito.
Process to ensure that health products are
Percentage of female Anopheles mosquitoes
oocyst rate prequalification safe, appropriate and meet stringent quality
with oocysts on the midgut
standards for international procurement
Motile stage of malaria parasite after
Use of medicines either alone or in
ookinete fertilization of macrogamete and preceding preventive
combination to prevent malaria infections
oocyst formation chemotherapy
and their consequences
parasitaemia Presence of parasites in the blood
Any method of protection from or
parasitaemia, The presence of asexual parasites in the prevention of disease; when applied to
prophylaxis
asymptomatic blood without symptoms of illness chemotherapy, it is commonly termed
“chemoprophylaxis”.
Time between first drug administration and
parasite clearance
the first examination in which no parasites Complete prevention of erythrocytic
time prophylaxis,
are present in the blood by microscopy infection by destroying the pre-erythrocytic
causal
forms of the parasite
Number of asexual parasites per unit
parasite density volume of blood or per number of red blood Immunochromatographic lateral flow device
rapid diagnostic
cells for rapid detection of malaria parasite
test (RDT)
antigens
Presence of Plasmodium parasites in the
parasite density,
blood at parasite density below 100 rapid diagnostic Malaria rapid diagnostic test that can detect
low
parasites/μl test, combination a number of different malaria species
Period during which malaria parasitaemia is rapid diagnostic Proportion of positive results among all
patent period
detectable test positivity rate rapid diagnostic tests performed
242 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
243 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
244 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
destroy both blood and liver stages of the Measures of any kind against malaria-
parasite. vector control transmitting mosquitoes, intended to limit
their ability to transmit the disease
The stage of development of malaria
parasites growing within host red blood The degree to which a mosquito population
vector
cells from the ring stage to just before is susceptible (i.e., not resistant) to
trophozoite susceptibility
nuclear division. Trophozoites contain insecticides
malaria pigment that is visible by
The species of Anopheles mainly responsible
microscopy.
vector, principal for transmitting malaria in any particular
Symptomatic malaria parasitaemia without circumstance
uncomplicated
signs of severity or evidence of vital organ
malaria Species of Anopheles thought to play a
dysfunction
vector, secondary lesser role in transmission than the principal
In malaria, adult females of any mosquito or subsidiary vector; capable of maintaining malaria
species in which Plasmodium undergoes its transmission at a reduced level
sexual cycle (whereby the mosquito is the
Number of new infections that the
definitive host of the parasite) to the
vector population of a given vector would induce
infective sporozoite stage (completion of
vectorial capacity per case per day at a given place and time,
extrinsic development), ready for
assuming that the human population is and
transmission when a vertebrate host is
remains fully susceptible to malaria
bitten
A function of the public health services for
For malaria, the ability of the mosquito to
preventing reintroduction of malaria.
support completion of malaria parasite
Vigilance consists of close monitoring for
development after zygote formation and vigilance
any occurrence of malaria in receptive areas
vector oocyst formation, development and release
and application of the necessary measures
competence of sporozoites that migrate to salivary
to prevent re-establishment of transmission.
glands, allowing transmission of viable
sporozoites when the infective female
mosquito feeds again
245 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
246 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Research, Republic of India applications to evaluate new vector control tools in Africa; 4)
• Professor Dyann Wirth, Richard Pearson Strong Professor consulting with IVGGF from 2017 to February 2018 to provide
and Chair, Department of Immunology and Infectious technical support on developing guidelines for testing new
Diseases, Harvard T. H. Chan School of Public Health, vector control strategies, paid directly to Dr Burkot; 5) consulting
United States of America with PATH from 2017 to February 2018 to provide technical
advice on field trials for mosquito-proof housing products paid,
directly to Dr Burkot; 6) research support in a supervisory role
Systematic review production and management team and provided to James Cook University for evaluation of a new
GRADE analysis subgroup members (2019) malaria diagnostic test from October 2015 to March 2017; 7)
research support in a supervisory role provided to James Cook
• Mr Leslie Choi, Cochrane Infectious Diseases Group, University to undertake a malaria serologic survey in the
Liverpool School of Tropical Medicine, Liverpool, United Solomon Islands until June 2018; and 8) non-monetary support
Kingdom of Great Britain and Northern Ireland to Vestergaard in a supervisory role to evaluate the impact of
• Mr Joe Pryce, Cochrane Infectious Diseases Group, insecticide netting on malaria in Solomon Islands.
Liverpool School of Tropical Medicine, Liverpool, United
Kingdom of Great Britain and Northern Ireland Dr M. Coetzee reported a potential conflict of interest related to
• Ms Marty Richardson, Cochrane Infectious Diseases Group, a family member’s consulting work with AngloGold Ashanti in
Liverpool School of Tropical Medicine, Liverpool, United 2016 to carry out mosquito surveys and determine insecticide
Kingdom of Great Britain and Northern Ireland resistance in order to inform vector control strategies by gold
• Dr Vittoria Lutje, Cochrane Infectious Diseases Group, mining companies in Africa.
Liverpool School of Tropical Medicine, Liverpool, United
Professor M. Coosemans reported receiving a grant from the Bill
Kingdom of Great Britain and Northern Ireland
& Melinda Gates Foundation for studying the impact of
• Dr Deirdre Walshe, Cochrane Infectious Diseases Group,
repellents for malaria prevention in Cambodia and also reported
Liverpool School of Tropical Medicine, Liverpool, United
receiving repellent products for the study from SC Johnson for
Kingdom of Great Britain and Northern Ireland
work conducted in 2012–2014. He also reported receiving six
• Prof Paul Garner, Cochrane Infectious Diseases Group,
grants for the evaluation of public health pesticides from
Liverpool School of Tropical Medicine, Liverpool, United
WHOPES from 2007, some of which continued until 2018.
Kingdom of Great Britain and Northern Ireland
Dr J. Hii reported receiving remuneration for consulting services
from WHO and from the Ministry of Health of Timor-Leste for
Guidelines methodologist (2019) work conducted in 2017. He reported holding a grant from SC
Johnson that ceased in 2017 for the evaluation of transfluthrin,
Dr Joseph Okebe, Guidelines Methodologist, Disease Control and receiving travel and accommodation support from Bayer
and Elimination Team, Medical Research Council Unit, Republic Crop Science to attend the 4th Bayer Vector Control Expert
of the Gambia Meeting in 2017. He reported holding a WHO/TDR research
grant that focused on studying the magnitude and identifying
Declaration of interests (2019)
causes for residual transmission in The Kingdom of Thailand and
Participants in the technical consultations or sessions for Viet Nam (completed in 2018), and reported a plan to study the
development of the Guidelines reported relevant interests. The impact of socio-ecological systems and resilience (SESR)-based
declared interests, as per WHO regulations, were assessed by strategies on dengue vector control in schools and neighbouring
the WHO Secretariat, with support from the Office of household communities in Cambodia, which in November 2017
Compliance, Risk Management and Ethics as needed. WHO was was awaiting ethical approval.
of the opinion that these declarations did not constitute conflicts
Members of the Guidelines Development Group (GDG) (2021)
of interest and that the considered experts could participate in
the consultations on the Guidelines subject to the public • Dr Dorothy Achu, Programme manager, National Malaria
disclosure of their interests, which was conducted. Control Programme, Yaoundé, the Republic of Cameroon
• Prof Basil Brooke, Associate professor, University
The relevant declared interests are summarized as follows:
Witwatersrand/National Institute for Communicable
Dr T. Burkot reported several potential conflicts of interest Disease, Johannesburg, Republic of South Africa
related to consulting payments, research support and non- • Prof Ahmad Ali Enayati, Head, Medical Entomology
monetary support, as follows: 1) consulting with Intellectual Department, School of Public Health, Mazandaran
Ventures Global Good Fund (IVGGF), the non-profit arm of University of Medical Sciences, Sari, Islamic Republic of Iran
Intellectual Ventures Laboratory. Work was conducted from • Ms Mihirini Hewavitharane, Entomology Technical Manager,
October 2014 to March 2015 through James Cook University; 2) PMI VectorLink Project, Abt Associates, Phnom Penh,
consulting with IVGGF for a secondment in 2017 to develop a Kingdom of Cambodia
vector control strategy on mosquitoproof housing and methods • Dr Seth Irish, Research entomologist, Centers for Disease
to age-grade mosquitoes through James Cook University; 3) Control and Prevention, Atlanta, United States of America
consulting with the non-profit Programme for Appropriate • Prof Fang Jing, Director, Institute for Health Sciences,
Technology in Health (PATH) in 2017 to support grant Kunming Medical University, Yunnan Province, People’s
247 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Members of the Guidelines Steering Group (2021 & 2022) • Ms Kallista Chan Department of Disease Control, Faculty of
Infectious Tropical Diseases, London School of Hygiene and
• Dr Samira Al-Eryani, WHO Regional Office for the Eastern Tropical Medicine, London, United Kingdom of Great Britain
Mediterranean, Cairo, Arab Republic of Egypt and Northern Ireland
• Dr Haroldo Bezerra, WHO Regional Office for the Americas, • Mr Leslie Choi, Cochrane Infectious Diseases Group,
248 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
249 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
17(3): e1003055. https://doi.org/10.1371/journal.pmed.100305. Tanzania. This was a collaboration with Ifakara Health Institute,
She was also a guest editor for a Malaria Journal thematic series United Republic of Tanzania; Swiss Tropical & Public Health
on Housing and Malaria between 2015 and 2016. Institute, Swiss Confederation; Liverpool School of Tropical
Medicine, UK. This project investigated housing characteristics
Dr Tusting also was involved in studies and reviews related to that were associated with risk of mosquito biting but did not
larval source management (LSM) as a vector control tool but all evaluate the impact of housing modifications on malaria
these date to 2015 or earlier and she has not received any
support towards work on this topic since and so it was Caroline Jones: reported being a co-Investigator on a Wellcome
concluded that this did not constitute a conflict of interest. Trust Collaborative Award: Improving the efficacy of malaria
prevention in an insecticide resistant Africa which aimed to
It was determined that Dr Tusting could participate in all parts of investigate the factors limiting the efficacy of current tools to
the meeting except for decision-making with respect to prevent malaria, largely insecticide-treated nets, and to identify
recommendations related to housing improvements. the most cost effective, complementary interventions that would
drive malaria transmission towards zero. Although this project
Five members of the External Review Group reported relevant
could consider interventions under discussion by the ERG, it did
interests; it was assessed that all members could fully participate
not seek to systematically evaluate a particular tool. She also
as the remit of the Review Group was limited to identifying
reported being a co-investigator on a DfID/MRC/Wellcome
factual errors, providing clarity and commenting on implications
Trust Joint Global Health Trials funded project: Can improved
for implementation not changing the recommendations
housing provide additional protection against clinical malaria
formulated by the GDG. It was concluded that their expertise in
over current best practice? A household-randomised controlled
some of these areas would be valuable, particularly on
trial.
implementation considerations and factors to be considered
associated with gender and social determinants, equity, and Neil Lobo: reported being a co-principal investigator on
human rights. ‘Screening mosquito entry points into houses with novel long
lasting insecticidal netting to reduce indoor vector densities and
The relevant declared interests for the ERG are summarized as
mitigate pyrethroid resistance’ in collaboration with Durham
follows:
University.
Umberto D’Alessandro: reported receiving remuneration for the
No interests related to the topics of the meetings were disclosed
following activities which were topics of the meeting. He
by the methodologist or systematic review teams.
declared receiving research funding exceeding USD 5000 in the
last 4 years on three projects titled ‘Can improved housing Declaration of interests (2022)
provide additional protection against clinical malaria over current Members of the GDG, the ERG, the methodologist and members
best practice? A household-randomised controlled study. of systematic review teams who were commissioned to
Supported by the Joint Global Health Trial Scheme (Medical undertake reviews by WHO were requested to declare any
Research Council (MRC), Welcome Trust (WT), Department for interests related to the topic of the meeting. The declared
International Development (DfID)) and ‘Will raised buildings interests, as per WHO regulations, were assessed by the WHO
reduce malaria transmission in sub-Saharan Africa and keep Secretariat with support from the Office of Compliance, Risk
buildings cool?’ which is a collaboration with Durham University; Management and Ethics as needed.
and ‘Towards the end game: operational research on improving
rural housing in sub-Saharan Africa as a strategy to support The relevant declared interests for the GDG are summarized as
malaria elimination’ also a collaboration with Durham University. follows:
Jennifer Armistead: reported the following projects that she had Dr Lucy Tusting declared receiving remuneration for consulting
been involved in in the past 4 years, where funding exceeded services exceeding USD 5000 for WHO that ended in October
GBP 5000 and which concerned topics for discussion during the 2022. This agreement was for providing support and input into
meeting; Monitoring the deployment of PBO synergist ITNs in the development of the WHO Urban Malaria Framework. She
Ebonyi State, the Federal Republic of Nigeria, estimating also received research funding exceeding USD 5000 for a
coverage, and impact, funded by PMI; Impact of housing Medical Research Council (UK) fellowship that will continue until
modifications combined with piperonyl butoxide (PBO) long- November 2023. The fellowship is on the role of improved
lasting insecticidal nets (LLINs) on malaria burden in the Republic housing on malaria. She has also received a grant from the
of Uganda, a collaboration between CDC, London School of NovoNordisk Foundation that involves risk mapping of malaria
Hygiene & Tropical Medicine, UK and Infectious Disease and Aedes-borne diseases in Tanzania. The grant runs until
Research Collaboration, Kampala, The Republic of Uganda; 2026.
Determining the feasibility and effectiveness of larviciding,
funded by PMI collaboration with PATH. Charles Wondji declared receiving research support, including
grants, collaborations, sponsorships, and other funding from the
Maureen Coetzee: reported acting as supervisor for a PhD Innovative Vector Control Consortium (IVCC) exceeding USD
project to investigate whether integrated spatial information 5000. Ongoing studies aim to evaluate the entomological impact
tools could enable targeted urban planning interventions to of more recently developed indoor residual spraying (IRS)
control malaria and lymphatic filariasis in Dar es Salaam, products, dual active ingredient nets and pyrethroid-PBO nets
250 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
against insecticide-resistant mosquitoes. when sprayed on half the usual surface against natural
populations of Anopheles gambiae in Ghana, a small-scale field
Dr Josh Yukich declared receiving salary support from his pilot of Partial IRS with pirimiphos-methyl in households in
university through a project titled ‘New Nets’ to investigate the northern Ghana for Malaria Vector Control and evaluating the
cost and cost effectiveness of dual active ingredient nets and impact of attractive targeted sugar baits (ATSBs) and indoor
pyrethroid- PBO nets. He also declared supervising students residual spraying (IRS) in experimental huts.
engaged in the analysis of the effectiveness of IRS and he has
been engaged in similar analyses over the past several years Caroline Jones reported receiving research support within the
whilst being employed by Tulane university. He is acting as a last 4 years that exceeded USD 5000 for being a co-investigator
consultant for the University of California San Francisco to on UNITAID funded project: Broad One Health Endectocide-
design and develop data collection tools for a cost effectiveness based Malaria Intervention in Africa, for being a co-investigator
and willingness to pay study that involves topical repellents. on Wellcome Trust Collaborative Award: Improving the efficacy
of malaria prevention in an insecticide resistant Africa, for being
In summary, three members of the GDG declared potential a co-investigator on DfID/MRC/Wellcome Trust Joint Global
interests. Based on the detailed assessment of the information Health Trials funded project: Can improved housing provide
provided to WHO it was decided that Dr Lucy Tusting could additional protection against clinical malaria over current best
participate in all sessions, while Dr Josh Yukich was to be practice? A household-randomised controlled trial and lastly for
recused from the decision-making processes where the impact being a co-investigator on the Program for Appropriate
of dual active ingredient nets and topical repellents against Technology in Health (PATH) funded project: Dynamics of health
malaria were be determined and from the sessions where care utilization strategies in the context of RTS,S/AS01vaccine
recommendations were formulated. It was also concluded that introduction: a qualitative longitudinal study [in Kenya].
Prof Wondji was to be recused from the decision-making
processes where the impact of IRS and dual active ingredient Neil Lobo reported receiving research funding exceeding USD
nets against malaria were determined and from the sessions 5000 and/or non-monetary support valued at over USD 1000
where recommendations are formulated. overall within the last 4 years towards a project investigating
Spatial Repellent Products for Control of Vector-borne Diseases
The relevant declared interests for the ERG are summarized as by SC Johnson, and a project on innovative intervention for
follows: reducing outdoor malaria transmission by Widder Bros.
Dr Umberto D’Alessandro reported receiving renumeration for Melanie Renshaw reported receiving salary support exceeding
being a member of the external scientific advisory board for USD 5000 from the African Leaders Malaria Alliance.
Medicines for Malaria Venture until December 2018, travel
support for a meeting in Geneva in Sept 2017 and Oct 2018, In summary, five members of the ERG reported interests; it was,
and a donation of dihydroartemisinin piperaquine treatments for however, judged that none of these were relevant to the
malaria for a cluster randomized trial on mass drug recommendations under review and it was decided that all
administration from Guilin Pharma in 2018. He was also an members could fully participate particularly as the remit of the
investigator in a trial on the safety and efficacy of pyronaridine review group was limited to identifying factual errors, providing
artesunate in asymptomatic malaria-infected individuals. clarity and commenting on implications for implementation not
changing the recommendations formulated by the GDG. It was
Jennifer Armistead reported being employed by the US concluded that their expertise in some of these areas would be
President’s Malaria Initiative, who in turn has supported a valuable, particularly on implementation considerations and
number of projects in the past 4 years for which funding factors to be considered associated with gender and social
exceeded USD 5000 but for which she did not receive any determinants, equity, and human rights.
personal funding. The projects focused on the effect of indoor
residual spraying on Anopheles vector behaviors and their No interests related to the topics of the meetings were disclosed
impact on malaria transmission in the northern region of Ghana, by the methodologist or systematic review teams.
an evaluation of pirimiphos-methyl efficacy in experimental huts
251 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
emergency settings (4.2.6.2) • James Kelley, Technical Officer, Malaria and Neglected
Tropical Diseases, World Health Organization Regional
Office for the Western Pacific, Manila, Philippines
Members of the Guideline Development Group (2022) • Kim Lindblade, Team Lead, Elimination, Global Malaria
Programme, World Health Organization, Geneva,
• Professor Salim Abdulla, Chief Scientist, Ifakara Health Switzerland
Institute, United Republic of Tanzania (Male – Expertise: • Özge Tuncalp Mingard, Scientist, Sexual and Reproductive
Malaria research & policy-making) Health and Research, World Health Organization, Geneva,
• Dr Dorothy Achu, Manager, National Malaria Control Switzerland
Programme, Cameroon (Female – Expertise: Malaria control, • Laura Nic Lochlainn, Technical Officer, Immunizations,
end-user perspective, service-user, case management & Vaccines and Biologicals, World Health Organization,
chemoprevention) Geneva, Switzerland
• Professor Joseph Amon, Director, Office of Global, Dornsife • Sarah Marks, Consultant for the World Health Organization
School of Public Health, Drexel University, United States of supporting the Responsible Technical Officer
America (Male – Expertise: Human rights, epidemiology) • Abdisalan Noor, Team Leader, Information for Response,
• Dr Anup Anvikar, Scientist, ICMR-National Institute of Global Malaria Programme, World Health Organization,
Malaria Research, India (Male – Expertise: Malaria research, Geneva, Switzerland
drug resistance/AMR, malaria prevention) • Lynda Ozor, Malaria National Programme Officer, World
• Dr Matthew Coldiron (PDMC only), Medical Epidemiologist, Health Organization Country Office, Nigeria
Epicentre / Médecins Sans Frontières (MSF), USA / France • Charlotte Rasmussen, Technical Officer, Diagnostics,
(Male – Expertise: Malaria control in emergency / fragile Medicines & Resistance, World Health Organization,
situations) Geneva, Switzerland
• The late Dr Martin De Smet, Senior Health Advisor, • Lisa Rogers, Technical Officer, Nutrition and Food Safety,
Médecins Sans Frontières (MSF), Belgium (Male – Expertise: World Health Organization, Geneva, Switzerland
Malaria control in emergency/fragile situations) • Anthony Solomon, Medical Officer, Neglected Tropical
• Dr Corine Karema, Independent Consultant, African Leaders Diseases, World Health Organization, Geneva, Switzerland
Malaria Alliance (ALMA), Rwanda (Female – Expertise: • David Schellenberg (Responsible Technical Officer), Science
Malaria control) Advisor, Global Malaria Programme, World Health
• Professor Miriam Laufer, Director, Office of Student Organization, Geneva, Switzerland
Research, University of Maryland School of Medicine, • Jackson Sillah, Medical Officer, Tropical and Vector Borne
United States of America (Female – Expertise: Malaria drug Diseases, World Health Organization Regional Office for
resistance) Africa, Brazzaville, Congo
• Mrs Olivia Ngou, Executive Director, Impact Santé Afrique, • Neena Valecha, Regional Malaria Adviser, World Health
Cameroon (Female – Expertise: Civil society) Organization Regional Office for South-East Asia, New
• Professor Melissa Penny, Professor and Unit Head, Swiss Delhi, India
Tropical and Public Health Institute (Swiss TPH), Switzerland • Wilson Were, Medical Officer, Child Health and
(Female – Expertise: Mathematical modelling for malaria) Development, World Health Organization, Geneva,
• Dr Francisco Saute, Scientific Director, Manhiça Health Switzerland
Research Center (CISM), Mozambique (Male – Expertise:
Malaria control programming & research)
• The late Dr Samuel Smith, Manager, National Malaria Members of the External Review Group (2022)
Control Programme, Sierra Leone (Male – Expertise: Malaria
control programming) • Professor Umberto d’Alessandro, Director, Medical
• Dr Allan Schapira, Visiting Consultant, Bicol University Research Council Unit, Gambia (Malaria Policy Advisory
College of Medicine, Phillippines (Male – Expertise: Malaria Group [MPAG] member)
control and research) • Mrs Valentina Buj de Lauwerier, Global Malaria and Health
• Professor Robert Snow, Scientist, KEMRI-Wellcome Trust Partnerships Advisor, Health Section, Programme Division,
collaboration, Kenya (Male – Expertise: Malaria UNICEF, New York, United States of America
epidemiology & control) • Professor Graham Brown, Professor Emeritus (MDA, PMC
and SMC only), University of Melbourne, Australia
• Dr Caroline Jones, Senior Social Scientist, KEMRI-Wellcome
Members of the Guideline Steering Group (2022) Trust Research Programme, Kenya (MPAG member)
• Dr Estrella Lasry (MDA, PMC, IPTp, IPTsc, and PDMC only),
• Sheick Oumar Coulibaly, Technical Officer, Diagnostic and Senior Disease Advisor Malaria, Technical Advice and
Laboratory Services, World Health Organization Regional Partnerships Department, Global Fund to Fight AIDS,
Office for Africa, Brazzaville, Congo Tuberculosis and Malaria, Geneva, Switzerland
• Mary Hamel, Senior Technical Officer, Immunization, • Dr Sussann Nasr (SMC only), Senior Malaria Advisor, Global
Vaccines and Biologicals, World Health Organization, Fund to Fight AIDS, Tuberculosis and Malaria, Geneva,
Geneva, Switzerland Switzerland
252 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• Dr Harriet Pasquale, HIV/AIDS and STI Program Director, Prevention, Atlanta, United States of America
National Ministry of Health, Republic of South Sudan • Dr Rose Zulliger, President’s Malaria Initiative, United States
• Dr Richard Steketee, Deputy Coordinator, U.S. President’s Agency for International Development, Washington DC,
Malaria Initiative (PMI), United States of America United States of America
253 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• Dr Julie Thwing, Malaria Branch, Division of Parasitic of Tropical Medicine, United Kingdom of Great Britain and
Diseases and Malaria, Centers for Disease Control and Northern Ireland
Prevention, Atlanta, United States of America
Declaration of interests (2022)
Members of the Guideline Development Group (GDG) were
requested to declare any interests related to the topic of the
MDA for burden reduction
meeting. The declared interests, as per WHO regulations, were
• Marisa Boily, Rollins School of Public Health, Emory assessed by the WHO Secretariat with support from the Office
University, Atlanta, United States of America of Compliance, Risk Management and Ethics as needed.
• Alexandra Busbee, Rollins School of Public Health, Emory
The relevant declared interests for the GDG are summarized as
University, Atlanta, United States of America
follows:
• Dr Julie Gutman, Malaria Branch, Division of Parasitic
Diseases and Malaria, Centers for Disease Control and Professor Salim Abdulla declared two chemoprevention research
Prevention, Atlanta, United States of America support grants his institute receives; he is involved in one of the
• Dr Jimee Hwang, U.S. President’s Malaria Initiative, Malaria studies as a technical advisor. The interests were assessed as
Branch, Division of Parasitic Diseases and Malaria, Centers related to the overall topic of discussion on malaria
for Disease Control and Prevention, Atlanta, United States chemoprevention, with one interest directly related to the topic
of America of PMC. One interest was considered non-personal in nature,
• Dr Monica Shah, Malaria Branch, Division of Parasitic academic and financially significant, and the other was
Diseases and Malaria, Centers for Disease Control and considered personal in nature, academic and financially
Prevention, Atlanta, United States of America insignificant. Professor Abdulla was allowed to join the
• Zachary Schneider, Malaria Branch, Division of Parasitic discussions as a full member of the GDG.
Diseases and Malaria, Centers for Disease Control and
Prevention, Atlanta, United States of America Professor Joseph Amon declared a research support grant a
previous employer received to fund activities related to MDA/
chemoprevention for other diseases. This interest was not
MDA for burden reduction in emergency settings current and of a non-personal nature. He was allowed to join the
discussions as a full member of the GDG.
• Dr Alaine Knipes, Malaria Branch, Division of Parasitic
Diseases and Malaria, Centers for Disease Control and The late Dr Martin De Smet declared his employment with an
Prevention, Atlanta, United States of America organization that is involved in the use of chemoprevention. This
• Dr Leah Moriarty, Malaria Branch, Division of Parasitic interest was of a non-personal nature and financially significant.
Diseases and Malaria, Centers for Disease Control and He was allowed to join the discussions as a full member of the
Prevention, Atlanta, United States of America GDG.
• Dr Dean Sayre, Malaria Branch, Division of Parasitic
Diseases and Malaria, Centers for Disease Control and Professor Miriam Laufer declared four research grants. The
Prevention, Atlanta, United States of America interests were assessed as related to the overall topic of
• Dr Monica Shah, Malaria Branch, Division of Parasitic discussion on malaria chemoprevention, with one interest
Diseases and Malaria, Centers for Disease Control and directly related to the topic of IPT during pregnancy and another
Prevention, Atlanta, United States of America interest directly related to the topic of IPT in school children.
• Dr Nelli Westercamp, Malaria Branch, Division of Parasitic The four interests were considered non-personal in nature,
Diseases and Malaria, Centers for Disease Control and academic, and financially significant. Professor Laufer was also
Prevention, Atlanta, United States of America senior author for the systematic review on IPT in school children
that was considered by the GDG, although she did not
contribute empirical data to the review. The systematic review
Preparation of background papers (2022) on IPT in school children was subjected to a third-party AMSTAR
assessment and found to be of good quality. Professor Laufer
• Mr Emmanuel Bache-Bache, Centre for Tropical Medicine
was allowed to join all GDG discussions as a full member, but
and Travel Medicine, Amsterdam UMC, University of
was a non-voting and non-chairing participant in discussions on
Amsterdam, The Netherlands
IPT in school children.
• Professor Martin Grobusch, Head, Centre for Tropical
Medicine and Travel Medicine, Amsterdam UMC, University Professor Melissa Penny declared financial research support
of Amsterdam, The Netherlands received by her institute related to the overall topic of discussion
• Dr Jasper Littmann, Associate Professor, Bergen Centre for on malaria chemoprevention, and grants that she held on the
Ethics and Priority Setting, University of Bergen, Norway broader subject of malaria. These interests were assessed as
• Dr Christopher Plowe, University of Maryland School of financially significant, of a non-personal nature and academic.
Medicine, Baltimore, United States of America She was able to join the discussions as a full member of the
GDG.
254 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
studies on malaria. This interest was considered non-personal in the discussions on malaria chemoprevention. He was allowed to
nature, academic and financially significant. He was allowed to join the discussions as a full member of the GDG.
join the discussions as a full member of the GDG.
Professor Robert Snow declared his employment and funding for
Dr Allan Schapira declared his role as Member of a Board of studies on various aspects of malaria but not specifically
Trustees for an organization working on malaria. He did not chemoprevention. This interest was considered non-personal in
receive any remuneration for this role. This interest was assessed nature, academic and financially significant. He was allowed to
as financially insignificant and of a personal nature. Dr Schapira’s join the discussions as a full member of the GDG.
position on the Board of Trustees was not seen to interfere with
255 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Members of the RTS,S/AS01 MPAG/SAGE Working Group Members of the Peer review group (External review group)
• Professor Ifedayo Adetifa, KEMRI-Wellcome Trust Research Members of the Peer review group include SAGE, MPAG, WHO
Programme, Kenya Regional Offices, external subject matter experts, selected
• Professor Nick Andrews, Public Health England, United national immunization and malaria programme managers, other
Kingdom of Great Britain and Northern Ireland interested parties (who have not been involved in the process to
• Dr Dafrossa Cyrily Lyimo, Independent consultant (and that point) and industry. Request for peer-review from industry
former National Immunization and Vaccine Development is coordinated through the International Federation of
Programme Manager), United Republic of Tanzania Pharmaceutical Manufacturers Association and the Developing
• Dr Corine Karema, Independent consultant (and former Country Vaccine Manufacturer Network. The list of external
Director of the Rwanda National Malaria Control reviewers is available upon request from the SAGE secretariat.
Programme), Rwanda
• Dr Eusebio Macete, Centro de Investigação em Saúde de Guidelines methodologist and systematic review team
Manhiça, Mozambique (Co-Chair) Two methodologists from the Cochrane Response – Gemma
• Professor Kim Mulholland, Murdoch Children’s Research Villanueva and Nicholas Henschke – were commissioned to
Institute, Australia support the development of the malaria vaccine
• Professor Kathleen Neuzil, Center for Vaccine Development recommendations. They provided a systematic review of
and Global Health (CVD), University of Maryland School of evidence, applied the PICO framework to conduct evidence
Medicine, United States of America assessments using GRADE, and supported the SAGE/MPAG
• The late Ms Adelaide Shearley, John Snow Inc., Zimbabwe Working Group in the transparent formulation of evidence-
• Professor Peter Smith, London School of Hygiene & Tropical informed recommendations.
Medicine, United Kingdom of Great Britain and Northern
Designated writer/editor
Ireland (Chair)
• Professor S. Patrick Kachur, Mailman School of Public Dr Laurence Slutsker drafted and consolidated a full evidence
Health, Columbia University, United States of America review for the SAGE/MPAG Working Group. WHO contracted
Dr Slutsker under an Agreement for Performance of Work
(APW).
Members of the RTS,S/AS01 MPAG/SAGE Working Group
Declaration of interests
Secretariat
All nine SAGE/MPAG Working Group members updated their
• Dr Pedro Alonso, Global Malaria Programme
Declarations of Interest in advance of the meeting. These were
• Dr Tracey Goodman, Expanded Programme on
assessed by the WHO Secretariat. Six members reported
Immunization
interests; it was assessed that all members could fully
• Mr John Grove, Quality Assurance / Norms & Standards
participate. The full summary of interests for the SAGE/MPAG
• Dr Joachim Hombach, Agenda, Policy & Strategy
Working Group is available on the WHO Malaria Vaccine
• Dr Melanie Marti, Agenda, Policy & Strategy
Implementation Programme website.
• Dr Kate O’Brien, Immunization, Vaccines and Biologicals
• Dr Vaseeharan Sathiyamoorthy, Research for Health All 15 SAGE members participating in the meeting updated their
• Rapporteur: Ms Cynthia Bergstrom, Consultant for WHO Declarations of Interest in advance of the meeting. These were
assessed by the WHO Secretariat. Eleven SAGE members
reported interests and zero SAGE members recused themselves
Members of the WHO Editorial Board from the discussion and decision-making during the malaria
vaccine session. The full summary of interests for SAGE
• Dr Madhava Ram Balakrishnan members is available on the meeting website.
• Dr Shalini Desai, Expanded Programme on Immunization
• Ms Eliane Furrer, Immunization, Vaccines and Biologicals All 17 MPAG members participating in the meeting updated
• Ms Tracey Goodman, Expanded Programme on their Declarations of Interest in advance of the meeting. These
Immunization were assessed by the WHO Secretariat. Thirteen members
• Dr Mary Hamel, Immunization, Vaccines and Biologicals reported interests and five MPAG members reported relevant
• Dr Joachim Hombach, Immunization, Vaccines and interests. Three members (Evelyn Ansah, Abdoulaye Djimde and
Biologicals Azra Ghani) recused themselves from the discussion and
• Dr Dianliang Lei, Norms, Standards and Biologicals decision-making during the malaria vaccine session. It was
• Dr Melanie Marti, Immunization, Vaccines and Biologicals assessed that the remaining members could fully participate in
• Dr Marie-Perre Preziosi, Immunization, Vaccines and all sessions. The full summary of interests for MPAG members is
Biologicals available on the meeting website.
• Dr David Schellenberg, Global Malaria Programme
• Ms Erin Shutes, Global Malaria Programme • Professor Evelyn Ansah, University of Health & Allied
Sciences, Ghana: declared research support and her role as
256 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
the Ghana Co-Investigator on funding from PATH for the years (current). Included data analysis and modelling of
Health Utilization Study on a qualitative assessment of the public health impact of routine implementation and
pilot implementation of RTS,S. This interest was assessed as assessment of seasonal implementation related to RTS,s.
non-personal, specific and financially significant. This interest is assessed as non-personal, specific and
• Professor Abdoulaye Djimde, Head, Molecular financially significant.
Epidemiology Drug Resistance Unit, University of Mali, Mali: • Professor Caroline Jones, Senior Social Scientist, KEMRI-
declared his role as a sub-investigator on the RTS,S – SMC Wellcome Trust Research, Kenya: declared her role as a
trial which contributed minimal salary support through the mentor for a post-doctorate student on the study
London School of Tropical Medicine & Hygiene. Professor conducted in collaboration with PATH entitled “Dynamics
Djimde was a co-author on: Chandramohan D, Zongo I, of health care utilization strategies in the context of RTS,S/
Sagara I, Cairns M, Yerbanga RS, Diarra M, Nikièma F, Tapily AS01 vaccine introduction: a qualitative longitudinal study
A, Sompougdou F, Issiaka D, Zoungrana C, Sanogo K, Haro in Kenya” (2018- 2020). The institution received support for
A, Kaya M, Sienou AA, Traore S, Mahamar A, Thera I, Diarra the post-doc who has now left the institution. This interest
K, Dolo A, Kuepfer I, Snell P, Milligan P, Ockenhouse C, is assessed as non-personal, specific and non-financially
Ofori-Anyinam O, Tinto H, Djimde A, Ouédraogo JB, Dicko significant.* Although specific to the malaria vaccine, the
A, Greenwood B. Seasonal Malaria Vaccination with or interest was assessed as that of the post-doc, not Professor
without Seasonal Malaria Chemoprevention. N Engl J Med. Jones as the mentor.
2021 Sep 9;385(11):1005-1017. doi: 10.1056/ • Professor Dyann Wirth, Richard Pearson Strong Professor
NEJMoa2026330. Epub 2021 Aug 25. PMID: 34432975. and Chair, Harvard T.H. Chan School of Public Health, USA:
This interest is assessed as non-personal, specific and declared a research grant to Harvard University received
financially significant. from PATH and her role as the Principal Investigator for
• Professor Azra Ghani, Infectious Diseases Epidemiology, Mal095 using RTS,S to look at the issue of allele specific
Imperial College, UK: declared research support to Imperial immunity. This interest is assessed as non-personal, specific
College from the Global Fund on different projects related and financially significant.* Although related to the malaria
to modelling impact estimates for malaria including global vaccine, this work is to understand gaps in immunity
scenarios that incorporate RTS,s from 2016 to 2019 and in provided by RTS,S and will inform the development of
2021. This interest is assessed as non-personal, specific and future malaria vaccines. This research is not being
financially significant; and research support funding from considered as evidence for the decision on the malaria
multiple organizations for work on malaria and COVID-19 vaccine and the work will continue regardless of the
research including Bill & Melinda Gates Foundation (BMGF), outcome.
MVI, MMV, IVCC, MRC, Wellcome Trust, NIH over three
257 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
of pharmacokinetics and simulations of dosing, they used raw • Professor K. Barnes, (co-Chair)
clinical and laboratory data from the Worldwide Antimalarial • Professor F. Binka
Resistance Network on the concentrations of antimalarial agents • Dr S. Lutalo
in plasma or whole blood measured with validated assays in • Dr E. Juma
individual patients. The data had either been included in peer- • Professor O. Mokuolu
reviewed publications or been submitted to regulatory • Dr S. Parikh, Department of Medicine, Yale University
authorities for drug registration. Population pharmacokinetics School of Public Health, Connecticut, USA
models were constructed, and the plasma or whole blood • Dr D. Sinclair
concentration profiles of antimalarial medicines were simulated • Dr J. Tarning, Faculty of Tropical Medicine, Mahidol
(typically 1000 times) for different weight categories. University, Bangkok, Thailand
• Dr D.J. Terlouw, Malawi-Liverpool Wellcome Trust Clinical
The GDG met in two technical meetings, in November 2013 and Research Programme, Blantyre, Malawi
June 2014, to develop and finalize recommendations based on • Professor N. White (co-Chair)
the GRADE tables constructed on the basis of answers to the
PICO questions. The Guidelines were written by a subcommittee
Guideline Steering Group
of the group. At various times during preparation of the
Guidelines, sections of the document or recommendations were • Dr A. Bosman, Global Malaria Programme, WHO, Geneva,
reviewed by external experts and users who were not members Switzerland
of the group; these external peer reviewers are listed • Dr K. Carter, Malaria Regional Adviser, WHO Regional
below. Treatment recommendations were agreed by consensus, Office for the Americas, Washington D.C., United States of
supported by systematic reviews and review of information on America
pharmacokinetics and pharmacodynamics. Areas of • Dr N.Dhingra-Kumar, Health Systems Policies and
disagreement were discussed extensively to reach consensus; Workforce, WHO, Geneva, Switzerland
voting was not required. • Dr M. Gomes, Special Programme for Research and Training
in Tropical Diseases, WHO, Geneva, Switzerland
Members of the GDG • Dr P.E. Olumese (Secretary), Global Malaria Programme
WHO, Geneva, Switzerland
• Professor K.I. Barnes, Division of Clinical Pharmacology,
• Dr F. Pagnoni, Special Programme for Research and Training
University of Cape Town, South Africa
in Tropical Diseases, WHO, Geneva, Switzerland
• Professor F. Binka, (co-Chair), University of Health and Allied
• Dr A.E.C. Rietveld, Global Malaria Programme WHO,
Sciences, Ho, Volta Region, Ghana
Geneva, Switzerland
• Professor A. Bjorkman, Division of Infectious Diseases,
• Dr P. Ringwald, Global Malaria Programme WHO, Geneva,
Karolinska University Hospital, Stockholm, Sweden
Switzerland
• Professor M.A. Faiz, Dev Care Foundation, Dhaka,
• Dr M. Warsame, Global Malaria Programme WHO, Geneva,
Bangladesh
Switzerland
• Professor O. Gaye, Service de Parasitologie, Faculté de
• Dr W. Were, Child and Adolescent Health, WHO, Geneva,
Médicine, Université Cheikh Anta Diop, Dakar-Fann,
Switzerland
Senegal
• Dr S. Lutalo, King Faisal Hospital, Kigali, Rwanda
• Dr E. Juma, Kenya Medical Research Institute, Centre for External reviewers
Clinical Research, Nairobi, Kenya
• Dr A. McCarthy, Tropical Medicine and International Health • Dr F. ter-Kuile, Liverpool School of Tropical Medicine,
Clinic, Division of Infectious Diseases, Ottawa Hospital Liverpool, United Kingdom
General Campus, Ottawa, Canada • Dr R. McGready, Shoklo Malaria Research Unit, Faculty of
• Professor O. Mokuolu, Department of Paediatrics, Tropical Medicine, Mahidol University, Bangkok, Thailand
University of Ilorin Teaching Hospital, Ilorin, Nigeria • Professor F. Nosten, Shoklo Malaria Research Unit, Faculty
• Dr D. Sinclair, International Health Group, Liverpool School of Tropical Medicine, Mahidol University, Bangkok, Thailand
of Tropical Medicine, Liverpool, United Kingdom
• Dr L. Slutsker, Centers for Disease Control and Prevention,
Guidelines methodologist
Atlanta, Georgia, United States of America
Professor P. Garner, Liverpool School of Tropical Medicine,
• Dr E. Tjitra, National Institute of Health and Development,
Liverpool, United Kingdom
Ministry of Health, Jakarta, Indonesia
• Dr N. Valecha, National Institute of Malaria Research, New
Declaration of interests
Delhi, India
Participants in the technical consultation for the review of the
• Professor N. White (co-Chair), Faculty of Tropical Medicine,
Guidelines for the treatment of malaria and the external expert
Mahidol University, Bangkok, Thailand
reviewers of the Guidelines reported relevant interests, in
accordance with WHO procedures. These were discussed
Members of the sub-group on dose recommendations extensively by the committee. Although it was considered that
none of the declared interests had direct relevance to the
258 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
deliberations or recommendations of the meeting, the panel • Professor Philip Rosenthal, Professor, Department of
members with declared interests were excluded from the Medicine, University of California, United States of America
subcommittees on GRADE and recommendations and the • Professor Terrie Taylor, Professor, Department of
drafting group. The declared interests, as per WHO regulations, Osteopathic Medical Specialties, College of Osteopathic
were reviewed through the Legal Department of WHO. Medicine, Michigan State University, United States of
America
Dr K. Barnes reported being a grants co-recipient from the
Medicines for Malaria Venture to undertake clinical trials to
evaluate antimalarial medicines. Members of the Guidelines Steering Group (2022)
Dr F. Binka reported being a member of the INDEPTH network • Andrea Bosman, Coordinator, Global Malaria Programme,
that was a recipient of a research grant from the Bill & Melinda World Health Organization, Geneva, Switzerland
Gates Foundation to conduct Phase IV post licensure studies on • Maurice Bucagu, Family, Women, Children and Adolescents,
“Euratesim”. World Health Organization, Geneva, Switzerland
• Maria Bustos, Technical Officer, Malaria Control, WHO
Dr P. Garner reported receiving a grant from the Department for Country Office, Thailand
International Development (UK) to help ensure global guidelines • Jane Cunningham, Technical Officer, Global Malaria
and decisions are based on reliable evidence. Programme, World Health Organization, Geneva,
Switzerland
Dr N. Valecha reported serving as an investigator for a clinical • Bayo Fatunmbi, Technical Officer, Malaria, WHO Country
trial supported by the Department of Science and Technology Office, Uganda
India, and Ranbaxy Laboratories Limited. There were no • Elizabeth Juma, WHO Country Office, Ghana
monetary benefits and no conflicts with the subject of this • Roberto Montoya, Regional Malaria Adviser, WHO Regional
review. Office for the Americas, United States of America
• Deus Mubangizi, Coordinator, Prequalification Regulation
Professor N. White reported being an advisor to all and Prequalification, Access to Medicines and Health
pharmaceutical companies developing new antimalarial Products, World Health Organization, Geneva, Switzerland
medicines. This is done on a pro bono basis; it did not include • Peter Olumese, (Secretary) Medical Officer, Global Malaria
consultancy fees or any form of remuneration. Programme, World Health Organization, Geneva,
Switzerland
Members of the GDG (2022) • Shanthi Pal, Technical Officer, Pharmacovigilance,
Regulation and Safety/ Regulation and Prequalification,
• Dr Dorothy Achu, Programme Manager, National Malaria
World Health Organization, Geneva, Switzerland
Control Programme, Yaoundé, Cameroon
• Pascal Ringwald, Medical Officer, Global Malaria
• Professor Karen Barnes, Clinical Pharmacology, University
Programme, World Health Organization, Geneva,
of Cape Town, South Africa
Switzerland
• Dr Constance Bart-Plange, Independent Malaria Consultant,
• Neena Valecha, Regional Malaria Adviser, WHO Regional
Accra, Ghana
Office for South-East Asia, India
• Professor Adrianus Dondorp, Deputy Director, Mahidol
• Wilson Were, Medical Officer, Child Health and
Oxford Tropical Medicine Research Unit, Mahidol
Development HQ/UHL/MCA/CHD, Maternal, Newborn,
University, Thailand
Child and Adolescent Health and Ageing, World Health
• Professor Sanjeev Krishna, (Co-chair) Molecular
Organization, Geneva, Switzerland
Parasitology and Medicine, St George’s University London,
• Ghasem Zamani, Regional Malaria Adviser, WHO Regional
United Kingdom of Great Britain and Northern Ireland
Office for the Eastern Mediterranean, Egypt
• Professor Marcus Lacerda, Infectious Disease Researcher,
Tropical Medicine Foundation
• Dr Miriam Laufer, Director, Office of Student Research, Members of the External Review Group (ERG) (2022)
University of Maryland School of Medicine, United States of
America • Professor Ahmed Adeel, Independent Consultant, United
• Professor Rose Leke, Professor, Immunology and States of America
Parasitology, Faculty of Medicine and Biomedical Sciences, • Professor Umberto d’Alessandro, Director, Medical
University of Yaoundé, Cameroon Research Council Unit, Gambia
• Professor Olugbenga Mokuolu, (Co-chair) Professor,
Paediatrics & Child Health, Department of Paediatrics,
College of Health Sciences, University of Ilorin, Nigeria Members of the Systematic Review Team members (2022)
• Professor Bernhards Ogutu, Senior Principal Clinical
Research Scientist, Kenya Medical Research Institute, Kenya • Stephanie Dellicour, Liverpool School of Tropical Medicine,
• Professor Stephen Rogerson, Professor, Department of United Kingdom of Great Britain and Northern Ireland
Infectious Diseases, Doherty Institute, University of • Martha Chaplin, Liverpool School of Tropical Medicine,
Melbourne, Australia Centre for Evidence Synthesis, United Kingdom of Great
259 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Britain and Northern Ireland receiving around US$ 3000 per month, and her institution
• Paul Garner, Cochrane Infectious Diseases Group, Liverpool University of Cape Town is currently running projects on
School of Tropical Medicine, Centre for Evidence Synthesis, antimalarial pharmacology translational research with funding
United Kingdom of Great Britain and Northern Ireland from Bill & Melinda Gates Foundation and the South African
• Patricia Graves, College of Public Health Medical & Vet Ministry of Health of around US$ 450 000. She is not a direct
Sciences, Australian Institute of Tropical Health and beneficiary of this funding. Although the funding was for studies
Medicine, Australia with antimalarial medicines, , it is not related to the subject of
• Jenny Hill, Liverpool School of Tropical Medicine, United this Guideline. It was judged that none of these research projects
Kingdom of Great Britain and Northern Ireland or appointments had any direct relationship to the agenda of this
• Paul Hine, Liverpool School of Tropical Medicine, United meeting. These declarations were assessed as non-personal and
Kingdom of Great Britain and Northern Ireland non-specific, and of no direct or indirect personal or financial
• Feiko ter Kuile, Liverpool School of Tropical Medicine, benefit; therefore, they were considered non-significant
United Kingdom of Great Britain and Northern Ireland
• Makoto Saito, WorldWide Antimalarial Resistance Network, Professor Arjen Dondrop declared that, starting in February
Institute of Medical Science, University of Tokyo, Japan 2021, he has chaired the Malaria Advisory Council of Novartis,
• Kasia Stepniewska, WorldWide Antimalarial Resistance but he does not receive remuneration for this consultancy. In
Network, Infectious Disease Data Observatory, Oxford, 2014, his research group received a grant from Guilin Pharma
United Kingdom of Great Britain and Northern Ireland (now Fosun) to study intravenous artesunate. His group is
• Melissa Taylor, Liverpool School of Tropical Medicine, currently investigating the efficacy and safety of triple ACTs for
United Kingdom of Great Britain and Northern Ireland uncomplicated falciparum malaria in the context of increasing
• Rebecca Thomas, Cochrane Infectious Diseases Group, antimalarial drug resistance. One of the triple ACTs, artemether-
Liverpool School of Tropical Medicine, United Kingdom of lumefantrine-amodiaquine, is provided for free by Fosun
Great Britain and Northern Ireland Pharma. These declarations were assessed as not directly related
to any of the medicines being discussed by this GDG. These
declarations were assessed as non-personal and non-specific,
and of no direct or indirect personal or financial benefit;
Guidelines methodologist and co-chair (2022)
therefore, they were considered non-significant.
Leonila Dans, Independent Methodologist and World Health
Professor Lacerda declared institutional grants from Medicines
Organization consultant, Philippines
for Malaria Venture and Bill & Melinda Gates Foundation to
Declarations of interest (2022) study the operational feasibility of appropriate P. vivax radical
Members of the GDG, the ERG, the methodologist and members cure with tafenoquine or primaquine. These interests were
of systematic review teams who were commissioned to assessed as being of no direct or indirect personal or financial
undertake reviews by WHO were requested to declare any benefit; therefore, they were considered non-significant.
interests related to the topic of the meeting. The declared
Professor Terrie Taylor declared serving on two advisory boards
interests, as per WHO regulations, were assessed by the WHO
of Novartis AG and receiving US$ 3125 in 2019; her University
Secretariat with support from the Office of Compliance, Risk
and research group has received several rounds of research
Management and Ethics as needed. Below is a summary of the
funding from the United States National Institutes of Health.
declared interests of members.
None of these were directly related to the considerations of the
Professor Karen Barnes declared that she is currently serving as GDG, and it was determined that she could join the discussions
staff with Oxford University in the WorldWide Antimalarial of the group. These interests were assessed as non-personal and
Resistance Network, Infectious Disease Data Observatory, non-specific.
10.5 Recommendations for interventions in the final phase of elimination and prevention of
re-establishment
The following outlines the constitution of the Guidelines • Mass drug administration for reduction of transmission of P.
Development Group, Guidelines Steering Group, and External falciparum in moderate to high transmission settings
Review Group for the recommendations listed below, published (4.2.6.4)
in 2022. Also indicated are members of the systematic review • Mass drug administration for reduction of transmission of P.
production and management team as well as the guidelines vivax (4.2.6.5)
methodologist. Final compositions of these groups are shown as • Mass relapse prevention to reduce transmission of P. vivax
of the date of finalization of the Guidelines. (4.2.6.6)
• Mass testing and treatment (6.1.1)
Recommendations • Targeted drug administration (6.2.1)
• Targeted testing and treatment (6.2.2)
• Mass drug administration for reduction of transmission of P.
• Targeted testing and treatment at points of entry (6.2.3)
falciparum in very low to low transmission settings (4.2.6.3)
260 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
• Reactive drug administration (6.3.1) Migration Programme, World Health Organization, Geneva,
• Reactive case detection and treatment (6.3.2) Switzerland
• Reactive indoor residual spraying (6.3.3) • Dr Mika Kawano, Technical Officer, Border Health Risk
Dissemination, World Health Organization, Geneva,
Switzerland
Members of the Guidelines Development Group (2022) • Dr James Kelley, Technical Officer, Malaria and Neglected
Tropical Diseases, WHO Regional Office for the Western
• Dr Jane Achan, Senior Research Advisor, Malaria Pacific, Manila, Philippines
Consortium, United Kingdom of Great Britain and Northern • Dr Jan Kolaczinski, Head, Vector Control and Insecticide
Ireland (Female – Expertise: Malaria control and case Resistance Unit, Global Malaria Programme, World Health
management) Organization, Geneva, Switzerland
• Dr Mohammed Alzahrani, General Director of Vector-borne • Dr Kim Lindblade (Responsible Technical Officer), Head,
& Zoonotic Diseases Department, Public Health Agency, Elimination Unit, Global Malaria Programme, World Health
Ministry of Health, Saudi Arabia (Male – Expertise: Malaria Organization, Geneva, Switzerland
elimination programme management) • Dr Abdisalan Noor, Head, Information for Response Unit,
• Dr Kevin Baird, Director, Eijkman Oxford Clinical Research Global Malaria Programme, World Health Organization,
Unit, University of Oxford, Indonesia (Male – Expertise: Geneva, Switzerland
Malaria elimination and P. vivax) • Dr Peter Olumese, Medical Officer, Diagnostics, Medicines
• Professor Teun Bousema, Radboud University Medical & Resistance, Global Malaria Programme, World Health
Center, The Netherlands (Male – Expertise: Malaria Organization, Geneva, Switzerland
transmission) • Dr Risintha Premaratne, Technical Officer, Malaria Control,
• Dr Marcus Lacerda, Tropical Medicine Foundation Dr Heitor WHO Regional Office for South-East Asia, New Delhi, India
Vieira Dourado, Manaus, Brazil (Male – Expertise: P. vivax) • Charlotte Rasmussen, Technical Officer, Diagnostics,
• Associate Professor Dionicia Gamboa, Institute of Tropical Medicines & Resistance, World Health Organization,
Medicine, Alexander von Humboldt, Cayetano Heredia Geneva, Switzerland
University, Peru (Female – Expertise: Malaria transmission) • Dr Pascal Ringwald, Coordinator, Office of the Director,
• Professor Kevin Marsh (Co-chair), Kenya Academy of Global Malaria Programme, World Health Organization,
Sciences, Kenya (Male – Expertise: Clinical malaria Geneva, Switzerland
epidemiology) • Dr Anthony Solomon, Medical Officer, Neglected Tropical
• Dr Kamini Mendis, Independent Consultant, Sri Lanka Diseases, World Health Organization, Geneva, Switzerland
(Female – Expertise : Malaria elimination) • Dr Ghasem Zamani, Regional Adviser, Malaria and Vector
• Professor Melissa Penny, Professor and Unit Head, Swiss Control, WHO Regional Office for the Eastern
Tropical and Public Health Institute (Swiss TPH), Switzerland Mediterranean, Cairo, Egypt
(Female - Expertise: Mathematical modelling for malaria)
• Dr Allan Schapira, Visiting Consultant, Bicol University
Members of the External Review Group (2022)
College of Medicine, Phillippines (Male - Expertise: Malaria
control and research) • Dr Gao Qi, Chair of National Malaria Expert Group, Wuxi,
• Dr Siv Sovannaroth, Manager, National Malaria Programme, China (MPAG member)
Cambodia (Male – Expertise : Malaria elimination • Dr Azra Ghani, Professor, Imperial College, London, United
programme management) Kingdom of Great Britain and Northern Ireland (MPAG
• Dr Chansuda Wongsrichanalai, Consultant, Thailand (Female member)
– Expertise: P. vivax) • Dr Jimee Hwang, U.S. President’s Malaria Initiative, Malaria
Branch, Division of Parasitic Diseases and Malaria, Centers
Members of the Guidelines Steering Group (2022) for Disease Control and Prevention, Atlanta, United States
of America
• Dr Ebenezer Sheshi Baba, Medical Officer, Tropical and • Dr Jenarun Jelip, Ministry of Health, Kuala Lumpur, Malaysia
Vector Borne Diseases, WHO Regional Office for Africa, • Dr Roopal Patel, Senior Disease Advisor, Malaria, the Global
Brazzaville, Congo Fund to Fight AIDS, Tuberculosis and Malaria; Geneva,
• Dr Maurice Bucagu, Medical Officer, Maternal Health, Switzerland
World Health Organization, Geneva, Switzerland • Dr Frank Richards, Senior Advisor, Onchocerciasis,
• Dr Jane Cunningham, Technical Officer, Diagnostics, Lymphatic Filariasis, Schistosomiasis and Malaria, The
Medicines & Resistance, Global Malaria Programme, World Carter Center (Chair of the Malaria Elimination Oversight
Health Organization, Geneva, Switzerland Committee)
• Dr Blanca Escribano Ferrer, Medical Officer, Malaria and • Dr Francisco Saúte, Director General, Centro de
Neglected Tropical Diseases, WHO Regional Office for the Investigação,em Saúde de Manhiça, Manhiça, Mozambique
Americas, Washington, DC, United States of America • Dr Stephen Vreden, Vice-Chair, National Malaria Elimination
• Dr Shirin Heidari, Consultant, Gender, Equity and Human Taskforce, Paramaribo, Suriname
Rights, World Health Organization, Geneva, Switzerland
• Kanokporn Kaojaroen, Technical Officer, Health and
Systematic review team members (2022)
261 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
262 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
this interest does not present a serious conflict with respect to https://doi.org/10.1186/s12936-019-2907-7. As the paper
the meeting but would be disclosed to the GDG members could be perceived as presenting a bias against implementation
of MDA, Dr Mendis was recused from making judgments related
Dr Marcus Lacerda reported research interests related to use of to any of the MDA recommendations.
tafenoquine, an 8-aminoquinoline, for radical cure of P. vivax
infections. It was determined that this interest does not present Professor Melissa Penny declared financial research support her
a conflict with respect to the meeting but would be disclosed to institute receives related to the overall topic of discussion on
GDG members. malaria elimination, and grants that she holds on the broader
subject of malaria. It was determined that this interest does not
Dr Kevin Marsh reported that he is an advisor to several WHO, present a conflict with respect to the meeting but would be
USAID, Malaria Vaccine Initiative and PATH groups. It was disclosed to GDG members.
determined that this interest does not present a conflict with
respect to the meeting topics but would be disclosed to GDG Dr Allan Schapira reported that he is on the Board of Trustees of
members. the UK-based charity, Malaria Consortium. It was determined
that this interest does not present a conflict with respect to the
Dr Kamini Mendis reported that she is the Director of the Board meeting but would be disclosed to GDG members.
of the Asia Pacific Leaders Malaria Alliance, and that she wrote
the following paper published in the Malaria Journal in 2019, on Dr Chansuda Wongsrichanalai reported two interests, a paid
invitation by the journal to participate in a debate series. The consultancy to Medicines for Malaria Venture on radical therapy
specific topics were pre-defined by the journal, and the authors for P. vivax and travel and per diem to attend international P.
could choose among the topics presented. Mendis K. (2019). vivax meetings. It was determined that these interests do not
Mass drug administration should be implemented as a tool to present a conflict with respect to the meeting topics but would
accelerate elimination: against. Malaria Journal. 18:281. be disclosed to GDG members.
263 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
References
1. WHO Handbook for Guideline Development 2nd edition. Geneva: World Health Organization 2014; Website
2. International travel and health. Geneva: World Health Organization 2012; Website
3. Guidance on the prioritization of insecticide-treated nets in situations where resources are limited. Geneva: World Health
Organization 2023; Website
4. World malaria report 2022. Geneva: World Health Organization 2022; Website
5. Global technical strategy for malaria 2016-2030, 2021 update. Geneva: World Health Organization 2021; Website
6. High burden to high impact: a targeted malaria response. Geneva: World Health Organization 2018; Website
7. Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, et al. : GRADE: an emerging consensus on rating quality of
evidence and strength of recommendations. BMJ (Clinical research ed.) 2008;336(7650):924-6 Pubmed Journal
8. International Covenant on Economic, Social and Cultural Rights. United National General Assembly resolution (21st session) 1966;
Website
9. State of inequality: HIV, tuberculosis and malaria. Geneva: World Health Organization 2021; Website
10. A framework for malaria elimination. Geneva: World Health Organization 2017; Website
11. Yekutiel P : Problems of epidemiology in malaria eradication. Bulletin of the World Health Organization 1960;22 669-83 Pubmed
Website
12. Cameron E, Battle KE, Bhatt S, Weiss DJ, Bisanzio D, Mappin B, et al. : Defining the relationship between infection prevalence and
clinical incidence of Plasmodium falciparum malaria. Nature communications 2015;6 8170 Pubmed Journal
13. Cox J, Sovannaroth S, Dy Soley L, Ngor P, Mellor S, Roca-Feltrer A : Novel approaches to risk stratification to support malaria
elimination: an example from Cambodia. Malaria journal 2014;13 371 Pubmed Journal
14. A research agenda for malaria eradication: monitoring, evaluation, and surveillance. PLoS medicine 2011;8(1):e1000400 Pubmed
Journal
15. Investing to overcome the global impact of neglected tropical diseases. Geneva: World Health Organization 2015; Website
16. Global vector control response 2017–2030. World Health Organization, Geneva 2017; Website
17. Bhatt S, Weiss DJ, Cameron E, Bisanzio D, Mappin B, Dalrymple U, et al. : The effect of malaria control on Plasmodium falciparum in
Africa between 2000 and 2015. Nature 2015;526(7572):207-211 Pubmed Journal
18. Framework for a national vector control needs assessment. Geneva: World Health Organization 2017; Website
19. Kafy HT, Ismail BA, Mnzava AP, Lines J, Abdin MSE, Eltaher JS, et al. : Impact of insecticide resistance in Anopheles arabiensis on
malaria incidence and prevalence in Sudan and the costs of mitigation. Proceedings of the National Academy of Sciences of the United
States of America 2017;114(52):E11267-E11275 Pubmed Journal
20. Kleinschmidt I, Bradley J, Knox TB, Mnzava AP, Kafy HT, Mbogo C, et al. : Implications of insecticide resistance for malaria vector
264 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
control with long-lasting insecticidal nets: a WHO-coordinated, prospective, international, observational cohort study. The Lancet.
Infectious diseases 2018;18(6):640-649 Pubmed Journal
21. Global plan for insecticide resistance management in malaria vectors. Geneva: World Health Organization 2012; Website
22. Manual for monitoring insecticide resistance in mosquito vectors and selecting appropriate interventions. Geneva: World Health
Organization 2022; Website
23. Framework for a national plan for monitoring and management of insecticide resistance in malaria vectors. Geneva: World Health
Organization 2017; Website
24. Lissenden N, Kont MD, Essandoh J, Ismail HM, Churcher TS, Lambert B, et al. : Review and Meta-Analysis of the Evidence for
Choosing between Specific Pyrethroids for Programmatic Purposes. Insects 2021;12(9):826 Pubmed Journal
25. Insecticide-treated nets for malaria transmission control in areas with insecticide-resistant mosquito populations: preferred product
characteristics. Geneva: World Health Organization 2021; Website
26. REX Consortium : Heterogeneity of selection and the evolution of resistance. Trends in ecology & evolution 2013;28(2):110-8
Pubmed Journal
27. Sternberg ED, Thomas MB : Insights from agriculture for the management of insecticide resistance in disease vectors. Evolutionary
applications 2018;11(4):404-414 Pubmed Journal
28. Huijben S, Paaijmans KP : Putting evolution in elimination: Winning our ongoing battle with evolving malaria mosquitoes and
parasites. Evolutionary applications 2018;11(4):415-430 Pubmed Journal
29. South A, Hastings IM : Insecticide resistance evolution with mixtures and sequences: a model-based explanation. Malaria journal
2018;17(1):80 Pubmed Journal
30. Malaria surveillance, monitoring and evaluation: a reference manual. Geneva: World Health Organization 2018; Website
31. Guidance note on the control of residual malaria parasite transmission. Geneva: World Health Organization 2014; Website
32. World Health Assembly : Global vector control response: an integrated approach for the control of vector-borne diseases. Geneva:
World Health Organization 2017;70 Website
33. Ethical issues associated with vector-borne diseases. Report of a scoping meeting, 23–24 February 2017. Geneva: World Health
Organization 2017; Website
34. Ethics and vector-borne diseases: WHO guidance. Geneva: World Health Organization 2020; Website
35. Conteh L, Shuford K, Agboraw E, Kont M, Kolaczinski J, Patouillard E : Costs and Cost-Effectiveness of Malaria Control
Interventions: A Systematic Literature Review. Value in Health 2021;24(8):1213-1222 Journal Website
36. ter Kuile FO, Terlouw DJ, Phillips-Howard PA, Hawley WA, Friedman JF, Kolczak MS, et al. : Impact of permethrin-treated bed nets
on malaria and all-cause morbidity in young children in an area of intense perennial malaria transmission in western Kenya: cross-
sectional survey. The American journal of tropical medicine and hygiene 2003;68(4 Suppl):100-7 Pubmed Website
37. Gimnig JE, Kolczak MS, Hightower AW, Vulule JM, Schoute E, Kamau L, et al. : Effect of permethrin-treated bed nets on the spatial
distribution of malaria vectors in western Kenya. The American journal of tropical medicine and hygiene 2003;68(4 Suppl):115-20
Pubmed Website
38. Gimnig JE, Vulule JM, Lo TQ, Kamau L, Kolczak MS, Phillips-Howard PA, et al. : Impact of permethrin-treated bed nets on
entomologic indices in an area of intense year-round malaria transmission. The American journal of tropical medicine and hygiene
265 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
39. Phillips-Howard PA, Nahlen BL, Kolczak MS, Hightower AW, ter Kuile FO, Alaii JA, et al. : Efficacy of permethrin-treated bed nets in
the prevention of mortality in young children in an area of high perennial malaria transmission in western Kenya. The American journal
of tropical medicine and hygiene 2003;68(4 Suppl):23-9 Pubmed Website
40. Hawley WA, Phillips-Howard PA, ter Kuile FO, Terlouw DJ, Vulule JM, Ombok M, et al. : Community-wide effects of permethrin-
treated bed nets on child mortality and malaria morbidity in western Kenya. The American journal of tropical medicine and hygiene
2003;68(4 Suppl):121-7 Pubmed Website
41. D'Alessandro U, Olaleye BO, McGuire W, Langerock P, Bennett S, Aikins MK, et al. : Mortality and morbidity from malaria in
Gambian children after introduction of an impregnated bednet programme. Lancet (London, England) 1995;345(8948):479-83 Pubmed
Website
42. Quiñones ML, Lines J, Thomson MC, Jawara M, Greenwood BM : Permethrin-treated bed nets do not have a 'mass-killing effect' on
village populations of Anopheles gambiae s.l. in The Gambia. Transactions of the Royal Society of Tropical Medicine and Hygiene
92(4):373-8 Pubmed Website
43. Snow RW, Lindsay SW, Hayes RJ, Greenwood BM : Permethrin-treated bed nets (mosquito nets) prevent malaria in Gambian
children. Transactions of The Royal Society of Tropical Medicine and Hygiene 1988;82(6):838-842 Pubmed Journal Website
44. Russell TL, Lwetoijera DW, Maliti D, Chipwaza B, Kihonda J, Charlwood JD, et al. : Impact of promoting longer-lasting insecticide
treatment of bed nets upon malaria transmission in a rural Tanzanian setting with pre-existing high coverage of untreated nets. Malaria
journal 2010;9 187 Pubmed Journal
45. Govella NJ, Okumu FO, Killeen GF : Insecticide-treated nets can reduce malaria transmission by mosquitoes which feed outdoors.
The American journal of tropical medicine and hygiene 2010;82(3):415-9 Pubmed Journal
46. Birget PLG, Koella JC : An Epidemiological Model of the Effects of Insecticide-Treated Bed Nets on Malaria Transmission. PloS one
2015;10(12):e0144173 Pubmed Journal
47. Malaria control in humanitarian emergencies: an inter-agency field handbook, 2nd ed. Geneva: World Health Organization 2013;
Website
48. Dolan G., ter Kuile FO, Jacoutot V., White NJ, Luxemburger C., Malankirii L., et al. : Bed nets for the prevention of malaria and
anaemia in pregnancy. Transactions of The Royal Society of Tropical Medicine and Hygiene 1993;87(6):620-626 Pubmed Journal
Website
49. Luxemburger C., Perea W.A., Delmas G., Pruja C., Pecoul B., Moren A. : Permethrin-impregnated bed nets for the prevention of
malaria in schoolchildren on the Thai-Burmese border. Transactions of The Royal Society of Tropical Medicine and Hygiene
1994;88(2):155-159 Pubmed Journal Website
50. Rowland M, Bouma M, Ducornez D, Durrani N, Rozendaal J, Schapira A, et al. : Pyrethroid-impregnated bed nets for personal
protection against malaria for Afghan refugees. Transactions of The Royal Society of Tropical Medicine and Hygiene
1996;90(4):357-361 Pubmed Journal Website
51. Rowland M, Hewitt S, Durrani N, Bano N, Wirtz R : Transmission and control of vivax malaria in Afghan refugee settlements in
Pakistan. Transactions of The Royal Society of Tropical Medicine and Hygiene 1997;91(3):252-255 Pubmed Journal Website
52. Rowland M, Mahmood P, Iqbal J, Carneiro I, Chavasse D : Indoor residual spraying with alphacypermethrin controls malaria in
Pakistan: a community-randomized trial. Tropical Medicine & International Health 2000;5(7):472-481 Pubmed Journal Website
53. Smithuis FM, Kyaw MK, Phe UO, van der Broek I, Katterman N, Rogers C, et al. : The effect of insecticide-treated bed nets on the
incidence and prevalence of malaria in children in an area of unstable seasonal transmission in western Myanmar. Malaria Journal
2013;12(1):363 Pubmed Journal Website
266 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
54. Messenger LA, Furnival-Adams J, Pelloquin B, Rowland M : Vector control for malaria prevention during humanitarian emergencies:
protocol for a systematic review and meta-analysis. BMJ Open 2021/07/27;11(7):e046325-e046325 Pubmed Journal Website
55. Pryce J, Richardson M, Lengeler C : Insecticide-treated nets for preventing malaria. Cochrane Database of Systematic Reviews
2018;(11): Pubmed Journal Website
56. Technical consultation on determining non-inferiority of vector control products within an established class: Report of a virtual
meeting 31 August–2 September 2021. Geneva: World Health Organization 2021; Website
57. Staedke SG, Gonahasa S, Dorsey G, Kamya MR, Maiteki-Sebuguzi C, Lynd A, et al. : Effect of long-lasting insecticidal nets with and
without piperonyl butoxide on malaria indicators in Uganda (LLINEUP): a pragmatic, cluster-randomised trial embedded in a national
LLIN distribution campaign. The Lancet 2020;395(10232):1292-1303 Pubmed Journal Website
58. Protopopoff N, Mosha JF, Lukole E, Charlwood JD, Wright A, Mwalimu CD, et al. : Effectiveness of a long-lasting piperonyl butoxide-
treated insecticidal net and indoor residual spray interventions, separately and together, against malaria transmitted by pyrethroid-
resistant mosquitoes: a cluster, randomised controlled, two-by-two factorial design trial. Lancet (London, England)
2018;391(10130):1577-1588 Pubmed Journal
59. Gleave K, Lissenden N, Richardson M, Choi L, Ranson H : Piperonyl butoxide (PBO) combined with pyrethroids in insecticide-treated
nets to prevent malaria in Africa. The Cochrane Database of Systematic Reviews 2021;5 CD012776 Pubmed Journal
60. Piperonyl butoxide (PBO) combined with pyrethroids in insecticide-treated nets to prevent malaria in Africa. 2021; Pubmed
61. Mosha JF, Kulkarni MA, Lukole E, Matowo NS, Pitt C, Messenger LA, et al. : Effectiveness and cost-effectiveness against malaria of
three types of dual-active-ingredient long-lasting insecticidal nets (LLINs) compared with pyrethroid-only LLINs in Tanzania: a four-arm,
cluster-randomised trial. Lancet (London, England) 2022;399(10331):1227-1241 Pubmed Journal
62. Accrombessi M, Cook J, Dangbenon E, Yovogan B, Akpovi H, Sovi A, et al. : Efficacy of pyriproxyfen-pyrethroid long-lasting
insecticidal nets (LLINs) and chlorfenapyr-pyrethroid LLINs compared with pyrethroid-only LLINs for malaria control in Benin: a cluster-
randomised, superiority trial. Lancet (London, England) 2023;401(10375):435-446 Pubmed Journal
63. Tiono AB, Ouédraogo A, Ouattara D, Bougouma EC, Coulibaly S, Diarra A, et al. : Efficacy of Olyset Duo, a bednet containing
pyriproxyfen and permethrin, versus a permethrin-only net against clinical malaria in an area with highly pyrethroid-resistant vectors in
rural Burkina Faso: a cluster-randomised controlled trial. Lancet (London, England) 2018;392(10147):569-580 Pubmed Journal
64. Achieving and maintaining universal coverage with long-lasting insecticidal nets for malaria control. Geneva: World Health
Organization 2017; Website
65. WHO recommendations on the sound management of old long-lasting insecticidal nets. Geneva: World Health Organization 2014;
Website
66. Stockholm Convention on Persistent Organic Pollutants (POPS). United Nations Environment Programme 2018; Website
67. Indoor residual spraying: An operational manual for IRS for malaria transmission, control and elimination. 2nd edition. Geneva:
World Health Organization 2015; Website
68. Meeting report on the WHO Evidence Review Group on assessing comparative effectiveness of new vector control tools. Geneva:
World Health Organization 2017; Website
69. Indoor residual spraying: use of indoor residual spraying for scaling up global malaria control and elimination. Geneva: World Health
Organization 2006; Website
70. Pluess B, Tanser FC, Lengeler C, Sharp BL : Indoor residual spraying for preventing malaria. The Cochrane database of systematic
reviews 2010;(4):CD006657 Pubmed Journal
267 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
71. Charlwood J.D, Qassim M, Elnsur E.I, Donnelly M, Petrarca V, Billingsley P.F, et al. : The impact of indoor residual spraying with
malathion on malaria in refugee camps in eastern Sudan. Acta Tropica 2001;80(1):1-8 Pubmed Journal Website
72. Rowland M, Hewitt S, Durrani N : Prevalence of malaria in Afghan refugee villages in Pakistan sprayed with lambdacyhalothrin or
malathion. Transactions of the Royal Society of Tropical Medicine and Hygiene 88(4):378-9 Pubmed
73. Wahid S, Stresman GH, Kamal SS, Sepulveda N, Kleinschmidt I, Bousema T, et al. : Heterogeneous malaria transmission in long-term
Afghan refugee populations: a cross-sectional study in five refugee camps in northern Pakistan. Malaria journal 2016;15 245 Pubmed
Journal
74. Choi L, Pryce J, Garner P : Indoor residual spraying for preventing malaria in communities using insecticide-treated nets. The
Cochrane database of systematic reviews 2019;(5):CD012688 Pubmed Journal Website
75. WHO Guidance for countries on combining indoor residual spraying and long-lasting insecticidal nets. Geneva: World Health
Organization 2014 ; Website
76. Risks associated with scale-back of vector control after malaria transmission has been reduced. Information note. Geneva: World
Health Organization 2015; Website
77. Maia MF, Kliner M, Richardson M, Lengeler C, Moore SJ : Mosquito repellents for malaria prevention. The Cochrane database of
systematic reviews 2018;(2):CD011595 Pubmed Journal Website
78. WHO Housing and health guidelines. Geneva: World Health Organization 2018; Website
79. Keeping the vector out: housing improvements for vector control and sustainable development. Geneva: World Health Organization
2017; Website
80. Tusting LS, Bottomley C, Gibson H, Kleinschmidt I, Tatem AJ, Lindsay SW, et al. : Housing Improvements and Malaria Risk in Sub-
Saharan Africa: A Multi-Country Analysis of Survey Data. PLoS medicine 2017;14(2):e1002234 Pubmed Journal
81. Larval source management: a supplementary measure for malaria vector control. An operational manual. Geneva: World Health
Organization 2013; Website
82. Choi L, Majambere S, Wilson AL : Larviciding to prevent malaria transmission. The Cochrane database of systematic reviews
2019;(8):CD012736 Pubmed Journal Website
83. Walshe DP, Garner P, Adeel AA, Pyke GH, Burkot TR : Larvivorous fish for preventing malaria transmission. The Cochrane database
of systematic reviews 2017;(12):CD008090 Pubmed Journal Website
84. Pryce J, Choi L, Richardson M, Malone D : Insecticide space spraying for preventing malaria transmission. The Cochrane database of
systematic reviews 2018;(11):CD012689 Pubmed Journal Website
85. Furnival-Adams JA, Olanga EA, Napier M, Garner M : House modifications for preventing malaria. The Cochrane database of
systematic reviews 2021;(1):CD013398 Pubmed Journal Website
86. Getawen SK, Ashine T, Massebo F, Woldeyes D, Lindtjørn B : Exploring the impact of house screening intervention on entomological
indices and incidence of malaria in Arba Minch town, southwest Ethiopia: A randomized control trial. Acta tropica 2018;181 84-94
Pubmed Journal
87. Kirby MJ, Ameh D, Bottomley C, Green C, Jawara M, Milligan PJ, et al. : Effect of two different house screening interventions on
exposure to malaria vectors and on anaemia in children in The Gambia: a randomised controlled trial. Lancet (London, England)
2009;374(9694):998-1009 Pubmed Journal
88. Barreaux AMG, Oumbouke WA, Brou N, Tia IZ, Ahoua Alou LP, Doudou DT, et al. : The role of human and mosquito behaviour in the
efficacy of a house-based intervention. Philosophical transactions of the Royal Society of London. Series B, Biological sciences
268 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
89. Global surveillance, prevention and control of chronic respiratory diseases : a comprehensive approach. Geneva: World Health
Organization 2007; Website
90. Exposure to household air pollution. Geneva: World Health Organization 2021; Website
91. Guidelines for indoor air quality: household fuel combustion. Geneva: World Health Organization 2014; Website
92. Sundell J, Levin H, Nazaroff WW, Cain WS, Fisk WJ, Grimsrud DT, et al. : Ventilation rates and health: multidisciplinary review of the
scientific literature. Indoor air 2011;21(3):191-204 Pubmed Journal
93. Knudsen JB, Pinder M, Jatta E, Jawara M, Yousuf MA, Søndergaard AT, et al. : Measuring ventilation in different typologies of rural
Gambian houses: a pilot experimental study. Malaria journal 2020;19(1):273 Pubmed Journal
94. Jatta E, Jawara M, Bradley J, Jeffries D, Kandeh B, Knudsen JB, et al. : How house design affects malaria mosquito density,
temperature, and relative humidity: an experimental study in rural Gambia. The Lancet Planetary Health 2018;2(11):e498-e508 Pubmed
Journal Website
95. Norms, standards and processes underpinning development of WHO recommendations on vector control. Geneva: World Health
Organization 2020; Website
96. Sicuri E, Bardají A, Nhampossa T, Maixenchs M, Nhacolo A, Nhalungo D, et al. : Cost-effectiveness of intermittent preventive
treatment of malaria in pregnancy in southern Mozambique. PloS one 2010;5(10):e13407 Pubmed Journal
97. Fernandes S, Sicuri E, Kayentao K, van Eijk AM, Hill J, Webster J, et al. : Cost-effectiveness of two versus three or more doses of
intermittent preventive treatment for malaria during pregnancy in sub-Saharan Africa: a modelling study of meta-analysis and cost data.
The Lancet. Global health 2015;3(3):e143-53 Pubmed Journal
98. Hansen KS, Ndyomugyenyi R, Magnussen P, Clarke SE : Cost-effectiveness analysis of three health interventions to prevent malaria
in pregnancy in an area of low transmission in Uganda. International health 2012;4(1):38-46 Pubmed Journal
99. Diengou NH, Cumber SN, Nkfusai CN, Mbinyui MS, Viyoff VZ, Bede F, et al. : Factors associated with the uptake of intermittent
preventive treatment of malaria in pregnancy in the Bamenda health districts, Cameroon. The Pan African medical journal 2020;35 42
Pubmed Journal
100. Nekaka R, Nteziyaremye J, Oboth P, Iramiot JS, Wandabwa J : Malaria preventive practices and delivery outcomes: A cross-
sectional study of parturient women in a tertiary hospital in Eastern Uganda. PloS one 2020;15(8):e0237407 Pubmed Journal
101. Ngapanya S, Mikomangwa W, Bwire G, Kilonzi M, Mlyuka H, Mutagonda R, et al. : Uptake of Intermittent Preventive Therapy
Among Pregnant Women Living in Dar es Salaam, Tanzania: a Descriptive Cross-sectional Study. SN Comprehensive Clinical Medicine
2020;2 408-413 Journal Website
102. Onyebuchi AK, Lawani LO, Iyoke CA, Onoh CR, Okeke NE : Adherence to intermittent preventive treatment for malaria with
sulphadoxine-pyrimethamine and outcome of pregnancy among parturients in South East Nigeria. Patient preference and adherence
2014;8 447-52 Pubmed Journal
103. Moberg J, Oxman AD, Rosenbaum S, Schünemann HJ, Guyatt G, Flottorp S, et al. : The GRADE Evidence to Decision (EtD)
framework for health system and public health decisions. Health research policy and systems 2018;16(1):45 Pubmed Journal
104. WHO policy brief for the implementation of intermittent preventive treatment of malaria in pregnancy using sulfadoxine-
pyrimethamine (IPTp-SP)SP. Geneva: World Health Organization 2014; Website
105. WHO recommendations on antenatal care for a positive pregnancy experience. Geneva: World Health Organization 2016; Website
269 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
106. Esu EB, Oringanje C, Meremikwu MM : Intermittent preventive treatment for malaria in infants. The Cochrane database of
systematic reviews 2021;7 CD011525 Pubmed Journal
107. Kobbe R, Kreuzberg C, Adjei S, Thompson B, Langefeld I, Thompson PA, et al. : A randomized controlled trial of extended
intermittent preventive antimalarial treatment in infants. Clinical infectious diseases : an official publication of the Infectious Diseases
Society of America 2007;45(1):16-25 Pubmed
108. Mockenhaupt FP, Reither K, Zanger P, Roepcke F, Danquah I, Saad E, et al. : Intermittent preventive treatment in infants as a means
of malaria control: a randomized, double-blind, placebo-controlled trial in northern Ghana. Antimicrobial agents and chemotherapy
2007;51(9):3273-81 Pubmed
109. Grobusch MP, Lell B, Schwarz NG, Gabor J, Dornemann J, Potschke M, et al. : Intermittent preventive treatment against malaria in
infants in Gabon--a randomized, double-blind, placebo-controlled trial. The Journal of infectious diseases 2007;196(11):1595-602
Pubmed
110. Bigira V, Kapisi J, Clark TD, Kinara S, Mwangwa F, Muhindo MK, et al. : Protective efficacy and safety of three antimalarial regimens
for the prevention of malaria in young Ugandan children: a randomized controlled trial. PLoS medicine 2014;11(8):e1001689 Pubmed
Journal
111. Griffin JT, Cairns M, Ghani AC, Roper C, Schellenberg D, Carneiro I, et al. : Protective efficacy of intermittent preventive treatment
of malaria in infants (IPTi) using sulfadoxine-pyrimethamine and parasite resistance. PloS one 2010;5(9):e12618 Pubmed Journal
112. Lahuerta M, Sutton R, Mansaray A, Eleeza O, Gleason B, Akinjeji A, et al. : Evaluation of health system readiness and coverage of
intermittent preventive treatment of malaria in infants (IPTi) in Kambia district to inform national scale-up in Sierra Leone. Malaria
journal 2021;20(1):74 Pubmed Journal
113. Manzi F, Schellenberg J, Hamis Y, Mushi AK, Shirima K, Mwita A, et al. : Intermittent preventive treatment for malaria and anaemia
control in Tanzanian infants; the development and implementation of a public health strategy. Transactions of the Royal Society of
Tropical Medicine and Hygiene 2009;103(1):79-86 Pubmed Journal
114. Shea BJ, Reeves BC, Wells G, Thuku M, Hamel C, Moran J, et al. : AMSTAR 2: a critical appraisal tool for systematic reviews that
include randomised or non-randomised studies of healthcare interventions, or both. BMJ (Clinical research ed.) 2017;358 j4008
Pubmed Journal
115. Intermittent preventive treatment for infants using sulfadoxine-pyrimethamine (SP-IPTi) for malaria control in Africa:
implementation field guide. Geneva: World Health Organization 2011; Website
116. Paton RS, Kamau A, Akech S, Agweyu A, Ogero M, Mwandawiro C, et al. : Malaria infection and severe disease risks in Africa.
Science (New York, N.Y.) 2021;373(6557):926-931 Pubmed Journal
117. Issiaka D, Barry A, Tounkara M, Traore T, Diarra I, Kone D, et al. : Determining the optimal mode for delivery of seasonal malaria
chemoprevention in Mali. American Journal of Tropical Medicine and Hygiene 2015;93(4 Supplement):496 Website
118. Barry A, Issiaka D, Traore T, Mahamar A, Diarra B, Sagara I, et al. : Optimal mode for delivery of seasonal malaria chemoprevention
in Ouelessebougou, Mali: A cluster randomized trial. PloS one 2018;13(3):e0193296 Pubmed Journal
119. Ceesay S, Hubbard E, Bojang K, Kandeh B, Kolley O, Jah H, et al. : Implementation of seasonal malaria chemoprevention in The
Gambia. American Journal of Tropical Medicine and Hygiene 2016;95(5 Supplement 1): Website
120. : Effectiveness of seasonal malaria chemoprevention at scale in west and central Africa: an observational study. Lancet (London,
England) 2020;396(10265):1829-1840 Pubmed Journal
121. Seasonal malaria chemoprevention with sulfadoxine-pyrimethamine plus amodiaquine in children: A field guide. Geneva: World
Health Organization 2013; Website
122. Clarke SE, Jukes MC, Njagi JK, Khasakhala L, Cundill B, Otido J, et al. : Effect of intermittent preventive treatment of malaria on
270 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
health and education in schoolchildren: a cluster-randomised, double-blind, placebo-controlled trial. Lancet (London, England)
2008;372(9633):127-138 Pubmed Journal
123. Nankabirwa J, Cundill B, Clarke S, Kabatereine N, Rosenthal PJ, Dorsey G, et al. : Efficacy, safety, and tolerability of three regimens
for prevention of malaria: a randomized, placebo-controlled trial in Ugandan schoolchildren. PloS one 2010;5(10):e13438 Pubmed
Journal
124. Matangila JR, Doua JY, Mitashi P, da Luz RI, Lutumba P, Van Geertruyden JP : Efficacy and safety of intermittent preventive
treatment in schoolchildren with sulfadoxine/pyrimethamine (SP) and SP plus piperaquine in Democratic Republic of the Congo: a
randomised controlled trial. International journal of antimicrobial agents 2017;49(3):339-347 Pubmed Journal
125. Cohee LM, Opondo C, Clarke SE, Halliday KE, Cano J, Shipper AG, et al. : Preventive malaria treatment among school-aged children
in sub-Saharan Africa: a systematic review and meta-analyses. The Lancet. Global health 2020;8(12):e1499-e1511 Pubmed Journal
126. Audibert C, Tchouatieu A-M : Perception of Malaria Chemoprevention Interventions in Infants and Children in Eight Sub-Saharan
African Countries: An End User Perspective Study. Tropical medicine and infectious disease 2021;6(2): Pubmed Journal
127. Maccario R, Rouhani S, Drake T, Nagy A, Bamadio M, Diarra S, et al. : Cost analysis of a school-based comprehensive malaria
program in primary schools in Sikasso region, Mali. BMC public health 2017;17(1):572 Pubmed Journal
128. Temperley M, Mueller DH, Njagi JK, Akhwale W, Clarke SE, Jukes MCH, et al. : Costs and cost-effectiveness of delivering
intermittent preventive treatment through schools in western Kenya. Malaria journal 2008;7 196 Pubmed Journal
129. Maiga H, Opondo C, Chico RM, Cohee LM, Sagara I, Traore OB, et al. : Overall and gender-specific effects of intermittent
preventive treatment of malaria with artemisinin-based combination therapies among schoolchildren in Mali: a three-group open label
randomized controlled trial. American Journal of Tropical Medicine & Hygiene 2022;
130. Rohner F, Zimmermann MB, Amon RJ, Vounatsou P, Tschannen AB, N'goran EK, et al. : In a randomized controlled trial of iron
fortification, anthelmintic treatment, and intermittent preventive treatment of malaria for anemia control in Ivorian children, only
anthelmintic treatment shows modest benefit. The Journal of nutrition 2010;140(3):635-41 Pubmed Journal
131. Cohee LM, Chilombe M, Ngwira A, Jemu SK, Mathanga DP, Laufer MK : Pilot Study of the Addition of Mass Treatment for Malaria
to Existing School-Based Programs to Treat Neglected Tropical Diseases. The American journal of tropical medicine and hygiene
2018;98(1):95-99 Pubmed Journal
132. Staedke SG, Maiteki-Sebuguzi C, Rehman AM, Kigozi SP, Gonahasa S, Okiring J, et al. : Assessment of community-level effects of
intermittent preventive treatment for malaria in schoolchildren in Jinja, Uganda (START-IPT trial): a cluster-randomised trial. The Lancet.
Global health 2018;6(6):e668-e679 Pubmed Journal
133. Haemoglobin concentrations for the diagnosis of anaemia and assessment of severity. Geneva: World Health Organization 2011;
Website
134. Phiri K, Esan M, van Hensbroek MB, Khairallah C, Faragher B, ter Kuile FO : Intermittent preventive therapy for malaria with
monthly artemether-lumefantrine for the post-discharge management of severe anaemia in children aged 4-59 months in southern
Malawi: a multicentre, randomised, placebo-controlled trial. The Lancet. Infectious diseases 2012;12(3):191-200 Pubmed Journal
135. Kwambai TK, Dhabangi A, Idro R, Opoka R, Watson V, Kariuki S, et al. : Malaria Chemoprevention in the Postdischarge
Management of Severe Anemia. The New England journal of medicine 2020;383(23):2242-2254 Pubmed Journal
136. Bojang KA, Milligan PJM, Conway DJ, Sisay-Joof F, Jallow M, Nwakanma DC, et al. : Prevention of the recurrence of anaemia in
Gambian children following discharge from hospital. PloS one 2010;5(6):e11227 Pubmed Journal
137. Nkosi-Gondwe T, Robberstad B, Blomberg B, Phiri KS, Lange S : Introducing post-discharge malaria chemoprevention (PMC) for
management of severe anemia in Malawian children: a qualitative study of community health workers' perceptions and motivation. BMC
health services research 2018;18(1):984 Pubmed Journal
271 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
138. Nkosi-Gondwe T, Robberstad B, Mukaka M, Idro R, Opoka RO, Banda S, et al. : Adherence to community versus facility-based
delivery of monthly malaria chemoprevention with dihydroartemisinin-piperaquine for the post-discharge management of severe
anemia in Malawian children: A cluster randomized trial. PloS one 2021;16(9):e0255769 Pubmed Journal
139. Svege S, Kaunda B, Robberstad B, Nkosi-Gondwe T, Phiri KS, Lange S : Post-discharge malaria chemoprevention (PMC) in Malawi:
caregivers` acceptance and preferences with regard to delivery methods. BMC health services research 2018;18(1):544 Pubmed Journal
140. Nadia J, Lu F : Historical experiences on mass drug administration for malaria control and elimination, its challenges and China's
experience: a narrative review. Acta tropica 2022;225 106209 Pubmed Journal
141. Mass drug administration for falciparum malaria: a practical field manual. Geneva: World Health Organization 2017; Website
142. Immunization Agenda 2030: A Global Strategy to Leave No One Behind. Geneva: World Health Organization 2020; Website
143. Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a
phase 3, individually randomised, controlled trial. Lancet (London, England) 2015;386(9988):31-45 Pubmed Journal
144. Mosquirix: Opinion on medicine for use outside EU. European Medicines Agency 2015; Website
145. Chandramohan D, Zongo I, Sagara I, Cairns M, Yerbanga R-S, Diarra M, et al. : Seasonal Malaria Vaccination with or without
Seasonal Malaria Chemoprevention. The New England journal of medicine 2021;385(11):1005-1017 Pubmed Journal
146. Datoo MS, Natama MH, Somé A, Traoré O, Rouamba T, Bellamy D, et al. : Efficacy of a low-dose candidate malaria vaccine, R21 in
adjuvant Matrix-M, with seasonal administration to children in Burkina Faso: a randomised controlled trial. Lancet (London, England)
2021;397(10287):1809-1818 Pubmed Journal
147. Butler D : Promising malaria vaccine to be tested in first large field trial. Nature 2019; Pubmed Journal
148. Minassian AM, Silk SE, Barrett JR, Nielsen CM, Miura K, Diouf A, et al. : Reduced blood-stage malaria growth and immune
correlates in humans following RH5 vaccination. Med (New York, N.Y.) 2021;2(6):701-719.e19 Pubmed Journal
149. Draper SJ, Angov E, Horii T, Miller LH, Srinivasan P, Theisen M, et al. : Recent advances in recombinant protein-based malaria
vaccines. Vaccine 2015;33(52):7433-43 Pubmed Journal
150. Adjuvant development for vaccines and for autoimmune and allergic diseases. Washington DC, United States of America. Small
Business Innovation Research and Small Business Technology Transfer 2020; Website
151. Guidelines on the quality, safety and efficacy of recombinant malaria vaccines targeting the pre-erythrocytic and blood stages of
Plasmodium falciparum, Annex 3, TRS No 980. Geneva: World Health Organization 2014; Website
152. WHO preferred product characteristics (PPC) for malaria vaccines. Geneva: World Health Organization 2014; Website
153. Malaria vaccine: WHO position paper - March 2022. Weekly Epidemiological Record,Vol. 97, No. 09, pp. 61-80. 4 March 2022.
Geneva: World Health Organization 2022; Website
154. Tinto H, Otieno W, Gesase S, Sorgho H, Otieno L, Liheluka E, et al. : Long-term incidence of severe malaria following RTS,S/AS01
vaccination in children and infants in Africa: an open-label 3-year extension study of a phase 3 randomised controlled trial. The Lancet.
Infectious diseases 2019;19(8):821-832 Pubmed Journal
155. Milligan P, Moore K : Statistical report on the results of the RTS,S/AS01 Malaria Vaccine Pilot Evaluation 24 months after the
vaccine was introduced (unpublished evidence). 2021;V1.3 6 Sept 2021 Website
156. Penny MA, Galactionova K, Tarantino M, Tanner M, Smith TA : The public health impact of malaria vaccine RTS,S in malaria
endemic Africa: country-specific predictions using 18 month follow-up Phase III data and simulation models. BMC medicine 2015;13
272 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
157. Vekemans J, Guerra Y, Lievens M, Benns S, Lapierre D, Leach A, et al. : Pooled analysis of safety data from pediatric Phase II RTS,S/
AS malaria candidate vaccine trials. Human vaccines 2011;7(12):1309-16 Pubmed Journal
158. Asante KP, Abdulla S, Agnandji S, Lyimo J, Vekemans J, Soulanoudjingar S, et al. : Safety and efficacy of the RTS,S/AS01E candidate
malaria vaccine given with expanded-programme-on-immunisation vaccines: 19 month follow-up of a randomised, open-label, phase 2
trial. The Lancet. Infectious diseases 2011;11(10):741-9 Pubmed Journal
159. Agnandji ST, Asante KP, Lyimo J, Vekemans J, Soulanoudjingar SS, Owusu R, et al. : Evaluation of the safety and immunogenicity of
the RTS,S/AS01E malaria candidate vaccine when integrated in the expanded program of immunization. The Journal of infectious
diseases 2010;202(7):1076-87 Pubmed Journal
160. Talaat KR, Ellis RD, Hurd J, Hentrich A, Gabriel E, Hynes NA, et al. : Safety and Immunogenicity of Pfs25-EPA/Alhydrogel®, a
Transmission Blocking Vaccine against Plasmodium falciparum: An Open Label Study in Malaria Naïve Adults. PloS one
2016;11(10):e0163144 Pubmed Journal
161. Guimarães LE, Baker B, Perricone C, Shoenfeld Y : Vaccines, adjuvants and autoimmunity. Pharmacological research 2015;100
190-209 Pubmed Journal
162. Malaria case management: operations manual. Geneva: World Health Organization 2009; Website
163. Integrated management of childhood illness for high HIV settings: chart booklet. Geneva: World Health Organization 2008;
Website
164. Universal access to malaria diagnostic testing - an operational manual. Geneva: World Health Organization 2011; Website
165. Malaria diagnosis: memorandum from a WHO meeting. Bulletin of the World Health Organization 1988;66(5):575-94 Pubmed
166. Malaria microscopy quality assurance manual, version 2. Geneva: World Health Organization 2016; Website
167. Kawamoto F, Billingsley PF : Rapid diagnosis of malaria by fluorescence microscopy. Parasitology today (Personal ed.)
1992;8(2):69-71 Pubmed
168. Malaria diagnosis: new perspectives. Geneva: World Health Organization 2003; Website
169. Malaria rapid diagnosis: making it work. Meeting report. World Health Organization. Regional Office for the Western Pacific 2003;
Website
170. The use of rapid diagnostic tests. World Health Organization. Regional Office for the Western Pacific 2004; Website
171. Transporting, storing and handling malaria rapid diagnostic tests in health clinics. Geneva: World Health Organization 2009;
Website
172. Malaria rapid diagnostic test performance. Results of WHO product testing of malaria RDTs: round 8. Geneva: World Health
Organization 2018; Website
173. False-negative RDT results and implications of new reports of P. falciparum hrp 2/3 gene deletions. Geneva: World Health
Organization 2017; Website
174. Abba K, Deeks JJ, Olliaro P, Naing C-M, Jackson SM, Takwoingi Y, et al. : Rapid diagnostic tests for diagnosing uncomplicated P.
falciparum malaria in endemic countries. The Cochrane database of systematic reviews 2011;(7):CD008122 Pubmed Journal
273 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
175. Thiam S, Thior M, Faye B, Ndiop M, Diouf ML, Diouf MB, et al. : Major reduction in anti-malarial drug consumption in Senegal after
nation-wide introduction of malaria rapid diagnostic tests. PloS one 2011;6(4):e18419 Pubmed Journal
176. Voller A : The immunodiagnosis of malaria. In: Wernsdorfer WH, McGregor I, editors. Malaria. Principles and Practice of
Malariology. Edinburgh: Churchill Livingstone 1988;1 815-827 Website
177. Bates I, Iboro J, Barnish G : Challenges in monitoring the impact of interventions against malaria using diagnostics. In: Reducing
malaria's burden. Evidence of effectiveness for decision-makers. Global Health Council, Washington D.C. 2003; 33-39 Website
178. WHO Evidence review group on malaria diagnosis in low transmission settings. Meeting Report. Geneva: World Health
Organization 2012; Website
179. Good procurement practices for artemisinin-based antimalarial medicines. Geneva: World Health Organization 2010; Website
180. Sinclair D, Zani B, Donegan S, Olliaro P, Garner P : Artemisinin-based combination therapy for treating uncomplicated malaria. The
Cochrane database of systematic reviews 2009;(3):CD007483 Pubmed Journal
181. Zani B, Gathu M, Donegan S, Olliaro PL, Sinclair D : Dihydroartemisinin-piperaquine for treating uncomplicated Plasmodium
falciparum malaria. The Cochrane database of systematic reviews 2014;(1):CD010927 Pubmed Journal
182. Tarning J, Zongo I, Somé FA, Rouamba N, Parikh S, Rosenthal PJ, et al. : Population pharmacokinetics and pharmacodynamics of
piperaquine in children with uncomplicated falciparum malaria. Clinical pharmacology and therapeutics 2012;91(3):497-505 Pubmed
Journal
183. Tona Lutete G, Mombo-Ngoma G, Assi S-B, Bigoga JD, Koukouikila-Koussounda F, Ntamabyaliro NY, et al. : Pyronaridine-
artesunate real-world safety, tolerability, and effectiveness in malaria patients in 5 African countries: A single-arm, open-label, cohort
event monitoring study. PLoS medicine 2021;18(6):e1003669 Pubmed Journal
184. Pryce J, Taylor M, Fox T, Hine P : Pyronaridine-artesunate for treating uncomplicated Plasmodium falciparum malaria. Cochrane
Database Syst Rev 2022;6(6): Pubmed Journal
185. : The effect of dosing regimens on the antimalarial efficacy of dihydroartemisinin-piperaquine: a pooled analysis of individual
patient data. PLoS medicine 2013;10(12):e1001564; discussion e1001564 Pubmed Journal
186. Graves PM, Gelband H, Garner P : Primaquine or other 8-aminoquinoline for reducing P. falciparum transmission. The Cochrane
database of systematic reviews 2014;(6):CD008152 Pubmed Journal
187. White NJ, Qiao LG, Qi G, Luzzatto L : Rationale for recommending a lower dose of primaquine as a Plasmodium falciparum
gametocytocide in populations where G6PD deficiency is common. Malaria journal 2012;11 418 Pubmed Journal
188. Recht J, Ashley E, White N : Safety of 8-aminoquinoline antimalarial medicines. World Health Organization, Geneva 2014; Website
189. Policy brief on single-dose primaquine as a gametocytocide in Plasmodium falciparum malaria. Geneva: World Health Organization
2015; Website
190. Desai M, ter Kuile FO, Nosten F, McGready R, Asamoa K, Brabin B, et al. : Epidemiology and burden of malaria in pregnancy. The
Lancet. Infectious diseases 2007;7(2):93-104 Pubmed
191. Accrombessi M, Yovo E, Fievet N, Cottrell G, Agbota G, Gartner A, et al. : Effects of Malaria in the First Trimester of Pregnancy on
Poor Maternal and Birth Outcomes in Benin. Clinical infectious diseases : an official publication of the Infectious Diseases Society of
America 2019;69(8):1385-1393 Pubmed Journal
192. Walker PGT, ter Kuile FO, Garske T, Menendez C, Ghani AC : Estimated risk of placental infection and low birthweight attributable
to Plasmodium falciparum malaria in Africa in 2010: a modelling study. The Lancet. Global health 2014;2(8):e460-7 Pubmed Journal
274 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
194. White TEK, Bushdid PB, Ritter S, Laffan SB, Clark RL : Artesunate-induced depletion of embryonic erythroblasts precedes
embryolethality and teratogenicity in vivo. Birth defects research. Part B, Developmental and reproductive toxicology
2006;77(5):413-29 Pubmed
195. Saito M, Mansoor R, Kennon K, Anvikar AR, Ashley EA, Chandramohan D, et al. : Efficacy and tolerability of artemisinin-based and
quinine-based treatments for uncomplicated falciparum malaria in pregnancy: a systematic review and individual patient data meta-
analysis. The Lancet. Infectious diseases 2020;20(8):943-952 Pubmed Journal
196. Burger RJ, van Eijk AM, Bussink M, Hill J, Ter Kuile FO : Artemisinin-Based Combination Therapy Versus Quinine or Other
Combinations for Treatment of Uncomplicated Plasmodium falciparum Malaria in the Second and Third Trimester of Pregnancy: A
Systematic Review and Meta-Analysis. Open forum infectious diseases 2016;3(1):ofv170 Pubmed Journal
197. Clark RL, Arima A, Makori N, Nakata Y, Bernard F, Gristwood W, et al. : Artesunate: developmental toxicity and toxicokinetics in
monkeys. Birth defects research. Part B, Developmental and reproductive toxicology 2008;83(4):418-34 Pubmed Journal
198. Clark RL : Embryotoxicity of the artemisinin antimalarials and potential consequences for use in women in the first trimester.
Reproductive toxicology (Elmsford, N.Y.) 2009;28(3):285-96 Pubmed Journal
199. Clark RL : Teratogen update: Malaria in pregnancy and the use of antimalarial drugs in the first trimester. Birth defects research
2020;112(18):1403-1449 Pubmed Journal
200. Moore KA, Simpson JA, Scoullar MJL, McGready R, Fowkes FJI : Quantification of the association between malaria in pregnancy
and stillbirth: a systematic review and meta-analysis. The Lancet. Global health 2017;5(11):e1101-e1112 Pubmed Journal
201. Saito M, McGready R, Titnto H, Rouamba T, Mosha D, Rulisa S, et al. : Pregnancy outcomes after first-trimester treatment with
artemisinin derivatives versus non-artemisinin antimalarials: a systematic review and individual patient data meta-analysis. The Lancet In
Press 2022 ; Journal
202. Dellicour S, Sevene E, McGready R, Tinto H, Mosha D, Manyando C, et al. : First-trimester artemisinin derivatives and quinine
treatments and the risk of adverse pregnancy outcomes in Africa and Asia: A meta-analysis of observational studies. PLoS medicine
2017;14(5):e1002290 Pubmed Journal
203. Mitchell AA : Special considerations in studies of drug induced birth defects. In: Strom BL, editor. Pharmacoepidemiology, 3rd
edition. Chichester: Wiley 2000; 749-763
204. Heinonen OP, Shapiro S, Slone D : Birth defects and drugs in pregnancy. Littleton: Publishing Sciences Group 1977;
205. PREGACT Study Group, Pekyi D, Ampromfi AA, Tinto H, Traoré-Coulibaly M, Tahita MC, et al. : Four Artemisinin-Based Treatments
in African Pregnant Women with Malaria. The New England journal of medicine 2016;374(10):913-27 Pubmed Journal
206. Pell C, Straus L, Andrew EVW, Meñaca A, Pool R : Social and cultural factors affecting uptake of interventions for malaria in
pregnancy in Africa: a systematic review of the qualitative research. PloS one 2011;6(7):e22452 Pubmed Journal
207. Achan J, Talisuna AO, Erhart A, Yeka A, Tibenderana JK, Baliraine FN, et al. : Quinine, an old anti-malarial drug in a modern world:
role in the treatment of malaria. Malaria journal 2011;10 144 Pubmed Journal
208. Adegnika AA, Breitling LP, Agnandji ST, Chai SK, Schütte D, Oyakhirome S, et al. : Effectiveness of quinine monotherapy for the
treatment of Plasmodium falciparum infection in pregnant women in Lambaréné, Gabon. The American journal of tropical medicine and
hygiene 2005;73(2):263-6 Pubmed
209. Al Khaja KAJ, Sequeira RP : Drug treatment and prevention of malaria in pregnancy: a critical review of the guidelines. Malaria
journal 2021;20(1):62 Pubmed Journal
275 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
210. Hill J, D'Mello-Guyett L, Hoyt J, van Eijk AM, ter Kuile FO, Webster J : Women's access and provider practices for the case
management of malaria during pregnancy: a systematic review and meta-analysis. PLoS medicine 2014;11(8):e1001688 Pubmed Journal
211. Riley C, Dellicour S, Ouma P, Kioko U, ter Kuile FO, Omar A, et al. : Knowledge and Adherence to the National Guidelines for
Malaria Case Management in Pregnancy among Healthcare Providers and Drug Outlet Dispensers in Rural, Western Kenya. PloS one
2016;11(1):e0145616 Pubmed Journal
212. Rao VB, Jensen TO, Jimenez BC, Robays JO, Lasry E, Sterk E, et al. : Malaria in pregnancy: a call for a safe, efficient, and patient-
centred approach to first-trimester treatment. The Lancet. Global health 2018;6(6):e607-e608 Pubmed Journal
213. Gething PW, Elyazar IRF, Moyes CL, Smith DL, Battle KE, Guerra CA, et al. : A long neglected world malaria map: Plasmodium vivax
endemicity in 2010. PLoS neglected tropical diseases 2012;6(9):e1814 Pubmed Journal
214. Mendis K, Sina BJ, Marchesini P, Carter R : The neglected burden of Plasmodium vivax malaria. The American journal of tropical
medicine and hygiene 64(1-2 Suppl):97-106 Pubmed
215. Singh B, Kim Sung L, Matusop A, Radhakrishnan A, Shamsul SSG, Cox-Singh J, et al. : A large focus of naturally acquired
Plasmodium knowlesi infections in human beings. Lancet (London, England) 2004;363(9414):1017-24 Pubmed
216. Sutherland CJ, Tanomsing N, Nolder D, Oguike M, Jennison C, Pukrittayakamee S, et al. : Two nonrecombining sympatric forms of
the human malaria parasite Plasmodium ovale occur globally. The Journal of infectious diseases 2010;201(10):1544-50 Pubmed Journal
217. Douglas NM, Lampah DA, Kenangalem E, Simpson JA, Poespoprodjo JR, Sugiarto P, et al. : Major burden of severe anemia from
non-falciparum malaria species in Southern Papua: a hospital-based surveillance study. PLoS medicine 2013;10(12):e1001575;
discussion e1001575 Pubmed Journal
218. Poespoprodjo JR, Fobia W, Kenangalem E, Lampah DA, Hasanuddin A, Warikar N, et al. : Vivax malaria: a major cause of morbidity
in early infancy. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 2009;48(12):1704-12
Pubmed Journal
219. Genton B, D'Acremont V, Rare L, Baea K, Reeder JC, Alpers MP, et al. : Plasmodium vivax and mixed infections are associated with
severe malaria in children: a prospective cohort study from Papua New Guinea. PLoS medicine 2008;5(6):e127 Pubmed Journal
220. Kochar DK, Das A, Kochar SK, Saxena V, Sirohi P, Garg S, et al. : Severe Plasmodium vivax malaria: a report on serial cases from
Bikaner in northwestern India. The American journal of tropical medicine and hygiene 2009;80(2):194-8 Pubmed
221. Alexandre MA, Ferreira CO, Siqueira AM, Magalhães BL, Mourão MPG, Lacerda MV, et al. : Severe Plasmodium vivax malaria,
Brazilian Amazon. Emerging infectious diseases 2010;16(10):1611-4 Pubmed Journal
222. Nosten F, McGready R, Simpson JA, Thwai KL, Balkan S, Cho T, et al. : Effects of Plasmodium vivax malaria in pregnancy. Lancet
(London, England) 1999;354(9178):546-9 Pubmed
223. William T, Menon J, Rajahram G, Chan L, Ma G, Donaldson S, et al. : Severe Plasmodium knowlesi malaria in a tertiary care hospital,
Sabah, Malaysia. Emerging infectious diseases 2011;17(7):1248-55 Pubmed Journal
224. Barber BE, William T, Grigg MJ, Menon J, Auburn S, Marfurt J, et al. : A prospective comparative study of knowlesi, falciparum, and
vivax malaria in Sabah, Malaysia: high proportion with severe disease from Plasmodium knowlesi and Plasmodium vivax but no mortality
with early referral and artesunate therapy. Clinical infectious diseases : an official publication of the Infectious Diseases Society of
America 2013;56(3):383-97 Pubmed Journal
225. Grigg MJ, William T, Barber BE, Parameswaran U, Bird E, Piera K, et al. : Combining parasite lactate dehydrogenase-based and
histidine-rich protein 2-based rapid tests to improve specificity for diagnosis of malaria Due to Plasmodium knowlesi and other
Plasmodium species in Sabah, Malaysia. Journal of clinical microbiology 2014;52(6):2053-60 Pubmed Journal
226. Gogtay N, Kannan S, Thatte UM, Olliaro PL, Sinclair D : Artemisinin-based combination therapy for treating uncomplicated
Plasmodium vivax malaria. The Cochrane database of systematic reviews 2013;(10):CD008492 Pubmed Journal
276 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
227. Testing for G6PD deficiency for safe use of primaquine in radical cure of P. vivax and P. ovale (Policy brief). Geneva: World Health
Organization 2016; Website
228. Guide to G6PD deficiency rapid diagnostic testing to support P. vivax radical cure. World Health Organization, Geneva 2018;
Website
229. Milligan R, Daher A, Villanueva G, Bergman H, Graves P : Primaquine alternative dosing schedules for preventing malaria relapse in
people with Plasmodium vivax. Cochrane Database Syst Rev 2020; Pubmed Journal
230. Commons RJ, Simpson JA, Thriemer K, Chu CS, Douglas NM, Abreha T, et al. : The haematological consequences of Plasmodium
vivax malaria after chloroquine treatment with and without primaquine: a WorldWide Antimalarial Resistance Network systematic
review and individual patient data meta-analysis. BMC medicine 2019;17(1):151 Pubmed Journal
231. Galappaththy GNL, Tharyan P, Kirubakaran R : Primaquine for preventing relapse in people with Plasmodium vivax malaria treated
with chloroquine. The Cochrane database of systematic reviews 2013;(10):CD004389 Pubmed Journal
232. Radeva-Petrova D, Kayentao K, ter Kuile FO, Sinclair D, Garner P : Drugs for preventing malaria in pregnant women in endemic
areas: any drug regimen versus placebo or no treatment. The Cochrane database of systematic reviews 2014;(10):CD000169 Pubmed
Journal
233. Management of severe malaria - A practical handbook, 3rd edition. Geneva: World Health Organization 2013; Website
234. Sinclair D, Donegan S, Isba R, Lalloo DG : Artesunate versus quinine for treating severe malaria. The Cochrane database of
systematic reviews 2012;(6):CD005967 Pubmed Journal
235. Information note on delayed haemolytic anaemia following treatment with artesunate. Geneva: World Health Organization 2013;
Website
236. Hendriksen ICE, Mtove G, Kent A, Gesase S, Reyburn H, Lemnge MM, et al. : Population pharmacokinetics of intramuscular
artesunate in African children with severe malaria: implications for a practical dosing regimen. Clinical pharmacology and therapeutics
2013;93(5):443-50 Pubmed Journal
237. Zaloumis SG, Tarning J, Krishna S, Price RN, White NJ, Davis TME, et al. : Population pharmacokinetics of intravenous artesunate: a
pooled analysis of individual data from patients with severe malaria. CPT: pharmacometrics & systems pharmacology 2014;3 e145
Pubmed Journal
238. Esu E, Effa EE, Opie ON, Uwaoma A, Meremikwu MM : Artemether for severe malaria. The Cochrane database of systematic
reviews 2014;(9):CD010678 Pubmed Journal
239. Okebe J, Eisenhut M : Pre-referral rectal artesunate for severe malaria. The Cochrane database of systematic reviews
2014;(5):CD009964 Pubmed Journal
240. Rectal artesunate for pre-referral treatment of severe malaria. Geneva: World Health Organization 2017; Website
241. Prequalification programme: A United Nations programme managed by WHO. Geneva: World Health Organization 2009; Website
242. Methods for surveillance of antimalarial drug efficacy. Geneva: World Health Organization 2009; Website
243. Methods and techniques for clinical trials on antimalarial drug efficacy: Genotyping to identify parasite populations. World Health
Organization, Geneva 2008; Website
244. Report on antimalarial drug efficacy, resistance and response: 10 years of surveillance (2010-2019). Geneva: World Health
Organization 2020; Website
277 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
245. Ajayi IO, Browne EN, Bateganya F, Yar D, Happi C, Falade CO, et al. : Effectiveness of artemisinin-based combination therapy used
in the context of home management of malaria: a report from three study sites in sub-Saharan Africa. Malaria journal 2008;7 190
Pubmed Journal
246. A practical handbook on the pharmacovigilance of antimalarial medicines. Geneva: World Health Organization 2008; Website
247. Stresman G, Whittaker C, Slater HC, Bousema T, Cook J : Quantifying Plasmodium falciparum infections clustering within
households to inform household-based intervention strategies for malaria control programs: An observational study and meta-analysis
from 41 malaria-endemic countries. PLoS medicine 2020;17(10):e1003370 Pubmed Journal
248. Sandfort M, Vantaux A, Kim S, Obadia T, Pepey A, Gardais S, et al. : Forest malaria in Cambodia: the occupational and spatial
clustering of Plasmodium vivax and Plasmodium falciparum infection risk in a cross-sectional survey in Mondulkiri province, Cambodia.
Malaria journal 2020;19(1):413 Pubmed Journal
249. Smith C, Whittaker M : Malaria elimination without stigmatization: a note of caution about the use of terminology in elimination
settings. Malaria journal 2014;13 377 Pubmed Journal
250. International health regulations (2005) - 3rd ed. Geneva: World Health Organization 2016; Website
251. Communicable disease surveillance and response systems: guide to monitoring and evaluating. Geneva: World Health Organization
2006; Website
252. WHO technical brief for countries preparing malaria funding requests for the Global Fund (2020-2022). Geneva: World Health
Organization 2020; Website
253. Alonso-Coello P, Schünemann HJ, Moberg J, Brignardello-Petersen R, Akl EA, Davoli M, et al. : [GRADE Evidence to Decision (EtD)
frameworks: a systematic and transparent approach to making well informed healthcare choices. 1: Introduction]. Gaceta sanitaria
32(2):166.e1-166.e10 Pubmed Journal
254. GRADE Handbook: Introduction to GRADE Handbook. Grading of Recommendations, Assessment, Development and Evaluation
(GRADE) Working Group 2013; Website
255. Schandelmaier S, Briel M, Varadhan R, Schmid CH, Devasenapathy N, Hayward RA, et al. : Development of the Instrument to
assess the Credibility of Effect Modification Analyses (ICEMAN) in randomized controlled trials and meta-analyses. CMAJ : Canadian
Medical Association journal = journal de l'Association medicale canadienne 2020;192(32):E901-E906 Pubmed Journal
256. Santesso N, Glenton C, Dahm P, Garner P, Akl EA, Alper B, et al. : GRADE guidelines 26: informative statements to communicate
the findings of systematic reviews of interventions. Journal of clinical epidemiology 2020;119 126-135 Pubmed Journal
257. World Health Organization : Meeting of Strategic Advisory Group of Experts on Immunization, October 2021: conclusions and
recommendations. Weekly Epidemiological Record 2021;96(50):613-632 Website
258. 20th meeting of the Malaria Policy Advisory Group (MPAG). Geneva: World Health Organization 2021; Website
259. WHO malaria terminology, 2021 update. Geneva: World Health Organization 2021; Website
278 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
1. ABBREVIATIONS
2. EXECUTIVE SUMMARY
3. INTRODUCTION
4. PREVENTION
Summary
Of the 23 included studies, 21 were cluster RCTs (six with households as the cluster and 15 with villages as the
cluster) and two were individual RCTs; 12 studies compared ITNs with untreated nets, and 11 studies compared
ITNs with no nets. Based on WHO regions, 12 studies were conducted in Africa (Burkina Faso, Republic of Côte
d’Ivoire, the Republic of Cameroon, Republic of the Gambia [two studies], Republic of the Ghana, the Republic of
Kenya [three studies], the Republic of Madagascar, the Republic of Sierra Leone and the United Republic of
Tanzania), six in the Americas (the Bolivarian Republic of Venezuela, the Republic of Colombia, the Republic of
Ecuador, the Republic of Nicaragua [two studies] and the Republic of Peru), four in South-East Asia (Republic of
India, the Republic of Union of Myanmar, The Kingdom of Thailand [two studies]) and one in the Eastern
Mediterranean (the Islamic Republic of Pakistan).
279 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Pyrethroid- the Evidence Plain language
No nets or
Timeframe measurements treated nets or (Quality of summary
curtains
curtains evidence)
All-cause
Relative risk 0.83 33 27 Pyrethroid-only nets or
(CI 95% 0.77 — 0.89) per 1000 per 1000
mortality curtains reduce the
Based on data from
129,714 participants in High child mortality from all
Difference: 6 fewer per 1000 causes compared to no
5 studies. (Randomized ( CI 95% 8 fewer nets or curtains.
controlled) — 4 fewer )
120 83
Relative risk 0.69 per 1000 per 1000 Pyrethroid-only nets or
P. falciparum (CI 95% 0.54 — 0.89) curtains reduce the
prevalence Based on data from prevalence of P.
Difference: 37 fewer per High
17,860 participants in 5 falciparum malaria
1000
studies. (Randomized compared to no nets or
( CI 95% 55
controlled) curtains.
fewer — 13 fewer
)
280 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Pyrethroid- the Evidence Plain language
No nets or
Timeframe measurements treated nets or (Quality of summary
curtains
curtains evidence)
Severe malaria
Relative risk 0.56 15 8 Pyrethroid-only nets or
(CI 95% 0.38 — 0.82) per 1000 per 1000 curtains reduce the
episodes Based on data from incidence of severe
31,173 participants in 2 High
Difference: 7 fewer per 1000 malaria episodes
studies. (Randomized compared to no nets or
( CI 95% 9 fewer
controlled) curtains.
— 3 fewer )
Attached Images
References
55. Pryce J, Richardson M, Lengeler C : Insecticide-treated nets for preventing malaria. Cochrane Database of
Systematic Reviews 2018;(11): Pubmed Journal Website
Summary
Of the 23 included studies, 21 were cluster RCTs (six with households as the cluster and 15 with villages as the
cluster) and two were individual RCTs; 12 studies compared ITNs with untreated nets, and 11 studies compared
ITNs with no nets. Based on WHO regions, 12 studies were conducted in Africa (Burkina Faso, Republic of Cote
d’Ivoire, Cameroon, Gambia (two studies), Ghana, Kenya (three studies), Madagascar, Sierra Leone, United Republic
of Tanzania), six in the Americas (Colombia, Ecuador, Nicaragua (two studies), Peru and Venezuela) and four in South-
East Asia (India, Myanmar, Thailand (two studies)) and one in the Eastern Mediterranean (Pakistan).
281 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Pyrethroid-only nets or curtains reduce the prevalence of P. falciparum malaria compared to untreated nets or
curtains.
(Risk ratio: 0.81; 95% CI (0.68–0.97); four studies; high certainty evidence)
Pyrethroid-only nets or curtains may reduce the incidence of uncomplicated P. vivax malaria episodes compared to
untreated nets or curtains.
(Rate ratio: 0.73; 95% CI (0.51–1.05); three studies; low certainty evidence)
he evidence is very uncertain about the effect of pyrethroid-only nets or curtains on P. vivax prevalence compared to
untreated nets or curtains.
(Risk ratio: 0.52; 95% CI (0.13–2.04); two studies; very low certainty evidence)
Intervention Certainty of
Comparator
Outcome Study results and Pyrethroid- the Evidence Plain language
Untreated nets
Timeframe measurements only nets or (Quality of summary
or curtains
curtains evidence)
All-cause
Relative risk 0.67 19 13 Pyrethroid-only nets or
(CI 95% 0.36 — 1.23) per 1000 per 1000 curtains probably
mortality Moderate
Based on data from reduce all-cause child
Due to serious
32,721 participants in 2 mortality compared to
Difference: 6 fewer per 1000 imprecision 1
studies. (Randomized untreated nets or
( CI 95% 12
controlled) curtains.
fewer — 4 more )
85 69 Pyrethroid-only nets or
P. falciparum Relative risk 0.81 per 1000 per 1000 curtains reduce the
prevalence (CI 95% 0.68 — 0.97)
prevalence of P.
Based on data from 300 High
Difference: 16 fewer per falciparum malaria
participants in 4 studies.
1000 compared to untreated
(Randomized controlled) ( CI 95% 27 nets or curtains.
fewer — 3 fewer )
282 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Pyrethroid- the Evidence Plain language
Untreated nets
Timeframe measurements only nets or (Quality of summary
or curtains
curtains evidence)
Any
Plasmodium
69 32
per 1000 per 1000 Pyrethroid-only nets or
spp. Relative risk 0.47
curtains probably
uncomplicated (CI 95% 0.17 — 1.28)
Difference: 37 fewer per Moderate reduce the incidence of
episodes Based on data from
1000 Due to serious uncomplicated malaria
(cumulative 7,082 participants in 2
( CI 95% 57 imprecision 5 episodes compared to
incidence) studies. (Randomized
fewer — 19 more untreated nets or
controlled)
) curtains.
1. Imprecision: serious.
2. Indirectness: serious. Imprecision: serious.
3. Inconsistency: serious. Imprecision: serious.
4. Indirectness: very serious. Imprecision: very serious.
5. Imprecision: serious.
6. Indirectness: very serious. Imprecision: serious.
Attached Images
References
55. Pryce J, Richardson M, Lengeler C : Insecticide-treated nets for preventing malaria. Cochrane Database of
Systematic Reviews 2018;(11): Pubmed Journal Website
283 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Summary
Two cRCTs from the Republic of Uganda and the United Republic of Tanzania were included in the review.
Certainty of
Comparator Intervention
Outcome Study results and the Evidence Plain language
Pyrethroid- Pyrethroid-
Timeframe measurements (Quality of summary
only LLINs PBO ITNs
evidence)
Pyrethroid-PBO ITNs
Parasite Odds ratio 0.88 248 225 probably reduce malaria
prevalence - 16 (CI 95% 0.74 — 1.04) per 1000 per 1000 parasite prevalence in
Moderate
to 18 months Based on data from areas of high insecticide
Difference: 23 fewer per Due to serious
11,822 participants in 2 resistance at 16- to
1000 inconsistency 2 18-month follow-up
studies. (Randomized
controlled) ( CI 95% 52 compared to
fewer — 7 more ) pyrethroid-only LLINs.
Pyrethroid-PBO ITNs
Parasite Odds ratio 0.79 350 298 probably reduce malaria
prevalence - 21 (CI 95% 0.67 — 0.95) per 1000 per 1000 parasite prevalence in
Moderate
to 25 months Based on data from areas of high insecticide
Difference: 52 fewer per Due to serious
10,603 participants in 2 resistance at 21- to
1000 inconsistency 3 25-month follow-up
studies. (Randomized
controlled) ( CI 95% 85 compared to
fewer — 12 fewer pyrethroid-only LLINs.
284 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Comparator Intervention
Outcome Study results and the Evidence Plain language
Pyrethroid- Pyrethroid-
Timeframe measurements (Quality of summary
only LLINs PBO ITNs
evidence)
Attached Images
References
59. Gleave K, Lissenden N, Richardson M, Choi L, Ranson H : Piperonyl butoxide (PBO) combined with pyrethroids in
insecticide-treated nets to prevent malaria in Africa. The Cochrane Database of Systematic Reviews 2021;5
CD012776 Pubmed Journal
60. Piperonyl butoxide (PBO) combined with pyrethroids in insecticide-treated nets to prevent malaria in Africa.
2021; Pubmed
Summary
Ten experimental hut trials from Republic of Benin, Burkina Faso, Republic of Cameroon, Republic of Côte d’Ivoire
and United Republic of Tanzania were included in the review.
Certainty of
Comparator Intervention
Outcome Study results and the Evidence Plain language
Pyrethroid- Pyrethroid-
Timeframe measurements (Quality of summary
only LLINs PBO ITNs
evidence)
Mosquito High
mortality -
Relative risk 1.84
(CI 95% 1.6 — 2.11)
238 438 Not downgraded
Unwashed
pyrethroid-PBO ITNs
285 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Comparator Intervention
Outcome Study results and the Evidence Plain language
Pyrethroid- Pyrethroid-
Timeframe measurements (Quality of summary
only LLINs PBO ITNs
evidence)
1. Systematic review [60] with included studies: Toé 2018, Toé 2018, Koudou 2011, Toé 2018, Bayili 2017, Pennetier
2013, Corbel 2010, Bayili 2017, Toé 2018. Baseline/comparator: Control arm of reference used for intervention.
2. Systematic review [60] with included studies: Koudou 2011, Pennetier 2013, Bayili 2017, Bayili 2017, Corbel 2010.
Baseline/comparator: Control arm of reference used for intervention.
3. Systematic review [60] with included studies: Bayili 2017, Toé 2018, Toé 2018, Pennetier 2013, Toé 2018, Corbel
2010, Bayili 2017, Toé 2018. Baseline/comparator: Control arm of reference used for intervention.
4. Systematic review [60] with included studies: Pennetier 2013, Corbel 2010, Bayili 2017, Bayili 2017. Baseline/
comparator: Control arm of reference used for intervention.
Attached Images
References
59. Gleave K, Lissenden N, Richardson M, Choi L, Ranson H : Piperonyl butoxide (PBO) combined with pyrethroids in
insecticide-treated nets to prevent malaria in Africa. The Cochrane Database of Systematic Reviews 2021;5
CD012776 Pubmed Journal
286 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
60. Piperonyl butoxide (PBO) combined with pyrethroids in insecticide-treated nets to prevent malaria in Africa.
2021; Pubmed
Summary
The systematic review [Barker et al unpublished evidence] included two RCTs, one from Benin [62] and one from the
United Republic of Tanzania [61] that compared the epidemiological impact against malaria of pyrethroid-
chlorfenapyr ITNs (alphacypermethrin-chlorfenapyr) against pyrethroid-only LLINs (alphacypermethrin). Both trials
were conducted in areas of high malaria transmission and pyrethroid-resistance. The review provided high to
moderate certainty evidence that incidence of clinical malaria was lower in areas where pyrethroid-chlorfenapyr
ITNs were deployed than in those with pyrethroid-only LLINs, at one and two years after ITN deployment (one-year
incidence rate ratio (IRR): 0.44; 95% CI: 0.37–0.52; two-year IRR: 0.57; 95% CI: 0.51–0.63). The review also
provided high certainty evidence that prevalence of malaria infection was lower where pyrethroid-chlorfenapyr ITNs
were deployed than in those with pyrethroid-only LLINs, at several time points after ITN deployment (six-month
relative risk (RR): 0.50; 95% CI: 0.43–0.59; 12-month RR: 0.78; 95% CI: 0.72–0.85; 18-month RR: 075; 95% CI:
0.70–0.80; 24-month RR: 0.56; 95% CI: 0.50–0.63).
Intervention Certainty of
Comparator
Outcome Study results and Pyrethroid- the Evidence Plain language
Pyrethroid-
Timeframe measurements chlorfenapyr (Quality of summary
only ITNs
ITNs evidence)
2000-person years (2
678 487 RCTs) Length of time
per 1000 per 1000 observed: <1 month to
Malaria case Rate ratio 0.72
24 months Based on
incidence (CI 95% 0.67 — 0.78) Difference: 190 fewer per Moderate data from at least
(overall) Based on data from 1000 Due to serious 61,183 participants (1
61,183 participants in 2
( CI 95% 224 inconsistency. 1 study) Absolute
studies. (Randomized fewer — 149 calculation performed
controlled) fewer ) manually as GRADEPro
cannot calculate using
IRR
2000-person years (2
487 213 RCTs) Length of time
Malaria case per 1000 per 1000 observed: <1 month to
Rate ratio 0.44
incidence (CI 95% 0.37 — 0.52)
12 months Based on
(1-year post- Difference: 272 fewer per data from at least
Based on data from 1000
intervention) 61,183 participants in 2 High 61,183 participants (1
( CI 95% 307 study) Absolute
studies. (Randomized fewer — 234 calculation performed
controlled) fewer ) manually as GRADEPro
cannot calculate using
IRR
287 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Pyrethroid- the Evidence Plain language
Pyrethroid-
Timeframe measurements chlorfenapyr (Quality of summary
only ITNs
ITNs evidence)
1. Inconsistency: serious. Point estimates vary widely (from 0.49 to 0.87 with no overlap of confidence intervals). This
heterogeneity appears to be unexplained but important (chi2, p <0.0001, I2 = 98%). But may not impact on a
recommendation for the intervention. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious.
Attached Images
288 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
References
61. Mosha JF, Kulkarni MA, Lukole E, Matowo NS, Pitt C, Messenger LA, et al. : Effectiveness and cost-effectiveness
against malaria of three types of dual-active-ingredient long-lasting insecticidal nets (LLINs) compared with
pyrethroid-only LLINs in Tanzania: a four-arm, cluster-randomised trial. Lancet (London, England)
2022;399(10331):1227-1241 Pubmed Journal
62. Accrombessi M, Cook J, Dangbenon E, Yovogan B, Akpovi H, Sovi A, et al. : Efficacy of pyriproxyfen-pyrethroid
long-lasting insecticidal nets (LLINs) and chlorfenapyr-pyrethroid LLINs compared with pyrethroid-only LLINs for
malaria control in Benin: a cluster-randomised, superiority trial. Lancet (London, England) 2023;401(10375):435-446
Pubmed Journal
Summary
The review [Barker et al unpublished evidence] compared the epidemiological impact against malaria of pyrethroid-
chlorfenapyr ITNs against pyrethroid-PBO ITNs (permethrin-piperonyl butoxide), based on one RCT [61] in the
United Republic of Tanzania. The review provided high to low certainty evidence that incidence of clinical malaria
was lower in areas where pyrethroid-chlorfenapyr ITNs were deployed than in those with pyrethroid-PBO ITNs, at
two years after ITN deployment, but possibly not at one year post-deployment (one-year IRR: 0.98; 95% CI:
0.71–1.36; two-year IRR: 0.65; 95% CI: 0.55–0.77). The review also provided high to moderate certainty evidence
that prevalence of malaria infection was generally lower where pyrethroid-chlorfenapyr ITNs were deployed,
compared to those with pyrethroid-only LLINs, at several time points after ITN deployment (12-month RR: 0.81;
95% CI: 0.68–0.98; 18-month RR: 0.94; 95% CI: 0.86–1.04; 24-month RR: 0.63; 95% CI: 0.56–0.71).
Intervention
Comparator
Pyrethroid- Certainty of
Pyrethroid-
Outcome Study results and chlorfenapyr the Evidence Plain language
PBO nets for
Timeframe measurements nets for (Quality of summary
prevention of
prevention of evidence)
malaria
malaria
2000-person years (1
333 227 RCT) Length of time
per 1000 per 1000 observed: <1 month to
Malaria case Rate ratio 0.68
24 months Based on
incidence (CI 95% 0.59 — 0.79) Difference: 107 fewer per data from at least
(overall) Based on data from 1000
61,183 participants in 1 High 61,183 participants (1
( CI 95% 137 study) Absolute
studies. (Randomized fewer — 70 fewer calculation performed
controlled) ) manually as GRADEPro
cannot calculate using
IRR
Malaria case
incidence
Rate ratio 0.98 133 131 2000-person years (1
RCT) Length of time
(CI 95% 0.71 — 1.36) per 1000 per 1000 Low
(1-year post- observed: <1 month to
Based on data from Due to very
intervention) 12 months Based on
61,183 participants in 1 Difference: 3 fewer per 1000 serious
data from at least
studies. (Randomized ( CI 95% 39 imprecision 1 61,183 participants (1
controlled) fewer — 48 more study) Absolute
289 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention
Comparator
Pyrethroid- Certainty of
Pyrethroid-
Outcome Study results and chlorfenapyr the Evidence Plain language
PBO nets for
Timeframe measurements nets for (Quality of summary
prevention of
prevention of evidence)
malaria
malaria
) calculation performed
manually as GRADEPro
cannot calculate using
IRR
2000-person years (1
483 315 RCT) Length of time
Malaria case per 1000 per 1000 observed: 12 months to
Rate ratio 0.65
incidence (CI 95% 0.55 — 0.77)
24 months Based on
(2-years post- Difference: 155 fewer per data from at least
Based on data from 1000
intervention) 61,183 participants in 1 High 61,183 participants (1
( CI 95% 198 study) Absolute
studies. (Randomized fewer — 101 calculation performed
controlled) fewer ) manually as GRADEPro
cannot calculate using
IRR
Parasite
Relative risk 0.81 192 156
prevalence (CI 95% 0.68 — 0.98) per 1000 per 1000
(12-months Moderate
Based on data from
follow-up) Difference: 37 fewer per Due to serious
2,197 participants in 1
1000 imprecision 2
studies. (Randomized
controlled) ( CI 95% 62
fewer — 4 fewer )
1. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Confidence intervals are very wide (39
fewer to 48 more) and may have crossed many important decision-making threshold (including line of no effect).
Publication bias: no serious.
2. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Confidence intervals are wide (62 fewer to
4 fewer) and may have crossed many important decision-making threshold. Publication bias: no serious.
3. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Confidence intervals are wide (from 61
fewer to 17 more) and may have crossed many important decision-making threshold (including line of no effect).
Publication bias: no serious.
290 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Attached Images
References
61. Mosha JF, Kulkarni MA, Lukole E, Matowo NS, Pitt C, Messenger LA, et al. : Effectiveness and cost-effectiveness
against malaria of three types of dual-active-ingredient long-lasting insecticidal nets (LLINs) compared with
pyrethroid-only LLINs in Tanzania: a four-arm, cluster-randomised trial. Lancet (London, England)
2022;399(10331):1227-1241 Pubmed Journal
62. Accrombessi M, Cook J, Dangbenon E, Yovogan B, Akpovi H, Sovi A, et al. : Efficacy of pyriproxyfen-pyrethroid
long-lasting insecticidal nets (LLINs) and chlorfenapyr-pyrethroid LLINs compared with pyrethroid-only LLINs for
malaria control in Benin: a cluster-randomised, superiority trial. Lancet (London, England) 2023;401(10375):435-446
Pubmed Journal
Summary
The systematic review [Barker et al unpublished evidence] included three trials from Benin [62], Burkina Faso [63] and
the United Republic of Tanzania [61] that compared the epidemiological impact against malaria of pyrethroid-
pyriproxyfen ITNs (either alphacypermethrin-pyriproxyfen or permethrin-pyriproxyfen) against that of pyrethroid-
only LLINs (either permethrin or alphacypermethrin). All three trials were conducted in areas of high malaria
transmission and pyrethroid-resistance. The review provided high-certainty evidence that incidence of clinical
malaria was lower in areas where pyrethroid-pyriproxyfen ITNs were deployed, compared to where pyrethroid-only
LLINs were deployed, at one and two years after ITN deployment (one-year incidence rate ratio (IRR): 0.81; 95% CI:
0.70–0.93; two-year IRR: 0.87; 95% CI: 0.80–0.95). The review also provided moderate to high certainty evidence
that prevalence of malaria infection was lower in areas where pyrethroid-pyriproxyfen ITNs were deployed than in
those where pyrethroid-only LLINs were deployed, at some, but not all, time points after ITN deployment (six-month
relative risk (RR): 0.96; 95% CI: 0.85–1.08; 12-month RR: 0.70; 95% CI: 0.60–0.80; 18-month RR: 0.98; 95% CI:
0.92–1.04; 24-month RR: 0.82; 95% CI: 0.75–0.90).
Intervention
Comparator
Pyrethroid- Certainty of
Pyrethroid-
Outcome Study results and pyriproxyfen the Evidence Plain language
only nets for
Timeframe measurements nets for (Quality of summary
prevention of
prevention of evidence)
malaria
malaria
2000-person years (3
1,037 929 RCTs); Length of time
per 1000 per 1000 observed: 5 months to
Malaria case Rate ratio 0.86
24 months; Based on
incidence (CI 95% 0.81 — 0.92) Difference: 145 fewer per data from at least
(overall) Based on data from 1000
63,163 participants in 2 High 63,163 participants (2
( CI 95% 197 studies); Absolute
studies. (Randomized fewer — 83 fewer calculation performed
controlled) ) manually as GRADEPro
cannot calculate using
IRR.
291 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention
Comparator
Pyrethroid- Certainty of
Pyrethroid-
Outcome Study results and pyriproxyfen the Evidence Plain language
only nets for
Timeframe measurements nets for (Quality of summary
prevention of
prevention of evidence)
malaria
malaria
2000 person-years; (2
487 393 RCTs); Length of time
Malaria case per 1000 per 1000 observed: < 1 month to
Rate ratio 0.81
incidence (CI 95% 0.7 — 0.93)
12 months Based on
(1-year post- Difference: 92 fewer per data from at least 61
Based on data from 1000
intervention) 61,183 participants in 1 High 183 participants (1
( CI 95% 146 study); Absolute
studies. (Randomized fewer — 34 fewer calculation performed
controlled) ) manually as GRADEPro
cannot calculate using
IRR.
Parasite
Relative risk 0.98 448 438
prevalence (CI 95% 0.92 — 1.04) per 1000 per 1000
(18-months Moderate
Based on data from
follow-up) Difference: 9 fewer per 1000 Due to serious
5,337 participants in 2
( CI 95% 36 imprecision. 2
studies. (Randomized
controlled) fewer — 18 more
)
292 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention
Comparator
Pyrethroid- Certainty of
Pyrethroid-
Outcome Study results and pyriproxyfen the Evidence Plain language
only nets for
Timeframe measurements nets for (Quality of summary
prevention of
prevention of evidence)
malaria
malaria
( CI 95% 114
controlled) fewer — 46 fewer
)
1. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Confidence intervals are wide (from 42
fewer to 22 more) and may have crossed many important decision-making thresholds (including line of no effect).
Publication bias: no serious.
2. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Confidence intervals are wide (from 36
fewer to 18 more) and may have crossed many important decision-making thresholds (including line of no effect).
Publication bias: no serious.
Attached Images
References
61. Mosha JF, Kulkarni MA, Lukole E, Matowo NS, Pitt C, Messenger LA, et al. : Effectiveness and cost-effectiveness
against malaria of three types of dual-active-ingredient long-lasting insecticidal nets (LLINs) compared with
pyrethroid-only LLINs in Tanzania: a four-arm, cluster-randomised trial. Lancet (London, England)
2022;399(10331):1227-1241 Pubmed Journal
62. Accrombessi M, Cook J, Dangbenon E, Yovogan B, Akpovi H, Sovi A, et al. : Efficacy of pyriproxyfen-pyrethroid
long-lasting insecticidal nets (LLINs) and chlorfenapyr-pyrethroid LLINs compared with pyrethroid-only LLINs for
malaria control in Benin: a cluster-randomised, superiority trial. Lancet (London, England) 2023;401(10375):435-446
Pubmed Journal
63. Tiono AB, Ouédraogo A, Ouattara D, Bougouma EC, Coulibaly S, Diarra A, et al. : Efficacy of Olyset Duo, a
bednet containing pyriproxyfen and permethrin, versus a permethrin-only net against clinical malaria in an area with
highly pyrethroid-resistant vectors in rural Burkina Faso: a cluster-randomised controlled trial. Lancet (London,
England) 2018;392(10147):569-580 Pubmed Journal
Summary
The review [Barker et al unpublished evidence] compared pyrethroid-pyriproxyfen ITNs (alphacypermethrin-
pyriproxyfen) to pyrethroid-PBO ITNs (permethrin-piperonyl butoxide) in terms of their epidemiological impact
against malaria, based on only one trial [61] conducted in the United Republic of Tanzania. The review provided high
to moderate certainty evidence that incidence of clinical malaria was higher at one year after ITN deployment (IRR:
2.04; 95% CI: 1.55–2.68) in areas where pyrethroid-pyriproxyfen ITNs were deployed that in those where
pyrethroid-PBO ITNs were deployed; there was little or no effect on malaria incidence two years post-deployment
(IRR: 1.10; 95% CI: 0.95–1.27). The review also provided high- to moderate-certainty evidence that pyrethroid-
pyriproxyfen only performed as well as, or worse than, pyrethroid-PBO ITNs in reducing prevalence of malaria
infection at all time points after ITN deployment (12-month RR: 1.13; 95% CI: 0.95–1.33; 18-month RR: 1.17; 95%
CI: 1.07–1.27; 24-month RR: 0.88; 95% CI: 0.75–1.03).
293 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention
Comparator
Pyrethroid- Certainty of
Pyrethroid-
Outcome Study results and pyriproxyfen the Evidence Plain language
PBO nets for
Timeframe measurements nets for (Quality of summary
prevention of
prevention of evidence)
malaria
malaria
2000-person years (1
333 416 RCT); Length of time
per 1000 per 1000 observed: <1 month to
Malaria case Rate ratio 1.25
24 months; Based on
incidence (CI 95% 1.1 — 1.41) Difference: 83 more per data from at least
(overall) Based on data from 1000
61,183 participants in 1 High 61,183 participants (1
( CI 95% 33 more study); Absolute
studies. (Randomized — 137 more ) calculation performed
controlled)
manually as GRADEPro
cannot calculate using
IRR.
2000-person years (1
131 266 RCT) Length of time
Malaria case per 1000 per 1000 observed: <1 month to
Rate ratio 2.04
incidence (CI 95% 1.55 — 2.68)
12 months Based on
(1-year post- Difference: 136 more per data from at least
Based on data from 1000
intervention) 61,183 participants in 1 High 61,183 participants (1
( CI 95% 72 more study) Absolute
studies. (Randomized — 220 more ) calculation performed
controlled)
manually as GRADEPro
cannot calculate using
IRR
2000-person years (1
483 531 RCT) Length of time
Malaria case per 1000 per 1000 observed: 12 months to
Rate ratio 1.1
incidence (CI 95% 0.95 — 1.27)
24 months Based on
(2-years post- Difference: 48 more per Moderate data from at least
Based on data from 1000
intervention) Due to serious 61,183 participants (1
61,183 participants in 1
( CI 95% 24 imprecision 1 study) Absolute
studies. (Randomized fewer — 130 calculation performed
controlled) more ) manually as GRADEPro
cannot calculate using
IRR
Parasite
Relative risk 1.17 433 506
prevalence (CI 95% 1.07 — 1.27) per 1000 per 1000
(18-months Based on data from
follow-up) 2,313 participants in 1 Difference: 74 more per High
studies. (Randomized 1000
controlled) ( CI 95% 30 more
— 117 more )
294 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention
Comparator
Pyrethroid- Certainty of
Pyrethroid-
Outcome Study results and pyriproxyfen the Evidence Plain language
PBO nets for
Timeframe measurements nets for (Quality of summary
prevention of
prevention of evidence)
malaria
malaria
1. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Confidence intervals are very wide (from 24
fewer 130 more) and may have crossed many important decision-making thresholds (including line of no effect).
Publication bias: no serious.
2. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Confidence intervals are wide (from 10
fewer to 63 more) and may have crossed many important decision-making thresholds (including line of no effect).
Publication bias: no serious.
3. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Confidence intervals are wide (from 67
fewer to 7 more) and may have crossed many important decision-making thresholds (including line of no effect).
Publication bias: no serious.
Attached Images
References
61. Mosha JF, Kulkarni MA, Lukole E, Matowo NS, Pitt C, Messenger LA, et al. : Effectiveness and cost-effectiveness
against malaria of three types of dual-active-ingredient long-lasting insecticidal nets (LLINs) compared with
pyrethroid-only LLINs in Tanzania: a four-arm, cluster-randomised trial. Lancet (London, England)
2022;399(10331):1227-1241 Pubmed Journal
62. Accrombessi M, Cook J, Dangbenon E, Yovogan B, Akpovi H, Sovi A, et al. : Efficacy of pyriproxyfen-pyrethroid
long-lasting insecticidal nets (LLINs) and chlorfenapyr-pyrethroid LLINs compared with pyrethroid-only LLINs for
malaria control in Benin: a cluster-randomised, superiority trial. Lancet (London, England) 2023;401(10375):435-446
Pubmed Journal
63. Tiono AB, Ouédraogo A, Ouattara D, Bougouma EC, Coulibaly S, Diarra A, et al. : Efficacy of Olyset Duo, a
bednet containing pyriproxyfen and permethrin, versus a permethrin-only net against clinical malaria in an area with
highly pyrethroid-resistant vectors in rural Burkina Faso: a cluster-randomised controlled trial. Lancet (London,
England) 2018;392(10147):569-580 Pubmed Journal
Summary
Of the four included ITN studies, two were cluster RCTs (one with households as the cluster and one with villages as
the cluster) and two were individual-level RCTs. The two individual-level RCTs were conducted on the
Myanmar–Thailand border, the village-level RCT was conducted in the Republic of Union of the Myanmar and the
295 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements no ITNs ITNs (Quality of summary
evidence)
70 39
P. falciparum Relative risk 0.55 per 1000 per 1000
ITNs reduce P.
case incidence (CI 95% 0.37 — 0.79)
Difference: 31 fewer per falciparum case
Based on data from High
1000 incidence compared to
3,200 participants in 4
no ITNs.
studies. ( CI 95% 44
fewer — 15 fewer
)
37 22
P. falciparum Relative risk 0.6 per 1000 per 1000
prevalence (CI 95% 0.4 — 0.88) ITNs reduce P.
Based on data from High falciparum prevalence
Difference: 15 fewer per
2,079 participants in 2 compared to no ITNs.
1000
studies. ( CI 95% 22
fewer — 4 fewer )
116 80
P. vivax case Relative risk 0.69 per 1000 per 1000
incidence (CI 95% 0.51 — 0.94) Moderate ITNs probably reduce P.
Based on data from Due to serious vivax case incidence
Difference: 36 fewer per
2,997 participants in 3 imprecision compared to no ITNs.
1000
studies. ( CI 95% 57
fewer — 7 fewer )
99 99
P. vivax Relative risk 1 per 1000 per 1000 Low ITNs may result in little
prevalence (CI 95% 0.75 — 1.34)
Due to very to no difference in P.
Based on data from
Difference: 0 fewer per 1000 serious vivax prevalence
2,079 participants in 2
( CI 95% 25 imprecision compared to no ITNs.
studies. fewer — 34 more
)
Attached Images
296 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
References
54. Messenger LA, Furnival-Adams J, Pelloquin B, Rowland M : Vector control for malaria prevention during
humanitarian emergencies: protocol for a systematic review and meta-analysis. BMJ Open 2021/07/
27;11(7):e046325-e046325 Pubmed Journal Website
Summary
The systematic review included 1 RCT from the United Republic of Tanzania that reported the effect of IRS on
malaria in an area of intense malaria transmission and another RCT from Islamic Republic of Pakistan that
investigated the epidemiological impact of IRS in an area with unstable malaria.
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No IRS IRS (Quality of summary
evidence)
Incidence of
malaria in
650 560
per 1000 per 1000
children under
Relative risk 0.86 Low IRS may reduce P.
5 years in areas (CI 95% 0.77 — 0.95) Due to serious
Difference: 90 fewer per falciparum incidence
of intense Based on data from 884 1000 indirectness, Due compared to no IRS in
malaria participants in 1 studies. ( CI 95% 150 to serious areas of intense malaria
transmission (Randomized controlled) fewer — 40 fewer imprecision 1 transmission.
)
297 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No IRS IRS (Quality of summary
evidence)
Incidence of 50 10
malaria in all Relative risk 0.12 per 1000 per 1000 Low IRS may reduce P.
ages in areas of (CI 95% 0.04 — 0.31)
Due to serious falciparum incidence
unstable Based on data from
Difference: 40 fewer per indirectness, Due compared to no IRS in
malaria 18,261 participants in 1
1000 to serious areas of unstable
studies. (Randomized ( CI 95% 50 imprecision 3 malaria
controlled) fewer — 40 fewer
)
Parasite
prevalence in
110 30
per 1000 per 1000
children aged Relative risk 0.24
Low IRS may reduce P.
5–15 years in (CI 95% 0.17 — 0.34)
Difference: 80 fewer per Due to serious falciparum prevalence
areas of Based on data from
1000 indirectness, Due compared to no IRS in
unstable 2,359 participants in 1
( CI 95% 90 to serious areas of unstable
malaria studies. (Randomized
fewer — 70 fewer imprecision 4 malaria
controlled)
)
Attached Images
References
70. Pluess B, Tanser FC, Lengeler C, Sharp BL : Indoor residual spraying for preventing malaria. The Cochrane
database of systematic reviews 2010;(4):CD006657 Pubmed Journal
Summary
The systematic review included 1 RCT from the United Republic of Tanzania that reported the effect of IRS
compared to ITNs on malaria in an area of intense malaria transmission and another study from the Republic of India
that investigated the epidemiological impact of IRS in an area with unstable malaria.
298 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements ITNs IRS (Quality of summary
evidence)
Incidence of
malaria in
630 550
per 1000 per 1000
children under
Relative risk 0.88 Low IRS may reduce P.
5 years in areas (CI 95% 0.78 — 0.98) Due to serious
Difference: 80 fewer per falciparum incidence
of intense Based on data from 818 1000 indirectness, Due compared to no ITNs in
malaria participants in 1 studies. ( CI 95% 140 to serious areas of intense malaria
transmission (Randomized controlled) fewer — 10 fewer imprecision 1 transmission.
)
Parasite
prevalence in
600 640
per 1000 per 1000
children under
Relative risk 1.06 Low IRS may result in little to
5 years in areas (CI 95% 0.91 — 1.22) Due to serious
Difference: 40 more per no difference in parasite
of intense Based on data from 449 1000 indirectness, Due prevalence compared to
malaria participants in 1 studies. ( CI 95% 50 to serious ITNs in areas of intense
transmission (Randomized controlled) fewer — 140 imprecision 2 malaria transmission.
more )
Incidence of
malaria in all Relative risk 1.48
20 30
per 1000 per 1000 Low IRS may increase
ages in areas of (CI 95% 1.37 — 1.6)
Due to serious incidence of malaria
unstable Based on data from
Difference: 10 more per imprecision, Due compared to ITNs in
malaria 88,100 participants in 1
1000 to serious areas of unstable
studies. (Randomized
( CI 95% 10 more indirectness 3 malaria.
controlled)
— 20 more )
Parasite
prevalence in all Relative risk 1.7
2 3
per 1000 per 1000 Low IRS may result in little to
ages in areas of (CI 95% 1.18 — 2.44)
Due to serious no difference in parasite
unstable Based on data from
Difference: 1 more per 1000 indirectness, Due prevalence compared to
malaria 52,934 participants in 1
( CI 95% 0 fewer to serious ITNs in areas of
studies. (Randomized
— 3 more ) imprecision 4 unstable malaria.
controlled)
299 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Attached Images
References
70. Pluess B, Tanser FC, Lengeler C, Sharp BL : Indoor residual spraying for preventing malaria. The Cochrane
database of systematic reviews 2010;(4):CD006657 Pubmed Journal
Summary
Of the four included IRS studies, one was a cluster RCT at the village-level and three were observational studies (one
controlled before-after, one before-after and one cross-sectional). The cRCT was conducted in The Republic of the
Sudan and the three observational studies were undertaken in the Islamic Republic of Pakistan.
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements no IRS IRS (Quality of summary
evidence)
P. falciparum Relative risk 1.31 257 337 Low IRS may result in little to
prevalence (CI 95% 0.91 — 1.88) per 1000 per 1000 Due to very no difference in P.
Based on data from 278
serious falciparum prevalence
participants in 1 studies.
Difference: 80 more per imprecision. compared to no IRS.
1000
300 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements no IRS IRS (Quality of summary
evidence)
( CI 95% 23
fewer — 226
more )
Very low
57 29 Due to serious
per 1000 per 1000 risk of bias; due
to serious
P. vivax Relative risk 0.51 Difference: 28 fewer per The evidence is very
indirectness.
incidence (CI 95% 0.49 — 0.52) 1000 uncertain about the
Upgraded
Based on data from ( CI 95% 29 effect of IRS on P. vivax
because all
480,372 participants in fewer — 27 fewer incidence compared to
plausible
1 studies. ) no IRS.
confounding
would reduce the
demonstrated
effect.
Very low
78 59 Due to serious
per 1000 per 1000 inconsistency;
due to serious
Difference: 19 fewer per indirectness; due
P. vivax Odds ratio 0.74 1000 The evidence is very
to serious
prevalence (CI 95% 0.25 — 2.14) ( CI 95% 57 uncertain about the
imprecision.
Based on data from fewer — 75 more effect of IRS on P. vivax
Upgraded
4,708 participants in 2 ) prevalence compared to
because all
studies. no IRS.
plausible
confounding
would reduce
demonstrated
effect.
Attached Images
References
54. Messenger LA, Furnival-Adams J, Pelloquin B, Rowland M : Vector control for malaria prevention during
humanitarian emergencies: protocol for a systematic review and meta-analysis. BMJ Open 2021/07/
27;11(7):e046325-e046325 Pubmed Journal Website
301 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Summary
Four RCTs were included in the systematic review. Studies were conducted in the Republic of Benin, the State of
Eritrea, the Republic of the Gambia and the United Republic of Tanzania.
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Pyrethroid-like
Timeframe measurements ITNs (Quality of summary
IRS plus ITNs
evidence)
Anaemia
Odds ratio 1.04 50 50 IRS using pyrethroid-like
prevalence (CI 95% 0.83 — 1.3) per 1000 per 1000 insecticides in addition
(haemoglobin Moderate to pyrethroid ITNs
Based on data from
<8g/dl) Difference: 0 fewer per 1000 Due to serious probably has little or no
12,940 participants in 2
( CI 95% 10 imprecision 4 effect on anaemia
studies. (Randomized
prevalence compared to
controlled) fewer — 10 more
pyrethroid ITNs alone
)
1. Imprecision: serious.
302 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Attached Images
References
74. Choi L, Pryce J, Garner P : Indoor residual spraying for preventing malaria in communities using
insecticide-treated nets. The Cochrane database of systematic reviews 2019;(5):CD012688 Pubmed Journal
Website
Summary
Four studies were included in the systematic review, of which only one was an RCT; the remaining three studies
were non-randomized. Studies were undertaken in Gambia, Kenya, Sri Lanka and United Republic of Tanzania.
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No larviciding Larviciding (Quality of summary
evidence)
303 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No larviciding Larviciding (Quality of summary
evidence)
120 90
Parasite per 1000 per 1000 Larviciding may reduce
prevalence of Odds ratio 0.72 parasite prevalence in
habitats <1km2 (CI 95% 0.58 — 0.89) areas where mosquito
Difference: 30 fewer per Low
(Observational (non- 1000 aquatic habitats are less
randomized)) than 1 km² compared to
( CI 95% 50
no larviciding
fewer — 10 fewer
)
Attached Images
References
82. Choi L, Majambere S, Wilson AL : Larviciding to prevent malaria transmission. The Cochrane database of
systematic reviews 2019;(8):CD012736 Pubmed Journal Website
Summary
The systematic review identified one study from the Republic of the Philippines that investigated the impact of
habitat manipulation by controlling the release of water from spillways (overflow channels) across streams to flush
304 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
downstream areas with water against malaria. It is unknown whether larval habitat manipulation has an effect on
malaria parasite prevalence compared to no larval habitat manipulation (relative risk: 0.01; 95% CI: 0.0–0.16; one
study; very low-certainty evidence).
Comparator Certainty of
Intervention
Outcome Study results and No larval the Evidence Plain language
Larval habitat
Timeframe measurements habitat (Quality of summary
manipulation
manipulation evidence)
1. Risk of Bias: very serious. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Publication
bias: no serious.
Attached Images
Summary
The systematic review identified one study from the Republic of India that investigated the impact of habitat
manipulation by controlling the release of water using floodgates on dams in areas with IRS. It is unknown whether
larval habitat manipulation combined with IRS has an effect on malaria clinical incidence compared to IRS
alone (odds ratios or relative risks could not be calculated because the numbers of participants in each arm or at
follow-up were not reported; one study; very low-certainty evidence).
Intervention Certainty of
Outcome Study results and Comparator Larval habitat the Evidence Plain language
Timeframe measurements IRS manipulation (Quality of summary
and IRS evidence)
The study did not report the number The evidence is very
of participants in either arm. At uncertain about the
Clinical malaria Based on data from
Very low
baseline, the mean annual incidence effect of using
incidence Due to serious
participants in 1 studies. rates were 1304 cases per 1000 floodgates on a dam to
risk of bias, due
(Observational (non- children in control villages versus 786 manipulate larval
to very serious
randomized)) per 1000 children in intervention habitats on clinical
villages. Following dam construction, imprecision 1 malaria incidence
compared to no larval
305 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Larval habitat the Evidence Plain language
Timeframe measurements IRS manipulation (Quality of summary
and IRS evidence)
1. Risk of Bias: serious. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Publication bias:
no serious.
2. Risk of Bias: serious. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Publication bias:
no serious.
Attached Images
Summary
Fifteen studies were included in the systematic review. Studies were undertaken in Comoros, Ethiopia, India (three
studies), Indonesia, Kenya, Republic of Korea (two studies), Sri Lanka (two studies), Sudan, and Tajikistan (two
studies).
Treated aquatic habitats included wells, domestic water containers, fishponds and pools (seven studies); river bed
pools below dams (two studies); rice field plots (four studies); and canals (two studies).
No studies reported on clinical malaria, EIR or adult vector densities; 12 studies reported on density of immature
stages; and five studies reported on the number of aquatic habitats positive for immature stages of the vector
species.
306 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Comparator Intervention
Outcome Study results and the Evidence Plain language
no larvivorous Larvivorous
Timeframe measurements (Quality of summary
fish fish
evidence)
Clinical malaria
(incidence)
No studies
Entomological
inoculation rate
No studies
Density of adult
malaria vectors
No studies
1. Inconsistency: serious.
Attached Images
References
83. Walshe DP, Garner P, Adeel AA, Pyke GH, Burkot TR : Larvivorous fish for preventing malaria transmission. The
Cochrane database of systematic reviews 2017;(12):CD008090 Pubmed Journal Website
307 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Summary
A total of six RCTs were included in the review. Studies were conducted among residents in Plurinational State of
Bolivia, Cambodia, Lao People’s Democratic Republic and United Republic of Tanzania, and in specific populations in
Pakistan (refugees) and Thailand (pregnant women).
It is unknown whether topical repellents have an effect on clinical malaria caused by P. falciparum
(Risk ratio: 0.65; 95% CI (0.40–1.07); three studies; very low certainty evidence)
Topical repellents may or may not have a protective effect against P. falciparum parasitaemia
(Risk ratio: 0.84; 95% CI (0.64–1.12); four studies; low certainty evidence)
Topical repellents may increase the number of clinical cases caused by P. vivax
(Risk ratio: 1.32; 95% CI (0.99–1.76); two studies; low certainty evidence)
Topical repellents may or may not have a protective effect against P. vivax parasitaemia
(Risk ratio: 1.07; 95% CI (0.80–1.41); three studies; low certainty evidence)
308 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator Certainty of
Intervention
Outcome Study results and placebo or no the Evidence Plain language
Topical
Timeframe measurements topical (Quality of summary
repellent
repellent evidence)
36 48
Clinical malaria Relative risk 1.32 per 1000 per 1000 Low Topical repellents may
(P. vivax) (CI 95% 0.99 — 1.76) Due to serious increase the number of
Based on data from risk of bias, Due P. vivax clinical cases
Difference: 12 more per
3,996 participants in 2 to serious compared to no topical
1000
studies. ( CI 95% 0 more imprecision 3 repellents.
— 28 more )
Attached Images
References
77. Maia MF, Kliner M, Richardson M, Lengeler C, Moore SJ : Mosquito repellents for malaria prevention. The
Cochrane database of systematic reviews 2018;(2):CD011595 Pubmed Journal Website
309 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Summary
Two RCTs were included in the systematic review. Studies were conducted in specific populations in the Republic of
Colombia (military personnel) and the Islamic Republic of Pakistan (Afghan refugees).
Insecticide-treated clothing may have a protective effect against clinical malaria caused by P. falciparum
(Risk ratio: 0.49; 95% CI (0.29–0.83); two studies; low certainty evidence)
Insecticide-treated clothing may have a protective effect against clinical malaria caused by P. vivax
(Risk ratio: 0.64; 95% CI (0.40–1.01); two studies; low certainty evidence)
35 17 Insecticide-treating
Clinical malaria Relative risk 0.49 per 1000 per 1000 Low
clothing may reduce P.
(P. falciparum) (CI 95% 0.29 — 0.83) Due to serious
falciparum clinical
Based on data from 997 risk of bias, Due
Difference: 18 fewer per malaria compared to no
participants in 2 studies. to serious
1000 insecticide-treated
( CI 95% 25 imprecision 1 clothing.
fewer — 6 fewer )
116 74 Insecticide-treating
Clinical malaria Relative risk 0.64 per 1000 per 1000 Low
clothing may reduce P.
(P. vivax) (CI 95% 0.4 — 1.01) Due to serious
vivax clinical malaria
Based on data from 997 risk of bias, Due
Difference: 42 fewer per compared to no
participants in 2 studies. to serious
1000 insecticide-treated
( CI 95% 69 imprecision 2 clothing.
fewer — 1 more )
Attached Images
References
77. Maia MF, Kliner M, Richardson M, Lengeler C, Moore SJ : Mosquito repellents for malaria prevention. The
Cochrane database of systematic reviews 2018;(2):CD011595 Pubmed Journal Website
Summary
Two RCTs were included in the systematic review. Studies were conducted in the People’s Republic of China and the
Republic of Indonesia.
310 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
Intervention Certainty of
placebo or no
Outcome Study results and Spatial/ the Evidence Plain language
malaria
Timeframe measurements airborne (Quality of summary
prevention
repellents evidence)
intervention
Very low
Parasitaemia (all Relative risk 0.24
10 2 Due to serious
The evidence is very
per 1000 per 1000 uncertain about the
species) (CI 95% 0.03 — 1.72) risk of bias, Due
effect of spatial
Based on data from to serious
Difference: 8 fewer per 1000 repellents on malaria
6,683 participants in 2 imprecision, Due
( CI 95% 10 parasitaemia compared
studies. to serious
fewer — 8 more ) to no spatial repellents.
inconsistency 1
Attached Images
References
77. Maia MF, Kliner M, Richardson M, Lengeler C, Moore SJ : Mosquito repellents for malaria prevention. The
Cochrane database of systematic reviews 2018;(2):CD011595 Pubmed Journal Website
Summary
The review included a single interrupted time series study from the Republic of India that reported the monthly
incidence of malaria over a four-year period, with at least one year prior and at least two years post-intervention.
It is not known if space spraying causes a step change in malaria incidence (1.00, 95% CI 0.51 to 1.92, 1 study, very
low-certainty evidence).
It is not known if if space spraying causes a change in the slope of malaria incidence over time (risk ratio 0.85, 95%
CI 0.79 to 0.91, 1 study, very low-certainty evidence).
311 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Comparator
Outcome Study results and Intervention the Evidence Plain language
no space
Timeframe measurements Space spraying (Quality of summary
spraying
evidence)
Very low
Malaria cases Relative risk 1 6 6 Due to serious
The evidence is very
per month (CI 95% 0.51 — 1.92) per 1000 per 1000 uncertain about the
risk of bias, Due
(Instant effect) Based on data from effect of space spraying
to serious
participants in 1 studies. Difference: 0 more per 1000 on monthly malaria
indirectness, Due
(Observational (non- ( CI 95% 3 fewer cases compared to no
to serious
randomized)) — 6 more ) space spraying.
imprecision 1
Malaria cases
per month Relative risk 0.85
6 1 Very low The evidence is very
per 1000 per 1000 Due to serious uncertain about the
(Effect after 12 (CI 95% 0.79 — 0.91)
risk of bias, Due effect of space spraying
months Based on data from
Difference: 5 fewer per 1000 to serious on monthly malaria
follow-up) participants in 1 studies.
(Observational (non- ( CI 95% 6 fewer indirectness, Due cases after 12 months
— 4 fewer ) to serious compared to no space
randomized))
imprecision 2 spraying.
Attached Images
References
84. Pryce J, Choi L, Richardson M, Malone D : Insecticide space spraying for preventing malaria transmission. The
Cochrane database of systematic reviews 2018;(11):CD012689 Pubmed Journal Website
Summary
Two cRCTs met the inclusion criteria and were included in the meta-analysis. One trial in the Federal Democratic
Republic of Ethiopia assessed screening of windows and doors. Another trial in the Republic of the Gambia assessed
full screening (screening of eaves, doors and windows), as well as screening of ceilings only.
Screening may reduce clinical malaria incidence caused by Plasmodium falciparum (rate ratio 0.38, 95% CI 0.18 to
0.82; 1 trial, low-certainty evidence; Ethiopian study).
Screening may have a small effect on malaria parasite prevalence, (RR 0.84, 95% CI 0.60 to 1.17; 1 trial; low-
certainty evidence).
Screening probably reduces anaemia (RR 0.61, 95% CI 0.42, 0.89; 705 participants; 1 trial, moderate-certainty
evidence).
Screening may reduce the entomological inoculation rate (EIR). In the Gambian trial, there was a mean difference in
EIR between the control houses and treatment houses ranging from 0.45 to 1.50 (CIs ranged from -0.46 to 2.41;
312 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
low-certainty evidence), The Ethiopian trial reported a mean difference in EIR of 4.57, favouring screening (95% CI
3.81 to 5.33; low-certainty evidence).
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No screening Screening (Quality of summary
evidence)
Clinical malaria 91 35
Relative risk 0.38 per 1000 per 1000
incidence Low Screening of houses
(CI 95% 0.18 — 0.82)
caused by P. Due to serious may reduce clinical P.
Based on data from
falciparum Difference: 56 fewer per risk of bias, Due falciparum malaria
participants in 1 studies.
1000 to serious incidence compared to
(Randomized controlled) ( CI 95% 75 imprecision 1 no screening.
Follow up: 6 months. fewer — 21 fewer
)
Attached Images
313 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
References
85. Furnival-Adams JA, Olanga EA, Napier M, Garner M : House modifications for preventing malaria. The Cochrane
database of systematic reviews 2021;(1):CD013398 Pubmed Journal Website
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Therapeutic
Timeframe measurements No medicine (Quality of summary
course of SP
evidence)
Low
birthweight, per Relative risk 0.75 25 19 Low
(CI 95% 0.71 — 0.78) per 1000 per 1000 Upgraded due to
dose of SP (low Therapeutic courses of
Based on data from clear dose-
prevalence – 80,519 participants in response
SP may improve low
Difference: 6 fewer per 1000
2.5%) birthweight in low-
98 studies. ( CI 95% 7 fewer gradient, Due to
burden areas.
(Observational (non- — 5 fewer ) serious
9 Critical randomized)) publication bias 1
Low
Relative risk 0.75 567 425 Low
birthweight, per per 1000 per 1000
(CI 95% 0.71 — 0.78) Upgraded due to Therapeutic courses of
dose of SP (high
Based on data from clear dose- SP may result in a large
prevalence – 80,519 participants in Difference: 142 fewer per response improvement in low
56.7%) 98 studies. 1000 gradient, Due to birthweight in high-
(Observational (non- ( CI 95% 164 serious burden areas.
9 Critical randomized)) fewer — 125 publication bias 2
fewer )
314 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Therapeutic
Timeframe measurements No medicine (Quality of summary
course of SP
evidence)
Maternal
malaria Relative risk 0.8
20 16
per 1000 per 1000 Moderate
infection at (CI 95% 0.75 — 0.85) Therapeutic courses of
Upgraded due to
delivery, per Based on data from SP probably decrease
Difference: 4 fewer per 1000 clear dose-
dose of SP participants in 72 maternal malaria
( CI 95% 5 fewer response
studies. (Observational infection at delivery.
— 3 fewer ) gradient 4
(non-randomized))
7 Critical
Stillbirths and/
Relative risk 0.68 10 7 Very low We are uncertain
(CI 95% 0.59 — 0.78) per 1000 per 1000
or abortions Due to serious whether therapeutic
Based on data from 0
inconsistency, courses of SP improve
participants in 46
Difference: 3 fewer per 1000 Due to serious or worsen stillbirths
6 Important studies. (Observational ( CI 95% 4 fewer indirectness 7 and/or abortions.
(non-randomized)) — 2 fewer )
Maternal
Relative risk 1.17 3 4 Low
(CI 95% 0.49 — 2.8) per 1000 per 1000 Very few events, Therapeutic courses of
deaths Based on data from Wide CIs include SP may result in little to
8,755 participants in 6 both no effect no difference in
Difference: 1 more per 1000
5 Important studies. (Randomized and appreciable maternal deaths.
( CI 95% 2 fewer
controlled) — 5 more ) risk 8
315 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Therapeutic
Timeframe measurements No medicine (Quality of summary
course of SP
evidence)
1. Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. Publication bias: serious. Due to
participation bias (women who did not attend ANC are likely to be different from those receiving three doses of IPTp).
Upgrade: clear dose-response gradient.
2. Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. Publication bias: serious. Due to
participation bias (women who did not attend ANC are likely to be different from those receiving three doses of IPTp).
Upgrade: clear dose-response gradient.
3. Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. Publication bias: serious. Due to
participation bias (women who did not attend ANC are likely to be different from those receiving three doses of IPTp).
Upgrade: clear dose-response gradient.
4. Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious. Upgrade:
clear dose-response gradient.
5. Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious. Upgrade:
clear dose-response gradient.
6. Inconsistency: serious. The magnitude of statistical heterogeneity was high, with I-squared 77.0%.. Indirectness: no
serious. Imprecision: no serious. Publication bias: serious. Due to participation bias (women who did not attend ANC are
likely to be different from those receiving three doses of IPTp).
7. Inconsistency: serious. Small numbers. The magnitude of statistical heterogeneity was high, with I-squared 79%.
Indirectness: serious. Low numbers contributing to outcome. Distinction is not always made between these outcomes in
participating studies. Imprecision: no serious. Publication bias: no serious.
8. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Very few events, Wide CIs include
both no effect and appreciable risk. Publication bias: no serious.
9. Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious. Upgrade:
clear dose-response gradient.
10. Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious. Upgrade:
clear dose-response gradient.
11. Risk of Bias: very serious. No events reported in comparison arm. Inconsistency: no serious. Indirectness: no
serious. Imprecision: no serious. Very few events. Publication bias: no serious.
12. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. CI crosses the null. Publication bias: no
serious.
Attached Images
316 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
Certainty of
No
Outcome Study results and Intervention the Evidence Plain language
intervention,
Timeframe measurements PMC (Quality of summary
or alternative
evidence)
medicines
Clinical malaria:
Rate ratio 0.42 Difference: 3,720 fewer per
DHAP (monthly
(CI 95% 0.33 — 0.54) 1000
doses from PMC with DHAP
Based on data from 147
( CI 95% 430 Moderate
6–24 months of participants in 1 studies. Due to serious
probably reduces
age) fewer — 325 incidence of clinical
(Randomized controlled) fewer ) imprecision 5 malaria.
Follow up: 36 months of
9 Critical age.
317 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
Certainty of
No
Outcome Study results and Intervention the Evidence Plain language
intervention,
Timeframe measurements PMC (Quality of summary
or alternative
evidence)
medicines
Severe malaria
Rate ratio 0.92 Difference: 1 fewer per 1000
incidence: SP Low
(CI 95% 0.47 — 1.81)
(various dosing ( CI 95% 9 fewer Due to serious PMC with SP may
Based on data from
regimens) — 11 more ) inconsistency, reduce severe malaria
1,347 participants in 2
Due to serious incidence.
studies. (Randomized
imprecision 6
8 Critical controlled)
Severe malaria
incidence: Difference: 8 more per 1000
DHAP (monthly Rate ratio 1.29 ( CI 95% 21
fewer — 144 Low
doses from (CI 95% 0.28 — 5.98) PMC with DHAP may
more ) Due to very
6–24 months of Based on data from 147 serious
increase severe malaria
age) 7 participants in 1 studies. incidence.
imprecision 8
(Randomized controlled)
8 Critical
Anaemia
incidence: AS- Difference: 70 fewer per
Rate ratio 0.77 1000
AQ (at 10, 14
(CI 95% 0.53 — 1.12)
140 fewer — 40 Moderate PMC with AS-AQ
weeks and 9 Based on data from 684 Due to serious probably reduces
months) more
participants in 1 studies. imprecision 9 anaemia incidence.
(Randomized controlled)
7 Critical
Anaemia
incidence: Difference: 80 fewer per
Rate ratio 0.72 1000
SP+AS (at 10,
(CI 95% 0.49 — 1.07)
( CI 95% 150 Moderate PMC with SP+AS
14 weeks and 9 Based on data from 676 Due to serious probably reduces
months) fewer — 20 more
participants in 1 studies. imprecision 10 anaemia incidence.
)
(Randomized controlled)
7 Critical
Anaemia
Rate ratio 0.82 Difference: 6 fewer per 1000
incidence: SP
(CI 95% 0.68 — 0.98)
(various dosing ( CI 95% 100 Moderate PMC with SP probably
Based on data from
regimens) fewer — 10 fewer Due to serious reduces anaemia
7,438 participants in 6
) inconsistency 11 incidence.
studies. (Randomized
7 Critical controlled)
Anaemia
incidence: MQ Difference: 20 fewer per
Rate ratio 1.06 1000
(at 10, 14
(CI 95% 0.78 — 1.44)
( CI 95% 60 Moderate PMC with MQ probably
weeks and 9 Based on data from 480 Due to serious increases anaemia
months) fewer — 130
participants in 1 studies. imprecision 12 incidence.
more )
(Randomized controlled)
7 Critical
318 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
Certainty of
No
Outcome Study results and Intervention the Evidence Plain language
intervention,
Timeframe measurements PMC (Quality of summary
or alternative
evidence)
medicines
All-cause
mortality: SP
Relative risk 0.93 23 21
(CI 95% 0.74 — 1.15) per 1000 per 1000
(various dosing Moderate PMC with SP probably
Based on data from
regimens) Due to serious reduces all-cause
14,588 participants in 9 Difference: 2 fewer per 1000
inconsistency 13 mortality slightly.
studies. (Randomized ( CI 95% 6 fewer
5 Important controlled) — 3 more )
All-cause
mortality: AS-
36 44
Relative risk 1.21 per 1000 per 1000
AQ (at 10, 14
(CI 95% 0.58 — 2.55) Moderate PMC with AS-AQ
weeks and 9 Based on data from 684 Due to serious probably increases all-
Difference: 8 more per 1000
months) participants in 1 studies. ( CI 95% 15 imprecision 14 cause mortality slightly.
(Randomized controlled) fewer — 56 more
5 Important )
All-cause
mortality:
10 3
per 1000 per 1000
DHAP (monthly Relative risk 0.33
Low
doses from (CI 95% 0.01 — 8.08) PMC with DHAP may
Difference: 7 fewer per 1000 Due to very
6–24 months of Based on data from 196 ( CI 95% 10 serious
reduce all-cause
age) participants in 1 studies. mortality slightly.
fewer — 71 more imprecision 15
(Randomized controlled)
)
5 Important
All-cause
mortality:
36 30
Relative risk 0.83 per 1000 per 1000
SP+AS (at 10,
(CI 95% 0.36 — 1.89) Moderate PMC with SP+AS
14 weeks and 9 Based on data from 676 Due to serious probably reduces all-
Difference: 6 fewer per 1000
months) participants in 1 studies. ( CI 95% 23 imprecision 16 cause mortality slightly.
(Randomized controlled) fewer — 32 more
5 Important )
1. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. The overall meta-analysis was
underpowered to detect a difference or to prove equivalence. Publication bias: no serious.
2. Per 1000 person years
3. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. The overall meta-analysis was
underpowered to detect a difference or to prove equivalence. Publication bias: no serious.
4. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Small population, wide CIs around effect
estimate. Publication bias: no serious.
5. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Very few infants contributed to this
319 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Attached Images
Comparator
Certainty of
No
Outcome Study results and Intervention the Evidence Plain language
intervention or
Timeframe measurements SMC (Quality of summary
alternative
evidence)
medicines
Clinical malaria:
children <5 Difference: 315 fewer per
years (various Rate ratio 0.27 1000
SMC probably reduces
regimens) (CI 95% 0.25 — 0.29) ( CI 95% 335 Moderate
fewer — 212 clinical malaria
Per 100 person- Based on data from Due to serious
fewer ) incidence in children <5
years participants in 8 studies. inconsistency 1 years.
(Randomized controlled)
7 Critical
320 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
Certainty of
No
Outcome Study results and Intervention the Evidence Plain language
intervention or
Timeframe measurements SMC (Quality of summary
alternative
evidence)
medicines
Clinical malaria:
children <5 Rate ratio 0.28 Difference: 338 fewer per 3–4 cycles of SMC with
years, 3–4 (CI 95% 0.26 — 0.31) 1000 Moderate SP+AQ probably
cycles, SP+AQ Based on data from ( CI 95% 374 Due to serious reduces clinical malaria
participants in 4 studies. fewer — 314 inconsistency 2 incidence in children <5
(Randomized controlled) fewer ) years.
7 Critical
Clinical malaria:
children <5 Rate ratio 0.22 Difference: 205 fewer per 5–6 cycles of SMC with
years, 5–6 (CI 95% 0.18 — 0.25) 1000 Moderate SP+AQ probably
cycles, SP+AQ Based on data from ( CI 95% 233 Due to serious reduces clinical malaria
participants in 2 studies. fewer — 168 inconsistency 3 incidence in children <5
(Randomized controlled) fewer ) years.
7 Critical
Clinical malaria:
children <5 Rate ratio 0.31 Difference: 122 fewer per
1000 5–6 cycles of SMC with
years, 5–6 (CI 95% 0.26 — 0.37)
( CI 95% 146 AS-AQ reduces clinical
cycles, AS-AQ Based on data from High
fewer — 103 malaria incidence in
participants in 1 studies.
fewer ) children <5 years.
(Randomized controlled)
7 Critical
Clinical malaria:
children <5 Rate ratio 0.14 Difference: 315 fewer per
1000 3–4 cycles of SMC with
years, 3–4 (CI 95% 0.1 — 0.2)
( CI 95% 450 SP+AS reduces clinical
cycles, SP+AS Based on data from High
fewer — 225 malaria incidence in
participants in 1 studies.
fewer ) children <5 years.
(Randomized controlled)
7 Critical
Clinical malaria
incidence: Difference: 170 fewer per
Rate ratio 0.27 1000 Low
children ≥5
(CI 95% 0.25 — 0.3) Due to serious SMC may reduce clinical
years (various ( CI 95% 189
Based on data from risk of bias, Due malaria incidence in
regimens) fewer — 158
participants in 3 studies. to serious children ≥ 5 years.
fewer )
(Randomized controlled) inconsistency 4
7 Critical
Clinical malaria:
children 5–9 Rate ratio 0.39 Difference: 2 fewer per 1000 3–4 cycles of SMC with
years, 3–4 (CI 95% 0.35 — 0.44) ( CI 95% 3 fewer Moderate SP+AQ probably
cycles, SP+AQ Based on data from — 2 fewer ) Due to serious reduces clinical malaria
participants in 1 studies. risk of bias 5 incidence in children
(Randomized controlled) 5–9 years.
7 Critical
321 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
Certainty of
No
Outcome Study results and Intervention the Evidence Plain language
intervention or
Timeframe measurements SMC (Quality of summary
alternative
evidence)
medicines
fewer )
7 Critical
Clinical malaria:
children <10 Rate ratio 0.4 Difference: 53 fewer per 3–4 cycles of SMC with
years, 3–4 (CI 95% 0.35 — 0.45) 1000 Moderate SP+AQ probably
cycles, SP+AQ Based on data from ( CI 95% 60 Due to serious reduces clinical malaria
participants in 2 studies. fewer — 46 fewer risk of bias 6 incidence in children
(Randomized controlled) ) <10 years.
7 Critical
Clinical malaria:
children <10 Rate ratio 0.17 Difference: 262 fewer per 5–6 cycles of SMC with
years, 5–6 (CI 95% 0.15 — 0.2) 1000 Moderate SP+AQ probably
cycles, SP+AQ Based on data from ( CI 95% 308 Due to serious reduces clinical malaria
participants in 1 studies. fewer — 231 risk of bias 7 incidence in children
(Randomized controlled) fewer ) <10 years.
7 Critical
Clinical malaria:
children 6–15 Rate ratio 0.15 Difference: 64 fewer per 3–4 cycles of SMC with
1000 Low
years, 3–4 (CI 95% 0.11 — 0.21) AS-AQ may reduce
Due to very
cycles, AS-AQ Based on data from ( CI 95% 89 clinical malaria
serious risk of
participants in 1 studies. fewer — 47 fewer incidence in children
) bias 8 6–15 years.
(Randomized controlled)
7 Critical
Incidence of
severe malaria, Rate ratio 0.57 Difference: 14 fewer per
1000 3–4 cycles of SMC with
children <5 (CI 95% 0.37 — 0.89)
( CI 95% 22 SP+AQ decreases
years, SP+AQ, Based on data from
2,000 participants in 3 fewer — 9 fewer ) High incidence of severe
3–4 cycles malaria in children <5
studies. (Randomized
years.
controlled)
9 Critical
Incidence of
severe malaria, Rate ratio 0.44 Difference: 4 fewer per 1000
( CI 95% 8 fewer 3–4 cycles of SMC with
children 5–9 (CI 95% 0.23 — 0.84)
— 2 fewer ) Moderate SP+AQ probably
years, SP+AQ, Based on data from
Due to serious decreases incidence of
3–4 cycles 2,000 participants in 1
risk of bias 9 severe malaria in
studies. (Randomized
children 5–9 years.
controlled)
9 Critical
Incidence of
Rate ratio 0.53 Difference: 11 fewer per
severe malaria,
(CI 95% 0.37 — 0.76) 1000 SMC decreases
children <10 Based on data from
( CI 95% 16 incidence of severe
years 2,000 participants in 4 High
fewer — 8 fewer ) malaria in children <10
studies. (Randomized years.
9 Critical controlled)
322 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
Certainty of
No
Outcome Study results and Intervention the Evidence Plain language
intervention or
Timeframe measurements SMC (Quality of summary
alternative
evidence)
medicines
Incidence of all-
cause Difference: 8 more per 1000
hospitalization, ( CI 95% 4 fewer 3-4 cycles of SMC with
Rate ratio 1.38 — 86 more )
children <5 SP+AQ probably
(CI 95% 0.71 — 2.67)
years, SP+AQ Moderate increases incidence of
Based on data from
moderate Due to serious all-cause hospitalization
2,000 participants in 2
transmission, imprecision 10 in children <5 years in
studies. (Randomized
3–4 cycles moderate transmission
controlled)
settings.
8 Critical
Incidence of all-
cause Difference: 50 fewer per
hospitalization, 1000
Rate ratio 0.54 ( CI 95% 87 3-4 cycles of SMC with
children <5
(CI 95% 0.31 — 0.94) fewer — 29 fewer SP+AQ reduces
years, SP+AQ, Based on data from ) incidence of all-cause
high 2,000 participants in 1 High
hospitalization in
transmission, studies. (Randomized children <5 years in high
3–4 cycles controlled) transmission settings.
8 Critical
Incidence of all-
cause Difference: 23 fewer per
Rate ratio 0.42 1000
hospitalization, 5–6 cycles of SMC with
(CI 95% 0.2 — 0.87) ( CI 95% 48
children <5 Based on data from
AS-AQ reduces
years, AS-AQ, fewer — 11 fewer High incidence of all-cause
2,000 participants in 1 )
5–6 cycles hospitalization in
studies. (Randomized
children <5 years.
controlled)
8 Critical
All-cause
mortality, Rate ratio 0.89 Difference: 8 fewer per 1000
( CI 95% 10 Low
children <5 (CI 95% 0.68 — 1.17)
fewer — 6 fewer ) Due to serious SMC may reduce all-
years (various Based on data from
inconsistency, cause mortality in
regimens) 2,000 participants in 6
Due to serious children <5 years.
studies. (Randomized
imprecision 11
controlled)
6 Important
All-cause
mortality, Rate ratio 0.86 Difference: 7 fewer per 1000
( CI 95% 10 Low
children <5 (CI 95% 0.64 — 1.16) 3–4 cycles of SMC with
fewer — 5 fewer ) Due to serious
years, SP+AQ, Based on data from SP+AQ may reduce all-
inconsistency,
3–4 cycles 2,000 participants in 4 cause mortality in
Due to serious
studies. (Randomized children <5 years.
imprecision 12
controlled)
6 Important
323 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
Certainty of
No
Outcome Study results and Intervention the Evidence Plain language
intervention or
Timeframe measurements SMC (Quality of summary
alternative
evidence)
medicines
All-cause
mortality, Rate ratio 1.04 Difference: 18 fewer per
1000 5–6 cycles of SMC with
children <5 (CI 95% 0.39 — 2.77)
( CI 95% 7 fewer Moderate AS-AQ probably has
years, AS-AQ, Based on data from
— 9 more ) Due to serious little or no difference on
5–6 cycles 2,000 participants in 1
imprecision 14 all-cause mortality in
studies. (Randomized
children <5 years.
controlled)
6 Important
All-cause
mortality, Rate ratio 0.97 Difference: 5 fewer per 1000
( CI 95% 4 fewer Low 3–4 cycles of SMC with
children 5–9 (CI 95% 0.6 — 1.57)
— 2 more ) Due to serious SP+AQ may have little
years, SP+AQ, Based on data from
risk of bias, Due or no difference on all-
3–4 cycles 2,000 participants in 1
to serious cause mortality in
studies. (Randomized
imprecision 15 children 5–9 years.
controlled)
6 Important
All-cause
mortality, Rate ratio 1.82 Difference: 0 fewer per 1000 We are uncertain
( CI 95% 0 fewer Very low
children 5–9 (CI 95% 0.16 — 20.24) whether 5–6 cycles of
— 4 more ) Due to serious
years, SP+AQ, Based on data from SMC with SP+AQ
imprecision, Due
5–6 cycles 2,000 participants in 1 increases or decreases
to very serious
studies. (Randomized all-cause mortality in
risk of bias 16 children 5–9 years.
controlled)
6 Important
Parasite 221 84
prevalence, per 1000 per 1000
Relative risk 0.38
children <5
(CI 95% 0.34 — 0.43) Moderate SMC probably reduces
years (various Based on data from 200 Difference: 137 fewer per Due to serious parasite prevalence in
regimens) participants in 9 studies. 1000 inconsistency 17 children <5 years.
(Randomized controlled) ( CI 95% 146
4 Important fewer — 126
fewer )
Parasite 159 45
prevalence, per 1000 per 1000
Relative risk 0.28
children <5 3–4 cycles of SMC with
(CI 95% 0.24 — 0.32)
years, SP+AQ, Based on data from 200 Difference: 114 fewer per SP+AQ reduces parasite
High
3–4 cycles 1000 prevalence in children
participants in 4 studies.
<5 years.
(Randomized controlled) ( CI 95% 121
4 Important fewer — 108
fewer )
324 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
Certainty of
No
Outcome Study results and Intervention the Evidence Plain language
intervention or
Timeframe measurements SMC (Quality of summary
alternative
evidence)
medicines
Parasite 196 47
prevalence, per 1000 per 1000
Relative risk 0.24
children <5 5–6 cycles of SMC with
(CI 95% 0.16 — 0.36)
years, AS-AQ, Based on data from 200 Difference: 149 fewer per AS-AQ reduces parasite
High
5–6 cycles 1000 prevalence in children
participants in 1 studies.
<5 years.
(Randomized controlled) ( CI 95% 165
4 Important fewer — 125
fewer )
Parasite 250 57
prevalence, per 1000 per 1000
Relative risk 0.23
children 5–9 5–6 cycles of SMC with
(CI 95% 0.11 — 0.48)
years, SP+AQ, Based on data from 200 Difference: 193 fewer per SP+AQ reduces parasite
High
5–6 cycles 1000 prevalence in children
participants in 1 studies.
5–9 years.
(Randomized controlled) ( CI 95% 223
4 Important fewer — 130
fewer )
Parasite
prevalence,
84 24
Relative risk 0.28 per 1000 per 1000
children <10 SMC with SP+AQ
(CI 95% 0.17 — 0.44)
years, SP+AQ, Based on data from 200
reduces parasite
Difference: 60 fewer per High
3–6 cycles prevalence of malaria in
participants in 2 studies. 1000
children <10 years.
(Randomized controlled) 70 fewer — 47
4 Important fewer
325 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
Certainty of
No
Outcome Study results and Intervention the Evidence Plain language
intervention or
Timeframe measurements SMC (Quality of summary
alternative
evidence)
medicines
Parasite
prevalence,
19 6
Relative risk 0.29 per 1000 per 1000
children <10 3–4 cycles of SMC with
(CI 95% 0.14 — 0.61)
years, SP+AQ, Based on data from 200
SP+AQ reduces parasite
Difference: 13 fewer per High
3–4 cycles prevalence in children
participants in 1 studies. 1000
<10 years.
(Randomized controlled) ( CI 95% 16
4 Important fewer — 7 fewer )
Parasite 215 58
prevalence, per 1000 per 1000
Relative risk 0.27
children <10 5–6 cycles of SMC with
(CI 95% 0.15 — 0.48)
years, SP+AQ, Based on data from 200 Difference: 157 fewer per SP+AQ reduces parasite
High
5–6 cycles 1000 prevalence in children
participants in 1 studies.
<10 years.
(Randomized controlled) ( CI 95% 183
4 Important fewer — 112
fewer )
524 440
Any anaemia, per 1000 per 1000
Relative risk 0.84
children <5
(CI 95% 0.8 — 0.88) Moderate SMC probably reduces
years Based on data from 200 Difference: 84 fewer per Due to serious any anaemia in children
participants in 6 studies. 1000 inconsistency 21 <5 years.
2 Not Important (Randomized controlled) ( CI 95% 105
fewer — 63 fewer
)
Anaemia
prevalence: Difference: 26 fewer per
Relative risk 0.47 SMC with SP+AQ
SP+AQ 1000
(CI 95% 0.35 — 0.63) High reduces anaemia
( CI 95% 32
(Randomized controlled) prevalence.
fewer — 18 fewer
2 Not Important )
326 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
Certainty of
No
Outcome Study results and Intervention the Evidence Plain language
intervention or
Timeframe measurements SMC (Quality of summary
alternative
evidence)
medicines
Moderate
anaemia in
100 82
Relative risk 0.82 per 1000 per 1000
children <5 SMC probably reduces
(CI 95% 0.73 — 0.93) Moderate
years (various Based on data from 200 Due to serious
moderate anaemia in
Difference: 18 fewer per
regimens) children <5 years
participants in 6 studies. 1000 inconsistency 24 slightly.
(Randomized controlled) ( CI 95% 27
2 Not Important fewer — 7 fewer )
Moderate
anaemia in
48 22
per 1000 per 1000
children <5
3–4 cycles of SMC with
years, SP+AQ, Relative risk 0.47
Difference: 26 fewer per SP+AQ decreases
3–4 cycles, (CI 95% 0.35 — 0.63)
1000 moderate anaemia in
moderate to Based on data from 200 High
( CI 95% 32 children <5 years, in
high participants in 2 studies.
fewer — 18 fewer moderate to high
transmission (Randomized controlled) ) transmission areas.
2 Not Important
Moderate
anaemia in
184 171
per 1000 per 1000
children <5 Relative risk 0.93 3–4 cycles of SMC with
years, SP+AQ, (CI 95% 0.81 — 1.07) SP+AQ reduces
Difference: 13 fewer per
3–4 cycles, low Based on data from 200 1000
High moderate anaemia in
transmission participants in 2 studies. children <5 years, in low
( CI 95% 35
(Randomized controlled) transmission areas.
fewer — 13 more
2 Not Important )
327 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
Certainty of
No
Outcome Study results and Intervention the Evidence Plain language
intervention or
Timeframe measurements SMC (Quality of summary
alternative
evidence)
medicines
fewer — 31 more
)
2 Not Important
Adverse events,
children up to
114 160
per 1000 per 1000
15 years, Relative risk 1.4
various (CI 95% 1.31 — 1.51)
Difference: 46 more per SMC increases adverse
regimens, 3–4 Based on data from
1000 High events in children up to
cycles, active 18,042 participants in 4
( CI 95% 35 more 15 years.
surveillance studies. (Randomized
— 58 more )
controlled)
5 Important
1. Inconsistency: serious. The magnitude of statistical heterogeneity was high, with I-squared > 50%.. Indirectness: no
serious. Imprecision: no serious. Publication bias: no serious.
2. Inconsistency: serious. The magnitude of statistical heterogeneity was high, with I-squared > 50%.. Indirectness: no
serious. Imprecision: no serious. Publication bias: no serious.
3. Inconsistency: serious. I-squared > 50%. Indirectness: no serious. Imprecision: no serious. Publication bias: no
serious.
4. Risk of Bias: serious. Randomization was imbalanced. Inconsistency: serious. The magnitude of statistical
heterogeneity was high, with I-squared: > 50%. Indirectness: no serious. Imprecision: no serious. Publication bias: no
serious.
5. Risk of Bias: serious. Imbalanced randomization. Inconsistency: no serious. Indirectness: no serious. Imprecision: no
serious. Publication bias: no serious.
6. Risk of Bias: serious. Inadequate/lack of blinding of outcome assessors, resulting in potential for detection bias.
Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious.
7. Risk of Bias: serious. Inadequate/lack of blinding of outcome assessors, resulting in potential for detection bias.
Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious.
8. Risk of Bias: very serious. Inadequate/lack of blinding of outcome assessors, resulting in potential for detection bias.
Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious.
9. Risk of Bias: serious. Outcome evaluated by health system staff aware of study arm. Inconsistency: no serious.
Indirectness: no serious. Imprecision: no serious. Publication bias: no serious.
10. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Wide CIs. Publication bias: no serious.
11. Inconsistency: serious. I-squared > 50%. Indirectness: no serious. Imprecision: serious. Wide range of effect sizes.
Publication bias: no serious.
12. Inconsistency: serious. Wide range of effect sizes. Indirectness: no serious. Imprecision: serious. Wide CIs.
Publication bias: no serious.
13. Risk of Bias: very serious. Extra method of finding deaths in intervention arm. Selective outcome reporting.
Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Wide CIs. Publication bias: no serious.
14. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Wide CIs. Only data from one study.
Publication bias: no serious.
15. Risk of Bias: serious. Outcome evaluated by health system staff aware of study arm. Inconsistency: no serious.
Indirectness: no serious. Imprecision: serious. Wide CIs. Publication bias: no serious.
16. Risk of Bias: very serious. Extra method of finding deaths in intervention arm. Inconsistency: no serious.
Indirectness: no serious. Imprecision: serious. Wide CIs. Publication bias: no serious.
17. Inconsistency: serious. I-squared > 50%. Indirectness: no serious. Imprecision: no serious. Publication bias: no
serious.
18. Inconsistency: serious. I-squared > 50%. Indirectness: no serious. Imprecision: no serious. Publication bias: no
serious.
19. Risk of Bias: serious. High loss to follow-up, much higher in control arm. Inconsistency: no serious. Indirectness: no
328 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Attached Images
Intervention
Certainty of
Comparator Therapeutic
Outcome Study results and the Evidence Plain language
No course of an
Timeframe measurements (Quality of summary
intervention antimalarial
evidence)
medicine
279 237
Relative risk 0.85 per 1000 per 1000 Low
Anaemia (CI 95% 0.77 — 0.92)
Due to serious Therapeutic courses of
Based on data from
Difference: 42 fewer per risk of bias, Due an antimalarial medicine
14,940 participants in
8 Critical 1000 to serious may decrease anaemia.
11 studies. (Randomized ( CI 95% 64 inconsistency 2
controlled) fewer — 22 fewer
)
329 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention
Certainty of
Comparator Therapeutic
Outcome Study results and the Evidence Plain language
No course of an
Timeframe measurements (Quality of summary
intervention antimalarial
evidence)
medicine
( CI 95% 209
fewer — 164
fewer )
1. Risk of Bias: serious. Participants and personnel giving the treatments were not blinded. Inconsistency: serious.
Unexplained between-study heterogeneity . Indirectness: no serious. Imprecision: no serious. Publication bias: no
serious.
2. Risk of Bias: serious. Participants and personnel giving the treatments were not blinded. Inconsistency: serious.
Unexplained between-study heterogeneity . Indirectness: no serious. Imprecision: no serious. Publication bias: no
serious.
3. Risk of Bias: serious. Participants and personnel giving the treatments were not blinded. Inconsistency: serious.
Unexplained between-study heterogeneity . Indirectness: no serious. Imprecision: no serious. Publication bias: no
serious.
Attached Images
Intervention
Certainty of
Comparator Therapeutic
Outcome Study results and the Evidence Plain language
Placebo or no courses of an
Timeframe measurements (Quality of summary
intervention antimalarial
evidence)
medicine
330 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention
Certainty of
Comparator Therapeutic
Outcome Study results and the Evidence Plain language
Placebo or no courses of an
Timeframe measurements (Quality of summary
intervention antimalarial
evidence)
medicine
All-cause
mortality Relative risk 0.77 21 16 Therapeutic courses of
per 1000 per 1000 an antimalarial medicine
(CI 95% 0.47 — 1.28)
(intervention probably reduce all-
Based on data from
plus post- Moderate cause mortality.
3,387 participants in 3 Difference: 5 fewer per 1000
intervention Due to serious However, the effect
studies. (Randomized ( CI 95% 11
period) imprecision 2 varies and it is possible
controlled) fewer — 6 more )
that it makes little to no
Follow up: 2 weeks to
difference for all-cause
8 Critical 26 weeks.
mortality.
All-cause re-
Hazard ratio 1.04 632 646 Therapeutic courses of
admission per 1000 per 1000
(CI 95% 0.83 — 1.3) an antimalarial medicine
(post-
Based on data from 558 Moderate probably result in little
intervention participants in 2 studies. Difference: 14 more per Due to serious to no difference in all-
period) (Randomized controlled) 1000 imprecision 4 cause re-admission in
Follow up: 15 weeks to ( CI 95% 68 the post-intervention
9 Critical 26 weeks. fewer — 95 more period.
)
Severe anaemia
Hazard ratio 0.74 289 223
re-admission per 1000 per 1000 Therapeutic courses of
(CI 95% 0.52 — 1.05)
(post- an antimalarial medicine
Based on data from 558 Moderate
intervention participants in 2 studies. Difference: 66 fewer per Due to serious
probably decrease
period) 1000 severe anaemia re-
(Randomized controlled) imprecision 6 admission in the post-
Follow up: 15 weeks to ( CI 95% 126
intervention period.
8 Critical 26 weeks. fewer — 12 more
)
331 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention
Certainty of
Comparator Therapeutic
Outcome Study results and the Evidence Plain language
Placebo or no courses of an
Timeframe measurements (Quality of summary
intervention antimalarial
evidence)
medicine
fewer )
Severe malaria
Hazard ratio 1.06 368 385 Therapeutic courses of
re-admission per 1000 per 1000
(CI 95% 0.81 — 1.39) an antimalarial medicine
(post-
Based on data from 558 Moderate probably result in little
intervention participants in 2 studies. Difference: 17 more per Due to serious to no difference in
period) (Randomized controlled) 1000 imprecision 8 severe malaria re-
Follow up: 15 weeks to ( CI 95% 58 admission in the post-
8 Critical 26 weeks. fewer — 104 intervention period.
more )
332 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention
Certainty of
Comparator Therapeutic
Outcome Study results and the Evidence Plain language
Placebo or no courses of an
Timeframe measurements (Quality of summary
intervention antimalarial
evidence)
medicine
1. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. The range of effect includes the null.
Publication bias: no serious.
2. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. The range of effect includes the null.
Publication bias: no serious.
3. Inconsistency: serious. Considerable heterogeneity with I-squared = 87%. Indirectness: no serious. Imprecision: no
serious. Publication bias: no serious.
4. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. The range of effect includes the null.
Publication bias: no serious.
5. Inconsistency: serious. Substantial heterogeneity with I-squared = 69%. Indirectness: no serious. Imprecision: no
serious. Publication bias: no serious.
6. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. The range of effect includes the null.
Publication bias: no serious.
7. Inconsistency: serious. Considerable heterogeneity with I-squared = 93%. Indirectness: no serious. Imprecision: no
serious. Publication bias: no serious.
8. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. The range of effect includes the null.
Publication bias: no serious.
9. Inconsistency: serious. Substantial heterogeneity with I-squared = 71%. Indirectness: no serious. Imprecision: no
serious. Publication bias: no serious.
10. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. The range of effect includes the null.
Publication bias: no serious.
11. Inconsistency: serious. Substantial heterogeneity with I-squared = 71%. Indirectness: no serious. Imprecision: no
serious. Publication bias: no serious.
12. Inconsistency: no serious. Indirectness: serious. ECG monitoring was conducted in a nested cardiac monitoring
study involving 33 children receiving DHAP (one study). Imprecision: no serious. Publication bias: no serious.
Attached Images
333 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No MDA MDA (Quality of summary
evidence)
Clinical malaria
incidence: RCT, Difference: 1 fewer per MDA may decrease
Rate ratio 0.41 1000
Pf, mod/high (CI 95% 0.04 — 4.42) Low
clinical malaria
transmission ( CI 95% 2 fewer incidence in delimited
Based on data from Due to very
1–3 months — 8 more ) moderate to high
144,422 participants in serious
post-MDA malaria transmission
1 studies. (Randomized imprecision 1 areas 1–3 months
controlled)
post-MDA.
9 Critical
Clinical malaria
incidence: RCT, Difference: 20 fewer per
Pf, low/very Rate ratio 0.58 1000 MDA may decrease
low (CI 95% 0.12 — 2.73) ( CI 95% 50 Low clinical malaria
transmission Based on data from fewer — 90 more Due to very incidence in delimited
1–3 months 130,651 participants in ) serious low to very low malaria
post-MDA 2 studies. (Randomized imprecision 2 transmission areas 1–3
controlled) months post-MDA.
9 Critical
Clinical malaria
incidence: RCT, Difference: 5 fewer per We are uncertain
1000 whether MDA
Pf, low/very Rate ratio 0.47
( CI 95% 7 fewer Very low increases or decreases
low (CI 95% 0.21 — 1.03)
— 0 fewer ) Due to serious clinical malaria
transmission Based on data from
risk of bias, Due incidence in delimited
4–12 months 26,576 participants in
to serious low to very low
post-MDA 3 studies. (Randomized
imprecision 3 transmission areas
controlled)
4–12 months post-
9 Critical MDA.
Clinical malaria
incidence: RCT, Difference: 4 fewer per
1000 MDA may decrease
Pf, low/very Rate ratio 0.77
( CI 95% 14 clinical malaria
low (CI 95% 0.2 — 3.03) Low
fewer — 34 more incidence in delimited
transmission Based on data from Due to very
) low to very low malaria
12–24 months 23,251 participants in serious
transmission areas
post-MDA 1 studies. (Randomized imprecision 4 12–24 months post-
controlled)
MDA.
9 Critical
Clinical malaria
Difference: 16 fewer per
Very low
incidence: RCT, Rate ratio 1.38 We are uncertain
1000 Due to serious
Pv (CI 95% 0.97 — 1.95) whether MDA
risk of bias, Due
4–12 months Based on data from ( CI 95% 39 increases or decreases
to serious
post-MDA 3,325 participants in 2 fewer — 1 fewer P. vivax clinical malaria
inconsistency,
studies. (Randomized ) incidence 4–12 months
Due to serious
controlled) post-MDA.
9 Critical imprecision 5
334 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No MDA MDA (Quality of summary
evidence)
Clinical malaria
Rate ratio 0.29
incidence: non- Difference: 45 fewer per We are uncertain
(CI 95% 0.26 — 0.31)
RCT, Pv 1000 whether MDA
Based on data from Very low
1–3 months ( CI 95% 48 increases or decreases
62,744 participants in Due to serious
post-MDA fewer — 41 P. vivax clinical malaria
2 studies. inconsistency 7
fewer ) incidence 1–3 months
(Observational (non-
post-MDA.
9 Critical randomized))
Clinical malaria
Rate ratio 0.72
incidence: non- Difference: 44 fewer per We are uncertain
(CI 95% 0.68 — 0.76)
RCT, Pv 1000 Very low whether MDA
Based on data from
4–12 months ( CI 95% 50 Due to very increases or decreases
11,300 participants in
post-MDA fewer — 37 serious risk of P. vivax clinical malaria
1 studies.
fewer ) bias 8 incidence 4–12 months
(Observational (non-
post-MDA.
9 Critical randomized))
Clinical malaria
Rate ratio 0.04
incidence: non- Difference: 150 fewer per We are uncertain
(CI 95% 0.02 — 0.07)
RCT, Pv 1000 Very low whether MDA
Based on data from
12–24 months ( CI 95% 153 Due to very increases or decreases
11,300 participants in
post-MDA fewer — 145 serious risk of P. vivax clinical malaria
1 studies.
fewer ) bias 9 incidence 12–24
(Observational (non-
months post-MDA.
9 Critical randomized))
All-cause
mortality: all
81 55
Relative risk 0.68 per 1 million per 1 million
ages, non-RCT, We are uncertain
(CI 95% 0.57 — 0.81)
Pf, mod/high Based on data from Very low
whether MDA
Difference: 26 fewer per 1
transmission increases or decreases
7,541,000 participants million Due to serious
<1 month post- all-cause mortality in
in 1 studies. ( CI 95% 35 risk of bias 10
MDA all ages <1 month post-
(Observational (non- fewer — 15 MDA.
randomized)) fewer )
7 Critical
All-cause
mortality: <5
250 85
per 1 million per 1 million
years, non- Relative risk 0.34
We are uncertain
RCT, Pf, mod/ (CI 95% 0.25 — 0.47)
Difference: 165 fewer per 1 whether MDA
high Based on data from Very low
million increases or decreases
transmission 1,353,070 participants Due to serious
( CI 95% 187 all-cause mortality in
<1 month post- in 1 studies. risk of bias 11
fewer — 132 children <5 years <1
MDA (Observational (non-
fewer ) month post-MDA.
randomized))
7 Critical
335 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No MDA MDA (Quality of summary
evidence)
All-cause
mortality: all
51 87
Odds ratio 1.77 per 1 million per 1 million
ages, non-RCT, We are uncertain
(CI 95% 1.54 — 2.04)
Pf, mod/high Based on data from Very low
whether MDA
Difference: 36 more per 1
transmission increases or decreases
11,419,200 million Due to serious
1–3 months all-cause mortality in
participants in 1 ( CI 95% 25 risk of bias 12
post-MDA all ages 1–3 months
studies. (Observational more — 48 more post-MDA.
(non-randomized)) )
7 Critical
All-cause
mortality: <5
106 118
per 1 million per 1 million
years, non- Odds ratio 1.13
We are uncertain
RCT, Pf, mod/ (CI 95% 0.87 — 1.46) Very low
Difference: 12 more per 1 whether MDA
high Based on data from Due to serious
million increases or decreases
transmission 2,008,720 participants risk of bias, Due
( CI 95% 12 all-cause mortality in
1–3 months in 1 studies. to serious
fewer — 42 more children <5 years 1–3
post-MDA (Observational (non- imprecision 13 months post-MDA.
)
randomized))
7 Critical
Parasite
prevalence:
418 251
We are uncertain
per 1000 per 1000
non-RCT, Pf, Relative risk 0.6 whether MDA
mod/high (CI 95% 0.55 — 0.67) increases or decreases
Difference: 167 fewer per Very low
transmission Based on data from P. falciparum parasite
1000 Due to serious
4–12 months 3,154 participants in 1 prevalence in moderate
( CI 95% 188 risk of bias 14 to high transmission
post-MDA studies. (Observational
fewer — 138 areas 4–12 months
(non-randomized))
fewer ) post-MDA.
6 Important
Parasite
incidence: RCT,
57 35
Rate ratio 0.61 per 1000 per 1000 MDA probably reduces
Pf, mod/high (CI 95% 0.4 — 0.92) the incidence of P.
transmission Moderate
Based on data from Difference: 22 fewer per falciparum in moderate
1–3 months Due to serious
820 participants in 1 1000 to high transmission
post-MDA risk of bias 15 areas 1–3 months
studies. (Randomized ( CI 95% 20
controlled) fewer — 205 post-MDA.
3 Not Important more )
Parasite
incidence: RCT,
108 98 We are uncertain
Rate ratio 0.91 per 1000 per 1000 whether MDA
Pf, mod/high Very low
(CI 95% 0.55 — 1.5) increases or decreases
transmission Due to serious
Based on data from Difference: 10 fewer per the incidence of P.
4–12 months risk of bias, Due
518 participants in 1 1000 falciparum in moderate
post-MDA to serious
studies. (Randomized ( CI 95% 53 to high transmission
imprecision 16 areas 4–12 months
controlled) fewer — 270
3 Not Important more ) post-MDA.
Parasite Moderate
incidence: RCT,
Rate ratio 0.37
(CI 95% 0.21 — 0.66)
12 5 Due to serious
MDA probably reduces
the incidence of P.
336 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No MDA MDA (Quality of summary
evidence)
Parasite
prevalence:
47 83
per 1000 per 1000
RCT, Pf, mod/ Relative risk 1.76 MDA may increase P.
high (CI 95% 0.58 — 5.36) Low falciparum parasite
Difference: 36 more per
transmission Based on data from Due to very prevalence slightly in
1000
1–3 months 786 participants in 1 serious moderate to high
( CI 95% 20
post-MDA studies. (Randomized imprecision 18 transmission areas 1–3
fewer — 205 months post-MDA.
controlled)
more )
6 Important
Parasite
prevalence:
483 570
per 1000 per 1000 MDA may increase P.
RCT, Pf, mod/ Relative risk 1.18
falciparum parasite
high (CI 95% 0.89 — 1.56) Low
Difference: 87 more per prevalence slightly in
transmission Based on data from Due to very
1000 moderate to high
4–12 months 1,497 participants in 1 serious
( CI 95% 53 transmission areas
post-MDA studies. (Randomized imprecision 19 4–12 months post-
fewer — 270
controlled)
more ) MDA.
6 Important
Parasite
prevalence:
723 615
We are uncertain
per 1000 per 1000
non-RCT, Pf, Relative risk 0.85 whether MDA
mod/high (CI 95% 0.78 — 0.93) increases or decreases
Difference: 108 fewer per Very low
transmission Based on data from P. falciparum parasite
1000 Due to serious
1–3 months 1,000 participants in 1 prevalence in moderate
( CI 95% 159 risk of bias 20 to high transmission
post-MDA studies. (Observational
fewer — 51 areas 1–3 months
(non-randomized))
fewer ) post-MDA.
6 Important
Parasite
prevalence:
431 332
per 1000 per 1000
non-RCT, Pf, Relative risk 0.77 MDA may decrease P.
mod/high (CI 95% 0.7 — 0.84) falciparum parasite
Difference: 99 fewer per
transmission Based on data from prevalence in moderate
3,261 participants in 1
1000 Low
12–24 months to high transmission
( CI 95% 129 areas 12–24 months
post-MDA studies. (Observational
fewer — 69 post-MDA.
(non-randomized))
fewer )
6 Important
Parasite
prevalence:
Relative risk 0.34
(CI 95% 0.06 — 1.97)
32 11 Very low
Due to serious
We are uncertain
whether MDA
Based on data from per 1000 per 1000 increases or decreases
RCT, Pf, low/ risk of bias, Due
337 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No MDA MDA (Quality of summary
evidence)
very low
transmission Difference: 21 fewer per P. falciparum parasite
12–24 months 1,390 participants in 1 1000 prevalence in low to
to serious
post-MDA studies. (Randomized ( CI 95% 30 very low transmission
fewer — 31 more indirectness 21
controlled) areas 12–24 months
) post-MDA.
6 Important
Parasite
prevalence:
35 4
per 1000 per 1000
RCT, Pf, low/ Relative risk 0.12 MDA probably
very low (CI 95% 0.03 — 0.52) decreases P. falciparum
Difference: 31 fewer per Moderate
transmission Based on data from parasite prevalence in
1000 Due to serious
<1 month post- 718 participants in 2 low to very low
( CI 95% 34 risk of bias 22 transmission areas <1
MDA studies. (Randomized
fewer — 17 month post-MDA.
controlled)
fewer )
6 Important
Parasite
prevalence:
24 6
per 1000 per 1000
RCT, Pf, low/ Relative risk 0.25 MDA probably
very low (CI 95% 0.15 — 0.41) decreases P. falciparum
Difference: 18 fewer per Moderate
transmission Based on data from parasite prevalence in
1000 Due to serious
1–3 months 6,511 participants in 8 low to very low
( CI 95% 20 risk of bias 23 transmission areas 1–3
post-MDA studies. (Randomized
fewer — 14 months post-MDA.
controlled)
fewer )
6 Important
Parasite 272 49
prevalence: Relative risk 0.18 per 1000 per 1000
RCT, Pv (CI 95% 0.08 — 0.4) MDA probably
Moderate
<1 month post- Based on data from decreases P. vivax
Difference: 223 fewer per Due to serious
MDA 234 participants in 1 parasite prevalence <1
1000 risk of bias 24
studies. (Randomized month post-MDA.
( CI 95% 250
controlled) fewer — 163
6 Important
fewer )
Parasite
prevalence:
19 16
per 1000 per 1000
RCT, Pf, low/ Relative risk 0.82 MDA may decrease P.
Low
very low (CI 95% 0.56 — 1.22) falciparum parasite
Difference: 3 fewer per Due to serious
transmission Based on data from prevalence in low to
1000 risk of bias, Due
4–12 months 5,102 participants in 6 very low transmission
( CI 95% 8 fewer to serious
post-MDA studies. (Randomized areas 4–12 months
— 4 more ) inconsistency 25 post-MDA.
controlled)
6 Important
338 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No MDA MDA (Quality of summary
evidence)
( CI 95% 119
fewer — 101
fewer )
Parasite
prevalence: Relative risk 1.01
96 97
per 1000 per 1000 Low MDA may have little or
RCT, Pv (CI 95% 0.87 — 1.18)
Due to serious no effect on P. vivax
4-12 months Based on data from
Difference: 1 more per 1000 risk of bias, Due parasite prevalence
post-MDA 6,255 participants in 5
( CI 95% 12 to serious 4–12 months post-
studies. (Randomized
fewer — 17 more inconsistency 27 MDA.
controlled)
6 Important )
Parasite 71 23
prevalence: Relative risk 0.32 per 1000 per 1000 We are uncertain
non-RCT, Pv (CI 95% 0.12 — 0.87) whether MDA
Very low
<1 month post- Based on data from increases or decreases
Difference: 48 fewer per Due to serious
MDA 449 participants in 1 P. vivax parasite
1000 risk of bias 29
studies. (Observational prevalence <1 month
( CI 95% 62
(non-randomized)) post-MDA.
6 Important fewer — 9 fewer
)
Parasite 231 42
prevalence: Relative risk 0.18 per 1000 per 1000 We are uncertain
non-RCT, Pv (CI 95% 0.1 — 0.33) Very low whether MDA
1–3 months Based on data from Due to very increases or decreases
Difference: 189 fewer per
post-MDA 1,024 participants in 2 serious risk of P. vivax parasite
1000
studies. (Observational ( CI 95% 208 bias 30 prevalence 1–3 months
(non-randomized)) post-MDA.
6 Important fewer — 155
fewer )
Parasite 71 24
prevalence: Relative risk 0.34 per 1000 per 1000 We are uncertain
non-RCT, Pv (CI 95% 0.15 — 0.78) Very low whether MDA
4–12 months Based on data from Due to very increases or decreases
Difference: 47 fewer per
post-MDA 939 participants in 1 serious risk of P. vivax parasite
1000
studies. (Observational ( CI 95% 60 bias 31 prevalence 4–12
(non-randomized)) months post-MDA.
6 Important fewer — 16
fewer )
339 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No MDA MDA (Quality of summary
evidence)
transmission million
0–3 months ( CI 95% 173
post-MDA studies. (Randomized fewer — 564
controlled) more )
5 Important
Serious
adverse events:
3,466 1,938
per 1 million per 1 million
RCT, Pf, low/ Odds ratio 1.47
very low (CI 95% 0.68 — 3.2) MDA probably
Difference: 1,528 fewer per Moderate
transmission Based on data from increases serious
1 million Due to serious
4–12 months 6,911 participants in 1 adverse events 4–12
( CI 95% 25,065 imprecision 33 months post-MDA.
post-MDA studies. (Randomized
fewer — 2,180
controlled)
fewer )
5 Important
Adverse
events: RCT, Pf,
133 333
Odds ratio 3.25 per 1000 per 1000
mod/high (CI 95% 0.68 — 15.53)
We are uncertain
transmission whether MDA
Based on data from 90 Difference: 200 more per Very low
1–3 months increases or decreases
participants in 1 1000 34
post-MDA adverse events 1–3
studies. (Randomized ( CI 95% 39 months post-MDA.
controlled) fewer — 571
5 Important more )
Adverse event
(vomiting):
43 24
Odds ratio 0.54 per 1000 per 1000
SP+AS +/-PQ,
(CI 95% 0.19 — 1.54)
RCT, Pf, low/ Moderate MDA with SP+AS +/-
Based on data from Difference: 19 fewer per
very low Due to serious PQ probably increases
703 participants in 1 1000
transmission imprecision 35 vomiting.
studies. (Randomized ( CI 95% 35
controlled) fewer — 22 more
4 Important )
340 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No MDA MDA (Quality of summary
evidence)
1. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Wide CIs that include both no
effect and substantial effect . Publication bias: no serious.
2. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Wide CIs that include both no
effect and substantial effect . Publication bias: no serious.
3. Risk of Bias: serious. High or unclear risk of bias in some/ all included studies. Inconsistency: very serious. I-
squared > 50%. Indirectness: no serious. Imprecision: serious. Wide CIs that include both no effect and substantial
341 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
342 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Attached Images
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No MDA MDA (Quality of summary
evidence)
All-cause
Relative risk 0.68 81 55
mortality: 3 per 1 million per 1 million
(CI 95% 0.57 — 0.81) We are uncertain about
rounds, <1
Based on data from Very low the effect of MDA on
month post- 7,541,000 participants Difference: 26 fewer per 1 Due to serious all-cause mortality in
MDA, all ages 1 in 1 studies. million risk of bias 2 all ages <1 month post-
(Observational (non- ( CI 95% 35 MDA.
7 Critical randomized)) fewer — 15
fewer )
343 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No MDA MDA (Quality of summary
evidence)
3 ( CI 95% 187
fewer — 132
randomized)) fewer )
7 Critical
All-cause
Odds ratio 1.77 51 87
mortality: 3 per 1 million per 1 million
(CI 95% 1.54 — 2.04) Very low We are uncertain about
rounds, 1–3
Based on data from Due to serious the effect of MDA on
months post- 11,419,200 Difference: 36 more per 1 risk of bias, Due all-cause mortality in
MDA, all ages 5 participants in 1 million to serious all ages 1–3 months
studies. (Observational ( CI 95% 25 imprecision 6 post-MDA.
7 Critical (non-randomized)) more — 48 more
)
All-cause
mortality: 3
106 118
Odds ratio 1.13
per 1 million per 1 million
rounds, 1–3 (CI 95% 0.87 — 1.46) Very low We are uncertain about
months post- Based on data from Due to serious the effect of MDA on
Difference: 12 more per 1
MDA, <5 years 2,008,720 participants risk of bias and all-cause mortality in
million
7 in 1 studies. serious children <5 years 1–3
( CI 95% 12
(Observational (non- imprecision 8 months post-MDA.
fewer — 42 more
randomized))
7 Critical )
Parasitologicall Based on data from Point estimates of changes in Very low We are uncertain about
y confirmed participants in 1 parasitologically-confirmed malaria Due to serious the effect of MDA on
malaria 0–1 studies. (Observational cases at health facilities decreased risk of bias 11 parasitologically
344 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements No MDA MDA (Quality of summary
evidence)
Attached Images
345 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
falciparum
Intervention: Mass drug administration (MDA)
Comparator: no MDA
Summary
The systematic review identified eight cRCTs in very low to low transmission settings of six countries (Cambodia,
Lao People’s Democratic Republic, Myanmar, United Republic of Tanzania, Viet Nam and Zambia) assessing the
impact of MDA on P. falciparum prevalence or incidence compared to no MDA (Schneider et al unpublished
evidence). Two studies used DP alone; five studies used DP plus single low-dose primaquine; and one study used
sulfadoxine-pyrimethamine/artesunate (SP+AS) plus a single dose of primaquine at 0.75 mg/kg. Most (5) studies
conducted three rounds of MDA within one year; one study conducted four rounds of MDA over 15 months;
one study conducted two rounds and one study conducted one round of MDA over a one-year period.
Meta-analyses of the results showed reductions in prevalence and incidence of P. falciparum infection, but not
clinical disease, 1–3 months after the last round of MDA. Multiple studies evaluated these outcomes at longer
time periods but either no impact was found or the evidence was of very low certainty. Adverse events were
often not measured in both arms, which complicated interpretation of the findings, but reported rates of adverse
events or serious adverse events were low. Markers of artemisinin resistance were measured in only one study,
which found no evidence of increases in drug-resistant parasites.
Intervention Certainty of
Outcome Study results and Comparator Mass drug the Evidence Plain language
Timeframe measurements no MDA administration (Quality of summary
(MDA) evidence)
1-3 months -
Incidence of
Rate ratio 0.58
(CI 95% 0.12 — 2.73)
6 4 Low
MDA may result in
little to no difference in
Based on data from per 1000 per 1000 Due to very the incidence of P.
clinical malaria
130,651 participants in serious falciparum clinical
2 studies. (Randomized imprecision 1 malaria between 1-3
controlled) months
24 6
Relative risk 0.25 per 1000 per 1000
1-3 months - (CI 95% 0.15 — 0.41) MDA probably reduces
Prevalence Moderate
Based on data from P. falciparum
Difference: 18 fewer per Due to serious
6,511 participants in 8 prevalence between
1000 risk of bias 2
studies. (Randomized 1-3 months
( CI 95% 20
controlled) fewer — 14
fewer )
1-3 months -
Incidence of
Rate ratio 0.37
(CI 95% 0.21 — 0.66)
12 5 MDA probably reduces
per 1000 per 1000 Moderate the incidence of P.
parasitaemia Based on data from
Due to serious falciparum
811 participants in 1
risk of bias 4 parasitaemia between
studies. (Randomized
1-3 months
controlled)
346 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Mass drug the Evidence Plain language
Timeframe measurements no MDA administration (Quality of summary
(MDA) evidence)
133 433
1-3 months - Relative risk 3.25 per 1000 per 1000 The evidence is very
Adverse events (CI 95% 0.68 — 15.53)
Very low uncertain about the
Based on data from 90
Difference: 300 more per Due to serious effect of MDA on
participants in 1
1000 indirectness 6 adverse events
studies. (Randomized ( CI 95% 43 between 1-3 months
controlled) fewer — 1,000
more )
12-24 months -
Incidence of
Rate ratio 0.77
(CI 95% 0.2 — 3.03)
17 13 Low
MDA may reduce the
per 1000 per 1000 incidence of P.
clinical malaria Based on data from Due to very
falciparum clinical
23,251 participants in serious
malaria between 12-24
1 studies. (Randomized imprecision 8 months
controlled)
0-3 months -
Serious
Odds ratio 3.61 0 1 MDA probably results
(CI 95% 0.43 — 30.03) per 1000 per 1000
Adverse Events Moderate in little to no difference
Based on data from
Due to serious in serious adverse
6,911 participants in 1 Difference: 1 more per 1000
imprecision 10 events between 0-3
studies. (Randomized ( CI 95% 0 more months
controlled) — 11 more )
Pf - Vomiting
among people
Odds ratio 0.54
(CI 95% 0.19 — 1.54)
43 24 MDA probably does
not increase vomiting
per 1000 per 1000 Moderate
receiving Based on data from among people
Due to serious
SP+AS with or 703 participants in 1 receiving SP+AS with
Difference: 19 fewer per imprecision 11 or without PQ vs
without PQ vs studies. (Randomized
1000
Placebo - Low/ controlled) Placebo
347 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Mass drug the Evidence Plain language
Timeframe measurements no MDA administration (Quality of summary
(MDA) evidence)
SAEs among
people who
0.03
Based on data from per 1000
received MDA 12
353,143 participants in
4 studies. (Randomized
controlled)
1-3 months -
Drug resistance
608 498
per 1000 per 1000 The evidence is very
markers Relative risk 0.82 uncertain about the
(PfKelch13) (CI 95% 0.45 — 1.51) effect of MDA on
Difference: 109 fewer per Very low
among people Based on data from 63 1000 Due to serious
artemisinin resistance
who were Pf participants in 1 markers (PfKelch13)
( CI 95% 334 risk of bias 13 among P. falciparum
positive studies. (Randomized
fewer — 310
controlled) infections between 1-3
more )
months
1-3 months -
Drug resistance
64 8
Relative risk 0.13 per 1000 per 1000 MDA may reduce the
markers Low
(CI 95% 0.05 — 0.3) proportion of drug
(PfKelch13) Due to serious
Based on data from Difference: 56 fewer per resistance markers
among all risk of bias, and
1,232 participants in 1 1000 (PfKelch13) among all
samples serious
studies. (Randomized ( CI 95% 61 samples between 1-3
imprecision 14 months
controlled) fewer — 45
fewer )
4-12 months -
Drug resistance
610 707
per 1000 per 1000 The evidence is very
markers Relative risk 1.16 uncertain about the
(PfKelch13) (CI 95% 0.83 — 1.61) effect of MDA on
Difference: 98 more per Very low
among people Based on data from 75 1000 Due to serious
artemisinin resistance
who were Pf participants in 1 markers (PfKelch13)
( CI 95% 104 risk of bias 15 among P. falciparum
positive studies. (Randomized
fewer — 372
controlled) infections between
more )
4-12 months
4-12 months -
Drug resistance
29 14
Relative risk 0.49 per 1000 per 1000 MDA may reduce the
markers Low
(CI 95% 0.28 — 0.85) proportion of drug
(PfKelch13) Due to serious
Based on data from Difference: 15 fewer per resistance markers
among all risk of bias, and
2,595 participants in 1 1000 (PfKelch13) among all
samples serious
studies. (Randomized ( CI 95% 21 samples between 4-12
imprecision 16 months
controlled) fewer — 4 fewer
)
348 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Mass drug the Evidence Plain language
Timeframe measurements no MDA administration (Quality of summary
(MDA) evidence)
12-24 months -
Drug resistance
714 764
per 1000 per 1000 The evidence is very
markers Relative risk 1.07 uncertain about the
(PfKelch13) (CI 95% 0.82 — 1.4) effect of MDA on
Difference: 50 more per Very low
among people Based on data from 78 1000 Due to serious
artemisinin resistance
who were Pf participants in 1 markers (PfKelch13)
( CI 95% 129 risk of bias 17 among P. falciparum
positive studies. (Randomized
fewer — 286
controlled) infections between
more )
12-24 months
12-24 months -
Drug resistance
25 17 MDA may result in
Relative risk 0.66 per 1000 per 1000
markers Low little to no reduction in
(CI 95% 0.4 — 1.11)
(PfKelch13) Due to serious drug resistance
Based on data from Difference: 8 fewer per
among all risk of bias, and markers (PfKelch13)
2,990 participants in 1 1000
samples serious among all samples
studies. (Randomized ( CI 95% 15
imprecision 18 between 12-24
controlled) fewer — 3 more ) months
1. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Substantial variability in point
estimates including both appreciable risk and appreciable benefit.. Publication bias: no serious.
2. Risk of Bias: serious. Some risk of bias in most/all included studies. Inconsistency: no serious. Indirectness: no
serious. Imprecision: no serious. Publication bias: no serious.
3. Risk of Bias: serious. Some risk of bias in most/all included studies. Inconsistency: serious. Completely non-
overlapping confidence intervals. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious.
4. Risk of Bias: serious. Some risk of bias in most/all included studies. Inconsistency: no serious. Indirectness: no
serious. Imprecision: no serious. Publication bias: no serious.
5. Risk of Bias: serious. Some risk of bias in most/all included studies. Inconsistency: very serious. I-squared 72%.
Indirectness: no serious. Imprecision: serious. Wide confidence intervals including both no effect and appreciable
benefit/ risk. Publication bias: no serious.
6. Inconsistency: no serious. Indirectness: serious. Self-reported symptoms, serious indirectness. Imprecision: very
serious. Wide confidence intervals including both no effect and appreciable benefit/ risk. Publication bias: no
serious.
7. Risk of Bias: serious. High risk of bias in all included studies. Inconsistency: no serious. Indirectness: serious.
McLean had contact tracing for neighbors in 50 km surrounding positive cases in the intervention but not control
arm; this effect measures the combined intervention.. Imprecision: very serious. Wide confidence intervals including
both no effect and appreciable benefit/ risk. Publication bias: no serious.
8. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Wide confidence intervals
including both no effect and appreciable benefit/ risk. Publication bias: no serious.
9. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Wide confidence intervals
including both no effect and appreciable benefit/ risk. Publication bias: no serious.
10. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Wide confidence intervals including
both no effect and appreciable benefit/ risk. Publication bias: no serious.
11. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Wide confidence intervals including
both no effect and appreciable benefit/ risk. Publication bias: no serious.
12. Risk of Bias: very serious. Although an RCT, data on AEs, SAEs and drug resistance markers was only collected
in the MDA arm, thus there is no control. Inconsistency: no serious. Indirectness: no serious. Imprecision: no
serious. Unable to calculate effect measure as there is no comparison group. Publication bias: no serious.
13. Risk of Bias: serious. Some risk of bias in most/all included studies. Inconsistency: no serious. Indirectness: no
serious. Imprecision: very serious. Wide confidence intervals including both no effect and appreciable benefit/ risk;
Small event numbers, does not meet Optimal Information Size. Publication bias: no serious.
349 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
14. Risk of Bias: serious. Some risk of bias in most/all included studies. Inconsistency: no serious. Indirectness: no
serious. Imprecision: serious. Small event numbers, does not meet Optimal Information Size. Publication bias: no
serious.
15. Risk of Bias: serious. Some risk of bias in most/all included studies. Inconsistency: no serious. Indirectness: no
serious. Imprecision: very serious. Wide confidence intervals including both no effect and appreciable benefit/ risk;
Small event numbers, does not meet Optimal Information Size. Publication bias: no serious.
16. Risk of Bias: serious. Some risk of bias in most/all included studies. Inconsistency: no serious. Indirectness: no
serious. Imprecision: serious. Small event numbers, does not meet Optimal Information Size. Publication bias: no
serious.
17. Risk of Bias: serious. Some risk of bias in most/all included studies. Inconsistency: no serious. Indirectness: no
serious. Imprecision: very serious. Wide confidence intervals including both no effect and appreciable benefit/ risk;
Small event numbers, does not meet Optimal Information Size. Publication bias: no serious.
18. Risk of Bias: serious. Some risk of bias in most/all included studies. Inconsistency: no serious. Indirectness: no
serious. Imprecision: serious. Small event numbers, does not meet Optimal Information Size. Publication bias: no
serious.
Attached Images
Summary
The systematic review identified two cRCTs and two NRSs in moderate to high transmission settings in four
countries (Burkina Faso, Gambia, Nigeria and Zambia) assessing the impact of MDA on P. falciparum compared to
no MDA (Schneider et al unpublished evidence). The cRCTs and NRSs were analysed and GRADEd separately.
Among the cRCTs, one study conducted four rounds of MDA with DP alone over 15 months and the other
conducted one round with SP+AS. Among the NRSs, one study provided nine rounds of sulfadoxine-
pyrimethamine every 10 weeks over 18 months and the other provided either chloroquine or amodiaquine in
combination with single low dose primaquine every 14 days for either eight or 15 rounds.
Meta-analyses of the results from the cRCTs showed little to no effect of MDA on P. falciparum prevalence or
incidence or the incidence of clinical malaria across all time points with low- to moderate-certainty. The results
from the NRSs were more likely to show a slight impact of MDA on P. falciparum prevalence at 4 – 12 and 12 –
24 months, with low-certainty evidence. Only one cRCT measured adverse events in a subset of both study arms
and found a small increase in adverse events in the MDA arm but the certainty of the evidence was very low.
None of the studies measured markers of drug resistance.
350 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Mass drug the Evidence Plain language
Timeframe measurements no MDA administration (Quality of summary
(MDA) evidence)
50 88
Relative risk 1.76 per 1000 per 1000
1-3 months - MDA may result in
(CI 95% 0.58 — 5.36) Low
Prevalence little to no difference in
Based on data from Due to very
Difference: 38 more per P. falciparum
786 participants in 1 serious
1000 prevalence between
studies. (Randomized ( CI 95% 21 imprecision 1 1-3 months
controlled) fewer — 219
more )
723 614
1-3 months - Relative risk 0.85 per 1000 per 1000 The evidence is very
Prevalence (CI 95% 0.78 — 0.93)
Very low uncertain about the
(NRS) Based on data from
Difference: 108 fewer per Due to serious effect of MDA on P.
1,000 participants in 1
1000 risk of bias 2 falciparum prevalence
studies. (Observational ( CI 95% 159 between 1-3 months
(non-randomized)) fewer — 51
fewer )
1-3 months -
Incidence of
Rate ratio 0.61
(CI 95% 0.4 — 0.92)
57 35 MDA probably reduces
per 1000 per 1000 Moderate the incidence of P.
parasitaemia Based on data from
Due to serious falciparum
820 participants in 1
imprecision 3 parasitaemia between
studies. (Randomized
1-3 months
controlled)
1-3 months -
Incidence of
Rate ratio 0.41
(CI 95% 0.04 — 4.42)
2 1 Low
MDA may result in
little to no difference in
Based on data from per 1000 per 1000 Due to very the incidence of P.
clinical malaria
144,422 participants in serious falciparum clinical
1 studies. (Randomized imprecision 4 malaria between 1-3
controlled) months
483 570
Relative risk 1.18 per 1000 per 1000
4-12 months - MDA may result in
(CI 95% 0.89 — 1.56) Low
Prevalence little to no difference in
Based on data from Due to very
Difference: 87 more per P. falciparum
1,497 participants in 1 serious
1000 prevalence between
studies. (Randomized ( CI 95% 53 imprecision 5 4-12 months
controlled) fewer — 271
more )
418 251
4- 12 months - Relative risk 0.6 per 1000 per 1000
Prevalence (CI 95% 0.55 — 0.67) MDA may reduce the
(NRS) Based on data from prevalence of P.
Difference: 167 fewer per Low
3,154 participants in 1 falciparum between
1000
studies. (Observational 4-12 months
( CI 95% 188
(non-randomized)) fewer — 138
fewer )
351 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Mass drug the Evidence Plain language
Timeframe measurements no MDA administration (Quality of summary
(MDA) evidence)
431 332
12-24 months - Relative risk 0.77 per 1000 per 1000
Prevalence (CI 95% 0.7 — 0.84) MDA may reduce P.
(NRS) Based on data from falciparum prevalence
Difference: 99 fewer per Low
3,261 participants in 1 between 12-24
1000
studies. (Observational months
( CI 95% 129
(non-randomized)) fewer — 69
fewer )
133 333
Odds ratio 3.25 per 1000 per 1000
Adverse events (CI 95% 0.68 — 15.53) Very low The evidence is very
Based on data from 90 Due to very uncertain about the
Difference: 200 more per
participants in 1 serious effect of MDA on
1000
studies. (Randomized ( CI 95% 39 inconsistency 7 adverse events
controlled) fewer — 572
more )
AEs among
people who
2 The evidence is very
Based on data from per 1000 uncertain about the
received MDA 8
336,821 participants in effect of MDA on
1 studies. (Randomized adverse events
controlled)
SAEs among
people who
0.01 The evidence is very
Based on data from per 1000 uncertain about the
received MDA 9
336,821 participants in effect of MDA on
1 studies. (Randomized adverse events
controlled)
1. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Wide confidence intervals
including both no effect and appreciable benefit/ risk. Publication bias: no serious.
2. Risk of Bias: serious. High risk of bias in all included studies. Inconsistency: no serious. Indirectness: no serious.
Imprecision: no serious. Publication bias: no serious.
3. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. GDG determined that the lower
confidence bound (5 fewer per 1000) was not an important reduction and concluded that the finding was imprecise..
Publication bias: no serious.
4. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Wide confidence intervals
including both no effect and appreciable benefit/ risk. Publication bias: no serious.
5. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Wide confidence intervals
including both no effect and appreciable benefit/ risk. Publication bias: no serious.
6. Risk of Bias: serious. High risk of bias in all included studies. Inconsistency: no serious. Indirectness: no serious.
Imprecision: very serious. Wide confidence intervals including both no effect and appreciable benefit/ risk.
Publication bias: no serious.
7. Inconsistency: very serious. Rates of events in both arms are much higher than in other studies; unclear how
352 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
questions were asked.. Indirectness: no serious. Imprecision: very serious. Wide confidence intervals including both
no effect and appreciable benefit/ risk. Publication bias: no serious.
8. Risk of Bias: very serious. Although an RCT, outcome was collected in MDA arm only, not in control group.
Inconsistency: no serious. Indirectness: no serious. Publication bias: no serious.
9. Risk of Bias: very serious. Although an RCT, outcome was collected in MDA arm only, not in control group.
Inconsistency: no serious. Indirectness: no serious. Publication bias: no serious.
Attached Images
Summary
The systematic review identified five cRCTs and seven NRSs in eight countries (Cambodia, India, Kenya, Lao
People’s Democratic Republic, Myanmar, Panama, Solomon Islands, Venezuela [Bolivarian Republic of] and Viet
Nam) assessing the impact of MDA on P. vivax transmission to no MDA (Schneider et al unpublished evidence). All
of the cRCTs used DP and four of the studies also administered single low-dose primaquine, but none of the
cRCTs used sufficient dosage of an 8-aminoquinoline to achieve radical cure of P. vivax hypnozoites1. One study
provided a single round of MDA while the other four conducted three rounds of MDA. Among the NRSs, only
one study reported radical cure of P. vivax. There was more variability in the design of MDA among the NRSs
with respect to drug regimens and number of rounds, ranging from a single round to 24 weekly rounds.
The meta-analysis of the data from cRCTs showed MDA may reduce P. vivax prevalence 1–3 months after the
last round of MDA but there was no impact of MDA on prevalence of P. vivax at later time periods. The certainty
of evidence obtained from the NRSs was very low across all time periods and outcomes. Data from a cRCT that
did not provide an 8-aminoquinoline medicine found that MDA probably did not increase the rate of severe
adverse events within 0 – 3 months.
1 The systematic review considered the following as the minimum adult dosage of 8-aminoquinoline medicines to achieve radical cure: 210
mg of primaquine over eight weeks; 1.25 g of plasmochin over 14 days. One study that contributed to the adverse events outcome (Comer
1971) considered its primaquine adult dosage regimen (40 mg of primaquine every two weeks for two years) to be radical cure, but as the
total dose for an eight-week period (i.e. 160 mg) was less than 210 mg, the systematic review did not consider this to be radical cure.
Intervention Certainty of
Outcome Study results and Comparator Mass drug the Evidence Plain language
Timeframe measurements no MDA administration (Quality of summary
(MDA) evidence)
231 42
1-3 months - Relative risk 0.18 per 1000 per 1000 Very low The evidence is very
Prevalence - (CI 95% 0.1 — 0.33)
Due to serious uncertain about the
NRS Based on data from
Difference: 189 fewer per risk of bias, Due effect of MDA on P.
1,024 participants in 2
1000 to serious vivax prevalence
studies. (Observational ( CI 95% 208 inconsistency 1 between 1-3 months
(non-randomized)) fewer — 155
fewer )
353 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Mass drug the Evidence Plain language
Timeframe measurements no MDA administration (Quality of summary
(MDA) evidence)
133 20
Relative risk 0.15 per 1000 per 1000
1-3 months - Low
(CI 95% 0.1 — 0.24)
Prevalence Due to serious MDA may reduce P.
Based on data from
Difference: 113 fewer per risk of bias, Due vivax prevalence
2,672 participants in 5
1000 to serious between 1-3 months
studies. (Randomized ( CI 95% 119 inconsistency 2
controlled) fewer — 101
fewer )
1-3 months -
Incidence of
Rate ratio 0.29 22 6 The evidence is very
(CI 95% 0.26 — 0.31) per 1000 per 1000
clinical malaria Based on data from Very low
uncertain about the
- NRS (low risk) effect of MDA on the
62,744 participants in Due to serious
incidence of P. vivax
2 studies. inconsistency 5 clinical malaria
(Observational (non-
between 1-3 months
randomized))
1-3 months -
Incidence of
Rate ratio 0.29 156 45 The evidence is very
(CI 95% 0.26 — 0.31) per 1000 per 1000
clinical malaria Based on data from Very low
uncertain about the
- NRS (high effect of MDA on the
62,744 participants in Difference: 111 fewer per Due to serious
risk) incidence of P. vivax
2 studies. 1000 inconsistency 6 clinical malaria
(Observational (non- 108 fewer — between 1-3 months
randomized)) 115 fewer
354 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Mass drug the Evidence Plain language
Timeframe measurements no MDA administration (Quality of summary
(MDA) evidence)
71 24
4-12 months - Relative risk 0.34 per 1000 per 1000 The evidence is very
Prevalence - (CI 95% 0.15 — 0.78) Very low
uncertain about the
NRS Based on data from Due to very
Difference: 47 fewer per effect of MDA on the
939 participants in 1 serious risk of
1000 prevalence of P. vivax
studies. (Observational ( CI 95% 60 bias 8 between 4-12 months
(non-randomized)) fewer — 16
fewer )
4-12 months -
Incidence of
Rate ratio 0.15
(CI 95% 0.07 — 0.34)
5 1 The evidence is very
per 1000 per 1000 Very low uncertain about the
parasitaemia- Based on data from
Due to very effect of MDA on the
NRS 223,990 participants in
serious risk of incidence of P. vivax
1 studies.
bias 9 parasitaemia between
(Observational (non-
4-12 months
randomized))
Very low
4-12 months - Rate ratio 1.38 41 57 Due to serious
The evidence is very
Incidence of (CI 95% 0.97 — 1.95) per 1000 per 1000 uncertain about the
risk of bias, Due
clinical malaria Based on data from effect of MDA on the
to serious
3,325 participants in 1 incidence of P. vivax
inconsistency,
studies. (Randomized clinical malaria
Due to serious
controlled) between 4-12 months
imprecision 10
4-12 months -
Incidence of
Rate ratio 0.72
(CI 95% 0.68 — 0.76)
156 112 The evidence is very
per 1000 per 1000 Very low
clinical malaria Based on data from uncertain about the
Due to very
- NRS 11,300 participants in effect of MDA on P.
serious risk of
1 studies. vivax clinical malaria
bias 11 between 4-12 months
(Observational (non-
randomized))
175 142
Relative risk 0.81 per 1000 per 1000
12-24 months - Low MDA may result in
(CI 95% 0.44 — 1.48)
Prevalence Due to serious little to no difference in
Based on data from
Difference: 33 fewer per risk of bias, Due P. vivax prevalence
243 participants in 1
1000 to serious between 12-24
studies. (Randomized ( CI 95% 98 imprecision 12 months
controlled) fewer — 84 more
)
12-24 months -
Incidence of
Rate ratio 0.04
(CI 95% 0.02 — 0.07)
156 6 The evidence is very
uncertain about the
per 1000 per 1000 Very low
clinical malaria Based on data from effect of MDA on the
Due to very
- NRS 11,300 participants in incidence of P. vivax
serious risk of
1 studies. clinical malaria
bias 13 between 12-24
(Observational (non-
randomized)) months
355 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Mass drug the Evidence Plain language
Timeframe measurements no MDA administration (Quality of summary
(MDA) evidence)
studies. (Randomized
months
controlled)
1. Risk of Bias: serious. High risk of bias in all included studies. Inconsistency: serious. Completely non-overlapping
confidence intervals. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious.
2. Risk of Bias: serious. High or unclear risk of bias in some/ all studies. Inconsistency: serious. Completely non-
overlapping confidence intervals; I-squared 84%. Indirectness: no serious. Imprecision: no serious. Publication bias:
no serious.
3. Risk of Bias: very serious. High risk of bias in all included studies.
4. Risk of Bias: very serious. High risk of bias in all included studies.
5. Risk of Bias: very serious. High risk of bias in all included studies. Inconsistency: serious. Completely non-
overlapping confidence intervals. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious.
6. Risk of Bias: very serious. High risk of bias in all included studies. Inconsistency: serious. Completely non-
overlapping confidence intervals. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious.
7. Risk of Bias: serious. High or unclear risk of bias in some/ all studies. Inconsistency: serious. I-squared 74%.
Indirectness: no serious. Imprecision: no serious. Publication bias: no serious.
8. Risk of Bias: very serious. High risk of bias in all included studies. Inconsistency: no serious. Indirectness: no
serious. Imprecision: no serious. Publication bias: no serious.
9. Risk of Bias: very serious. High risk of bias in all included studies. Inconsistency: no serious. Indirectness: no
serious. Imprecision: no serious. Publication bias: no serious.
10. Risk of Bias: serious. High or unclear risk of bias in some/ all studies. Inconsistency: serious. I-squared 52%.
Indirectness: no serious. Imprecision: serious. Wide confidence interval; include both null effect and appreciable
risk/ benefit. Publication bias: no serious.
11. Risk of Bias: very serious. High risk of bias in all included studies. Inconsistency: no serious. Indirectness: no
serious. Imprecision: no serious. Publication bias: no serious.
12. Risk of Bias: serious. High risk of bias in all included studies. Inconsistency: no serious. Indirectness: no serious.
Imprecision: serious. Wide confidence interval; include both null effect and appreciable risk/ benefit. Publication
bias: no serious.
13. Risk of Bias: very serious. High risk of bias in all included studies. Inconsistency: no serious. Indirectness: no
serious. Imprecision: no serious. Publication bias: no serious.
14. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Wide confidence interval; include both
null effect and appreciable risk/ benefit. Publication bias: no serious.
15. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Wide confidence interval; include both
null effect and appreciable risk/ benefit. Publication bias: no serious.
Attached Images
356 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Summary
The systematic review identified two NRSs that provided data on MRP for P. vivax (Shah et al unpublished
evidence). Studies were conducted in the Democratic People’s Republic of Korea in 2002 and in the Republic of
Azerbaijan in 1970–1971. Both studies provided primaquine for 14 days at 0.25 mg/kg per day, administered in
a single round prior to the peak transmission season. Both studies reported decreases in the incidence of P. vivax
1–3 months after the start of the intervention but the risk of bias in the studies was considered very serious.
Both studies found a decrease in the incidence of P. vivax 4–12 months after the intervention and one study
reported a decrease in the prevalence of P. vivax during that time period but the risk of bias in the studies was
considered very serious. Information on adverse events was obtained from the intervention group in one study:
no cases of severe haemolysis were reported, and side-effects were reported from less than 4% of 400 000
people. However, the overall certainty of the evidence was GRADEd as very low due to potential biases resulting
from the study designs.
357 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Mass relapse
Timeframe measurements No MRP (Quality of summary
prevention
evidence)
1-3 months -
Incidence of P.
Rate ratio 0.08
(CI 95% 0.07 — 0.08)
111 9 The evidence is very
per 1000 per 1000 Very low uncertain about the
vivax infection Based on data from
Due to very effect of MRP on the
- NRS 218,308 participants in
serious risk of incidence of P. vivax
2 studies.
bias 1 infection between 1-3
(Observational (non-
months
randomized))
4-12 months -
Prevalence -
Relative risk 0.07
(CI 95% 0.01 — 0.57)
4 0 The evidence is very
uncertain about the
per 1000 per 1000 Very low
NRS Based on data from effect of MRP on the
Due to risk of
6,710 participants in 1 prevalence of P. vivax
bias. 2 infection between
studies. (Observational
(non-randomized)) 4-12 months
Adverse events
40 Very low
per 1000 The evidence is very
Based on data from Due to serious
uncertain about the
333,946 participants in risk of bias, and
effect of MRP on
1 studies. very serious
adverse events
(Observational (non- indirectness 4
randomized))
1. Risk of Bias: very serious. Downgraded by 2 due to risk of bias. Many risk of bias domains judged as high risk or
not enough information to determine. High risk of bias due to confounding in both studies included for this outcome.
Inconsistency: no serious. Not downgraded for inconsistency. Both studies provided the same direction and a similar
magnitude (qualitatively) of effect. Indirectness: no serious. Not downgraded for indirectness since evidence was
judged to be sufficiently direct for the domains of population, intervention, comparator, direct comparison, and
outcome. Imprecision: no serious. Not downgraded for imprecision since lower and upper confidence limits indicate
the same direction of effect.
2. Risk of Bias: serious. Downgraded by 1 due to risk of bias. Quasi-experimental study design with a control group,
but allocation was not done at random and no baseline data were provided to assess potential confounders.
Inconsistency: no serious. Not downgraded for inconsistency due to single study result. Indirectness: no serious. Not
downgraded for indirectness since evidence was judged to be sufficiently direct for the domains of population,
intervention, comparator, direct comparison, and outcome. Imprecision: no serious. Not downgraded for imprecision
since lower and upper confidence limits indicate the same direction of effect.
3. Risk of Bias: very serious. Downgraded by 2 due to risk of bias. Many risk of bias domains judged as high risk or
not enough information to determine. High risk of bias due to confounding in both studies included for this outcome.
Inconsistency: no serious. Not downgraded for inconsistency. Both studies provided the same direction and a similar
magnitude (qualitatively) of effect. Indirectness: no serious. Not downgraded for indirectness since evidence was
judged to be sufficiently direct for the domains of population, intervention, comparator, direct comparison, and
outcome. Imprecision: no serious. Not downgraded for imprecision since lower and upper confidence limits indicate
the same direction of effect.
358 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
4. Risk of Bias: serious. Downgraded by 1 due to risk of bias. Quasi-experimental study design with a control group,
but allocation was not done at random and no baseline data were provided to assess potential confounders.
Inconsistency: no serious. Not downgraded for inconsistency due to single study result. Indirectness: very serious.
Downgraded by 2 due to indirectness. Side effects were not measured or reported in the control group, so evidence
is only provided in the intervention population. Imprecision: no serious. Not downgraded for imprecision since this
criteria is not applicable for this outcome (no effect measure presented). Upgrade: large magnitude of effect.
Attached Images
4.3. Vaccine
Summary
Systematic review summary
Three studies form the basis of these recommendations: two were individual randomized controlled trials (RCTs) and one
was an open-label extension study of an included RCT. One was a multicentre study comparing three or four doses of
the RTS,S/AS01 malaria vaccine to no malaria vaccination. The other RCT compared the RTS,S/AS01 malaria vaccine
alone to SMC alone, and also compared a combination of malaria vaccine and SMC to the malaria vaccine alone or SMC
alone. Based on WHO regions, all three studies were conducted in Africa, specifically: Burkina Faso (three studies),
Gabon, Ghana, Kenya (two studies), Malawi, Mali, Mozambique, and the United Republic of Tanzania (two studies).
In addition, data from the observational evaluation of the first 24 months of pilot implementation in Ghana, Malawi, and
Kenya were considered by MPAG/SAGE and included in the evidence summary.
The RCTs showed that RTS,S/AS01 reduces clinical malaria, hospital admissions with a positive malaria test,
hospitalization with severe malaria, all-cause hospital admissions, severe malaria anaemia and the need for blood
transfusions. Compared to SMC, RTS,S/AS01 is non-inferior in reducing clinical malaria and severe malaria anaemia and
may be superior in reducing hospitalization with severe malaria. The combination of RTS,S/AS01 and SMC is probably
better than SMC alone in reducing all-cause mortality and clinical malaria, and may reduce the need for blood
transfusions and all-cause hospital admissions. The pilot programme showed that delivery of RTS,S/AS01 through
routine systems probably reduces hospital admissions with severe malaria.
The RCTs had too few cases to determine an association between the vaccine and meningitis but the pilot study showed
that RTS,S/AS01 introduction was probably not associated with an increase in hospital admissions with meningitis.
There was uncertainty whether RTS,S/AS01 was associated with an increase in cerebral malaria in the RCTs but the pilot
programme showed that vaccine introduction was probably not associated with an increase in hospital admission with
cerebral malaria. One RCT found that vaccination with RTS,S/AS01 may be associated with an increase in deaths in girls,
but the other found no evidence that the effect of RTS,S/AS01 (alone or in combination with SMC) on mortality differed
between girls and boys compared to SMC alone. The pilot programme found that the effect of the RTS,S/AS01 vaccine
introduction on all-cause mortality probably did not differ between girls and boys.
359 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator RTS,S/AS01 the Evidence Plain language
Timeframe measurements No vaccination malaria (Quality of summary
vaccination evidence)
Protective
efficacy (%) Difference: 1,774 fewer per
against clinical 1000
malaria ( CI 95% 1,387
episodes; fewer — 2,186
4-doses of 36.3 fewer )
RTS,S/AS01 (CI 95% 31.8 — 40.5)
Based on data from
versus control 1 RTS,S/AS01 vaccination
5,950 participants in 1 High
Ph 3 randomized reduces clinical malaria.
trial 2009–2014 studies. 2 (Randomized
(month 0 to end controlled)
of study); median Follow up: 48 months.
of 48 months'
follow-up
6 Important
Protective
efficacy (%)
305 278
per 1000 per 1000
against clinical
malaria of
Difference: 27 fewer per
vaccine alone 7.9
1000
versus SMC (CI 99% -1 — 16)
( CI 95% 13 fewer RTS,S/AS01 vaccination
alone 3 Based on data from
3,953 participants in 1
— 40 fewer ) High is non inferior to SMC in
Phase 3b reducing clinical malaria.
randomized study studies. 4 (Randomized
2017–2020; 3 controlled)
years' follow-up
6 Important
Protective
efficacy (%)
305 113
per 1000 per 1000
against clinical
malaria of
Difference: 192 fewer per
vaccine + SMC 62.8 1000
combination The combination of
(CI 95% 58.4 — 66.8) ( CI 95% 182
versus SMC RTS,S/AS01 vaccination
Based on data from fewer — 200
alone 5 3,932 participants in 1 fewer )
High with SMC is superior to
Phase 3b SMC alone in reducing
studies. 6 (Randomized
randomized study clinical malaria.
controlled)
2017–2020; 3
years' follow-up
6 Important
Protective
efficacy (%) 32.2 Difference: 19 fewer per
against severe (CI 95% 13.7 — 46.9) 1000
malaria Based on data from ( CI 95% 4 fewer
— 35 fewer ) High RTS,S/AS01 vaccination
episodes; 4 5,950 participants in 1 9 reduces severe malaria.
vaccine doses studies. 8 (Randomized
versus control 7 controlled)
Ph 3 randomized Follow up: 48 months.
trial 2009–2014
360 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator RTS,S/AS01 the Evidence Plain language
Timeframe measurements No vaccination malaria (Quality of summary
vaccination evidence)
(month 0 to end
of study); median
of 48 months'
follow-up
9 Critical
Protective
efficacy (%)
6.8 6.7
per 1000 per 1000
against
hospitalization
Difference: 0.1 fewer per
due to severe 1000
malaria of -0.4 There may be little or no
( CI 95% 2 fewer
vaccine alone (CI 95% -60.2 — 37.1)
— 2.4 more )
Low difference between
versus SMC Based on data from Due to very RTS,S/AS01 vaccination
3,953 participants in 1 serious and SMC in reducing
alone 10
Phase 3b studies. 11 (Randomized imprecision 12 hospitalization with
controlled) severe malaria.
randomized study
2017–2020; 3
years' follow-up
9 Critical
Protective
efficacy (%)
6.8 2
per 1000 per 1000
against
hospitalization
Difference: 4.8 fewer per
due to severe 1000
malaria of 70.5 ( CI 95% 3.2 The combination of
vaccine + SMC (CI 95% 41.9 — 85) fewer — 5.7 fewer RTS,S/AS01 vaccination
combination ) Moderate
Based on data from with SMC may be
versus SMC Due to serious
3,932 participants in 1 superior to SMC alone in
alone 13 imprecision 15
studies. 14 (Randomized reducing hospitalization
Phase 3b controlled) with severe malaria.
randomized study
2017–2020, 3
years' follow-up
9 Critical
Incidence rate
ratio for impact
of routine
RTS,S/AS01
vaccination on
hospitalization RTS,S/AS01 vaccine
0.7
with severe (CI 95% 0.54 — 0.92) Moderate
introduction is probably
malaria in associated with a
Based on data from Due to serious
implementing reduction in incidence of
27,678 participants in 1 imprecision 18
versus hospital admissions with
studies. 17
comparison severe malaria.
areas 16
Pilot
implementation
study 2019–2021
(month 0 to
361 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator RTS,S/AS01 the Evidence Plain language
Timeframe measurements No vaccination malaria (Quality of summary
vaccination evidence)
month 24)
9 Critical
Protective
efficacy (%) Difference: 11 fewer per
against severe 1000
malaria anaemia; ( CI 95% 1 fewer
4 vaccine doses 47.8 — 24 fewer )
versus control 19 (CI 95% 11.6 — 69.9)
Ph 3 randomized Based on data from Moderate RTS,S/AS01 vaccination
trial 2009–2014 5,950 participants in 1 Due to serious probably reduces severe
(month 0 to end studies. 20 (Randomized imprecision 21 malaria anaemia.
of study); median controlled)
of 48 months' Follow up: 48 months.
follow-up
6 Important
Protective
efficacy (%)
5.69 4.52
per 1000 per 1000
against severe
malaria anaemia
Difference: 1.17 fewer per
of vaccine alone 18.4
1000 There may be little or no
versus SMC (CI 95% -39.3 — 52.2) Low
( CI 95% 2.64 difference between
alone 22 Based on data from Due to very
fewer — 0.99 RTS,S/AS01 vaccination
Phase 3b 3,953 participants in 1 serious
more ) and SMC in reducing
randomized study studies. 23 (Randomized imprecision 24 severe malaria anaemia.
2017–2020, 3 controlled)
years' follow-up
6 Important
Protective
efficacy (%)
5.69 1.82
per 1000 per 1000
against severe
malaria anaemia
Difference: 3.87 fewer per
of vaccine + 1000
SMC 67.9 The combination of
( CI 95% 2.32
combination (CI 95% 34.1 — 84.3) RTS,S/AS01 vaccination
fewer — 4.71 Moderate
versus SMC Based on data from with SMC may be
fewer ) Due to serious
3,932 participants in 1 superior to SMC alone in
alone 25 imprecision 27
Phase 3b studies. 26 (Randomized reducing severe malaria
controlled) anaemia.
randomized study
2017–2020, 3
years' follow-up
6 Important
Protective 28.5
Difference: 15 fewer per RTS,S/AS01 vaccination
efficacy (%) (CI 95% 3.5 — 47.2) Moderate
1000 probably reduces the
against blood Based on data from Due to serious
need for blood
transfusions; 4 5,950 participants in 1 ( CI 95% 1 fewer imprecision 30 transfusions.
vaccine doses studies. 29 (Randomized — 31 fewer )
362 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator RTS,S/AS01 the Evidence Plain language
Timeframe measurements No vaccination malaria (Quality of summary
vaccination evidence)
versus control 28
Ph 3 randomized
trial 2009–2014
(month 0 to end
of study); median
controlled)
of 48 months'
follow-up
6 Important
Protective
efficacy (%)
4.22 3.79
per 1000 per 1000
against blood
transfusion of
Difference: 0.43 fewer per
vaccine alone 8.27 There may be little or no
1000
versus SMC (CI 95% -67.6 — 49.8) Low difference between
( CI 95% 1.75
alone 31 Based on data from Due to very RTS,S/AS01 vaccination
fewer — 1.6 more
Phase 3b 3,953 participants in 1 serious and SMC in reducing the
)
randomized study studies. 32 (Randomized imprecision 33 need for blood
2017–2020; 3 controlled) transfusions.
years' follow-up
9 Critical
Protective
efficacy (%)
4.22 1.45
per 1000 per 1000
against blood
transfusions of
Difference: 2.77 fewer per
vaccine + SMC 65.4 1000 The combination of
combination (CI 95% 22.9 — 84.5) ( CI 95% 1.32 Low RTS,S/AS01 vaccination
versus SMC Based on data from fewer — 3.49 Due to very with SMC may be
alone 34 3,932 participants in 1 fewer ) serious superior to SMC alone in
Phase 3b studies. 35 (Randomized imprecision 36 reducing the need for
randomized study controlled) blood transfusions.
2017–2020; 3
years' follow-up
9 Critical
Protective
efficacy (%) Difference: 59 fewer per
against all-cause 1000
hospital ( CI 95% 18 fewer
admissions; 4 — 103 fewer )
vaccine doses 16.5
versus control 37 (CI 95% 7.2 — 24.9)
RTS,S/AS01 vaccination
Ph 3 randomized Based on data from High
reduces all-cause
trial 2009–2014 5,950 participants in 1 39
hospital admissions.
(month 0 to end studies. 38 (Randomized
of study); median controlled)
of 48 months'
follow-up
9 Critical
363 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator RTS,S/AS01 the Evidence Plain language
Timeframe measurements No vaccination malaria (Quality of summary
vaccination evidence)
Protective
efficacy (%)
11 13.2
per 1000 per 1000
against all-cause
hospital
Difference: 2.2 more per
admissions of -22.3 1000 There may be little or no
vaccine alone (CI 95% -74.4 — 14.3) ( CI 95% 0.5 Low difference between
versus SMC Based on data from fewer — 5.6 more Due to very RTS,S/AS01 vaccination
alone 40 3,953 participants in 1 ) serious and SMC in reducing all-
Phase 3b studies. 41 (Randomized imprecision 42 cause hospital
randomized study controlled) admissions.
2017–2020; 3
years' follow-up
9 Critical
Protective
efficacy (%)
11 8.9
per 1000 per 1000
against all-cause
hospital
Difference: 2.1 fewer per
admissions of 1000
vaccine + SMC 18.7 The combination of
( CI 95% 4.28
combination (CI 95% -19.4 — 44.7)
fewer — 0.8 more
Low RTS,S/AS01 vaccination
versus SMC Based on data from Due to very with SMC may be
)
3,932 participants in 1 serious superior to SMC alone in
alone 43
Phase 3b studies. 44 (Randomized imprecision 45 reducing all-cause
controlled) hospital admissions.
randomized study
2017–2020; 3
years' follow-up
9 Critical
Incidence rate
ratio for impact
of routine
RTS,S/AS01
vaccination on
all-cause
hospital
admissions in 0.92 RTS,S/AS01 vaccine
implementing (CI 95% 0.83 — 1.03) Moderate introduction probably
versus Based on data from Due to serious has little or no difference
comparison 27,678 participants in 1 imprecision 48 on all-cause hospital
areas 46 studies. 47 admissions.
Pilot
implementation
study 2019–2021
(month 0 to
month 24)
9 Critical
364 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator RTS,S/AS01 the Evidence Plain language
Timeframe measurements No vaccination malaria (Quality of summary
vaccination evidence)
hospital
admissions with
a positive
malaria test in
implementing
versus
comparison
areas 49
Pilot
implementation
study 2019–2021
(month 0 to
month 24)
9 Critical
Protective
efficacy (%)
against all-cause
mortality; 3 or 4
vaccine doses
versus control 52 There were too few
Low
Ph 3 randomized deaths to determine the
Based on data from Due to very
trial 2009–2014 impact of RTS,S/AS01
8,922 participants in 1 serious
(month 0 to end vaccination on all-cause
studies. 53 (Randomized imprecision 54
of study); median mortality.
controlled)
of 48 months'
follow-up
9 Critical
Protective
efficacy (%)
4.59 3.97
per 1000 per 1000
against all-cause
mortality;
Difference: 0.62 fewer per
vaccine alone 12.1 There may be little or no
1000
versus SMC (CI 95% -55.7 — 50.4) Low difference between the
( CI 95% 1.97
alone 55 Based on data from Due to very impact of RTS,S/AS01
fewer — 1.45
Phase 3b 3,953 participants in 1 serious vaccination and SMC
more )
randomized study studies. 56 (Randomized imprecision 57 administration on all-
2017–2020; 3 controlled) cause mortality.
years' follow-up
9 Critical
Protective
efficacy (%)
4.59 2.18
per 1000 per 1000
against all-cause
52.3 The combination of
mortality of
(CI 95% 4.99 — 76) Difference: 2.41 fewer per RTS,S/AS01 vaccination
vaccine + SMC 1000
Moderate
Based on data from and SMC is probably
combination Due to serious
3,932 participants in 1 ( CI 95% 0.75 associated with a
versus SMC imprecision 60
studies. 59 (Randomized fewer — 3.35 reduction in all-cause
alone 58 controlled) fewer ) mortality.
Phase 3b
randomized study
2017–2020; 3
365 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator RTS,S/AS01 the Evidence Plain language
Timeframe measurements No vaccination malaria (Quality of summary
vaccination evidence)
years' follow-up
9 Critical
Incidence rate
ratio of
meningitis; 3 or
4 vaccine doses 10.5 Low There were too few
versus control 61 (CI 95% 1.41 — 78) Due to serious meningitis cases to
Post-hoc analysis Based on data from risk of bias, Due determine an association
of Ph 3 8,922 participants in 1 to serious with RTS,S/AS01
randomized trial studies. 62 imprecision 63 vaccination.
2009–2014
9 Critical
Incidence rate
ratio of
meningitis in
vaccine alone
versus SMC
alone versus There were no
combination of Low
meningitis cases to
vaccine with Based on data from Due to very
determine an association
6,861 participants in 1 serious
SMC 64 with RTS,S/AS01
Phase 3b studies. 65 (Randomized imprecision 66 vaccination.
randomized study controlled)
2017–2020; 3
years' follow-up
9 Critical
Incidence rate
ratio of hospital
admissions with
meningitis;
vaccine
There is probably no
implementing evidence that RTS,S/
versus 0.81
AS01 vaccine
comparison (CI 95% 0.43 — 1.55) Moderate
introduction is
Based on data from Due to serious
areas 67 associated with an
27,678 participants in 1 imprecision 69
Pilot increase in hospital
implementation studies. 68
admissions with
study 2019–2021 meningitis.
(month 0 to
month 24)
9 Critical
366 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator RTS,S/AS01 the Evidence Plain language
Timeframe measurements No vaccination malaria (Quality of summary
vaccination evidence)
groups 70
Post-hoc analysis
of Ph 3
randomized trial
2009–2014
9 Critical
Incidence rate
ratio of cerebral
malaria in
vaccine alone
versus SMC
alone vs There were too few
combination of Low
cerebral malaria cases to
vaccine with Based on data from Due to very
determine an association
5,920 participants in 1 serious
SMC 73 with RTS,S/AS01
Phase 3b studies. 74 (Randomized imprecision 75 vaccination.
randomized study controlled)
2017–2020; 3
years' follow-up
9 Critical
Incidence rate
ratio of hospital
admissions with
cerebral malaria;
vaccine
There is probably no
implementing evidence that RTS,S/
versus 0.77 Moderate
AS01 vaccine
comparison (CI 95% 0.44 — 1.35) Due to serious
introduction is
Based on data from inconsistency and
areas 76 associated with an
27,678 participants in 1 serious
Pilot increase in hospital
implementation studies. 77 imprecision 78 admissions with cerebral
study 2019–2021 malaria.
(month 0 to
month 24)
9 Critical
Female:male risk
ratio of vaccine
impact on all-
cause mortality;
4-dose + 3-dose 1.5 RTS,S/AS01 vaccination
versus control Low
(CI 95% 1.03 — 2.08) may be associated with
groups 79 Due to very
Based on data from an increase in deaths in
Post-hoc analysis serious
8,922 participants in 1 girls and a decrease in
of Ph 3 imprecision 81
studies. 80 deaths in boys.
randomized trial
2009–2014
9 Critical
367 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator RTS,S/AS01 the Evidence Plain language
Timeframe measurements No vaccination malaria (Quality of summary
vaccination evidence)
Female:male
rate ratio on
vaccine impact
on all-cause
mortality; 1.8
vaccine alone There may be no
(CI 95% 0.56 — 5.79) Low
versus SMC evidence that the effect
Based on data from Due to very
alone 82 of RTS,S/AS01
3,953 participants in 1 serious
Phase 3b vaccination differs
studies. 83 (Randomized imprecision 84
randomized study between girls and boys.
controlled)
2017–2020; 3
years' follow-up
9 Critical
Female:male
rate ratio for all-
cause mortality;
combination of
vaccine with 0.35
There may be no
SMC versus (CI 95% 0.06 — 1.98) Low
evidence that the effect
SMC alone 85 Based on data from Due to very
of RTS,S/AS01
Phase 3b 3,932 participants in 1 serious
vaccination differs
randomized study studies. 86 (Randomized imprecision 87 between girls and boys.
2017–2020; 3 controlled)
years' follow-up
9 Critical
Female:male
rate ratio of all-
cause mortality Moderate
Due to serious
ratio; vaccine
imprecision
implementing
because not yet
versus There is probably no
1.08 powered to assess
comparison evidence that the effect
(CI 95% 0.93 — 1.25) overall impact on
areas 88 of RTS,S/AS01 vaccine
Based on data from all-cause
Pilot introduction on all-cause
13,682 participants in 1 mortality,
implementation mortality differs
studies. 89 however well
study 2019–2021 between girls and boys.
powered to
(month 0 to detect gender
month 24) imbalance in all-
cause mortality 90
9 Critical
1. Clinical malaria episodes (from month 0 to end of study; median follow-up: 48 months) (modified ITT analysis) assessed
with: illness in a child brought to a study facility with a measured temperature of 37.5°C and P. falciparum asexual =
parasitaemia at a density of > 5000 parasites per cubic millimetre or a case of malaria meeting the primary case definition of
severe malaria. Severe malaria primary case definition = P. falciparum asexual parasitaemia at a density of > 5000 parasites
per cubic millimetre with one or more markers of disease severity and without diagnosis of a coexisting illness. Markers of
severe disease were prostration, respiratory distress, a Blantyre coma score of 2 (on a scale of 0 to = 5, with higher scores
indicating a higher level of consciousness), two or more observed or reported seizures, hypoglycaemia, acidosis, elevated
lactate level, or haemoglobin level of < 5 g per decilitre. Coexisting illnesses were defined as radiographically proven
pneumonia, meningitis established by analysis of cerebrospinal fluid, bacteraemia, or gastroenteritis with severe
dehydration); four-dose group = three doses of RTS,S/AS01 at months 0, 1, and 2 and a booster dose at month 20; Control
368 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
group received comparator vaccine at months 0, 1, 2, and 20; Protective efficacy = (1-hazard ratio); Per Protocol analysis: VE
28.5% (95% CI 6.3 to 45.7)
2. Primary study[143]. The number of cases averted over time was calculated as the sum of 3-monthly differences in the
estimated number of cases between the control and the RTS,S/AS01 groups (R3R and R3C combined up to the time of
booster dose and R3R and R3C separately after the booster dose) and expressed per 1000 participants vaccinated. Among
the older children, in the 12 months following administration of the first three doses, vaccine efficacy against clinical
(uncomplicated and severe) malaria was 51% (95% CI 47-55) (per protocol analysis). Baseline/comparator: . Supporting
references: [143],
3. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
SMC = combination group).
4. Primary study[145]. The RTS,S vaccine alone group had 1,540 clinical malaria cases over 5535.7 total person-years at
risk (PYAR) for an incidence rate of 278 cases (95% CI: 264.6 to 292.4) per 1000 PYAR; The SMC alone group had 1,661
cases over 5449.9 total PYAR for an incidence rate of 305 cases (95% CI: 290.5 to 319.8) per 1000 PYAR;. Baseline/
comparator: . Supporting references: [145], The 90, 95, and 99% CIs for the hazard ratio (HR) all excluded the pre-specified
non-inferiority margin of 1.20..
5. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
SMC = combination group).
6. Primary study[145]. The RTS,S + SMC combined group had 624 clinical malaria cases over 5508.0 total PYAR for an
incidence rate of 113 cases (95% CI: 104.7 to 122.5) per 1000 PYAR; The SMC alone group has 1,661 cases over 5449.9
total PYAR for an incidence rate of 305 cases (95% CI: 290.5 to 319.8) per 1000 PYAR;. Baseline/comparator: . Supporting
references: [145],
7. Assessed with P. falciparum asexual parasitaemia at a density of > 5000 parasites per cubic millimetre with one or more
markers of disease severity and without diagnosis of acoexisting illness. Markers of severe disease were prostration,
respiratory distress, a Blantyre coma score of 2 (on a scale of 0 to = 5, with higher scores indicating a higher level of
consciousness), two or more observed or reported seizures, hypoglycaemia, acidosis, elevated lactate level, or haemoglobin
level of < 5 g per decilitre. Coexisting illnesses were defined as radiographically proven pneumonia, meningitis established by
analysis of cerebrospinal fluid, bacteraemia, or gastroenteritis with severe dehydration). 4-dose group = three doses of
RTS,S/AS01 at months 0, 1, and 2 and a booster dose at month 20; Control group received = comparator vaccine at months
0, 1, 2, and 20. Protective efficacy = (1-hazard ratio). Per Protocol analysis: VE 28.5% (95%CI: 6.3 to 45.7)
8. Primary study[143]. Among the older children, in the 12 months following administration of the first three doses,
vaccine efficacy against severe malaria was 45% (95% CI 22-60) (per protocol analysis).. Baseline/comparator: . Supporting
references: [143], PP analysis VE: 28.5% (95% CI: 6.3 to 45.7); The number of cases averted overtime was calculated as the
sum of 3-monthly differences in the estimated number of cases between the control and the RTS,S/AS01 groups (R3R and
R3C combined up to the time of booster dose and R3R and R3C separately after the booster dose) and expressed per 1000
participants vaccinated..
9. Risk of Bias: no serious. Study was rated as unclear risk of bias due to heavy involvement of the funder in the project;
however, it has not been downgraded for risk of bias as this was the only concern and the study was carefully scrutinized by
independent experts and considered well conducted.. Inconsistency: no serious. Indirectness: no serious. Imprecision: no
serious. Publication bias: no serious.
10. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
SMC = combination group).
11. Primary study[145]. The RTS,S vaccine alone group had 37 severe malaria cases (of which 25 were severe malaria
anaemia) over 5535.7 total PYAR for an incidence rate of 6.7 severe malaria cases (95% CI: 4.8 to 9.2) per 1000 PYAR; The
SMC alone group had 37 cases (of which 31 were severe malaria anaemia) over 5449.9 total PYAR for a rate of 6.8 cases
(95% CI: 4.9 to 9.4) per 1000 PYAR;. Baseline/comparator: . Supporting references: [145], Most cases of severe malaria
were severe malaria anaemia (vaccine: 25/37; SMC: 31/37).
12. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Downgraded two levels due to
imprecision: few events and a very large CI that incorporates the possibility of benefit and harm. Publication bias: no
serious.
13. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
SMC = combination group).
14. Primary study[145]. Combination group of RTS,S + SMC had 11 severe malaria cases (of which 10 were severe malaria
anaemia) over 5508 total PYAR for an incidence rate of 2.0 severe malaria cases (95% CI: 1.1 to 3.6) per 1000 PYAR; The
369 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
SMC alone group has 37 cases (of which 31 were severe malaria anaemia) over 5449.9 total PYAR for a rate of 6.8 cases
(95% CI: 4.9 to 9.4) per 1000 PYAR;. Baseline/comparator: . Supporting references: [145],
15. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Downgraded one level due to imprecision:
few events and large CI. Publication bias: no serious.
16. Pilot implementation study designed to be analysed using cluster randomized control methodology. Across the three
countries, there was a total of 27 678 admissions to sentinel hospitals in children 1-59 months during the period from
vaccine introduction until 30 April 2021: 4,853 were vaccine-eligible based on their date of birth out of 13,918 total
admissions in areas where the vaccine was provided (implementating areas); 5,141 were vaccine-eligible out of 13,760 total
admissions in comparison areas
17. [155]. Among children eligible to have received all three primary doses of RTS,S/AS01, there was a total of 1107
admissions with severe malaria (out of 9,994 total age-eligible admissions), 418 from implementation areas and 689 from
comparison areas. Among children who were not eligible there were 2,703 total admissions with severe malaria (out of
17,684 total age-ineligible admissions) to have received any doses of RTS,S/AS01: 1313 from implementation areas and
1390 from comparison areas. The incidence rate ratio comparing incidence of admission with severe malaria between
implementing and comparison areas was 0.70 (95%CI 0.54 to 0.92), a reduction of 30% (95%CI 8% to 46%); there was no
evidence that effectiveness differed between cerebral malaria and other forms of severe malaria. Baseline/comparator: .
18. Risk of Bias: no serious. Not downgraded for risk of bias despite being an open-label study because the findings from
the household survey suggestthere is no evidence that the introduction of RTS,S/AS01 had a negative effect on the uptake
of other childhood vaccines, ITN use, care-seeking behaviour, or health worker behaviour in testing and treating for febrile
illness.. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Downgraded one level for imprecision: few
events and large CI. Publication bias: no serious.
19. Assessed with: a documented haemoglobin < 5·0 g per decilitre identified at clinical presentation to morbidity
surveillance system in association with a P. falciparum parasitaemia at a density of > 5000 parasites per cubic millimetre.
4-dose group = three doses of RTS,S/AS01 at months 0, 1, and 2 and a booster dose at month 20; Control group received =
comparator vaccine at months 0, 1, 2, and 20. Protective efficacy = (1-hazard ratio).
20. Primary study[143]. Baseline/comparator: .
21. Risk of Bias: no serious. Study was rated as unclear risk of bias due to heavy involvement of the funder within the
project; however, it has not been downgraded for ROB as this was the only concern and the study was carefully scrutinized
by independent experts and considered well conducted.. Inconsistency: no serious. Indirectness: no serious. Imprecision:
serious. Downgraded one level due to imprecision: few events and large confidence interval. Publication bias: no serious.
22. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
SMC = combination group).
23. Primary study[145]. The RTS,S vaccine group had 25 severe malaria anemia cases over 5535.7 total PYAR for an
incidence rate of 4.52 cases (95% CI: 3.05 to 6.68) per 1000 PYAR; The SMC alone group has 31 cases over 5449.9 total
PYAR for a rate of 5.69 cases (95% CI: 4.00 to 8.09) per 1000 PYAR;. Baseline/comparator: . Supporting references: [145],
24. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Downgraded two levels due to
imprecision: few events and a very large confidence interval that incorporates the possibility of benefit and harm.
Publication bias: no serious.
25. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
SMC = combination group).
26. Primary study[145]. The RTS,S vaccine and SMC combination group had 10 severe malaria anaemia cases over 5508
total PYAR for an incidence rate of 1.82 cases (95% CI: 0.977 to 3.37) per 1000 PYAR; The SMC alone group had 31 cases
over 5449.9 total PYAR for a rate of 5.69 cases (95% CI: 4.00 to 8.09) per 1000 PYAR;. Baseline/comparator: . Supporting
references: [145],
27. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Downgraded one level due to imprecision:
few events and a very large CI. Publication bias: no serious.
28. 4-dose group = three doses of RTS,S/AS01 at months 0, 1, and 2 and a booster dose at month 20; Control group
received = comparator vaccine at months 0, 1, 2, and 20. Protective efficacy = (1-hazard ratio).
29. Primary study[143]. Baseline/comparator: .
30. Risk of Bias: no serious. Study was rated as unclear risk of bias due to heavy involvement of the funder in the project;
however, it has not been downgraded for risk of bias as this was the only concern and the study was carefully scrutinized by
independent experts and considered well conducted.. Inconsistency: no serious. Indirectness: no serious. Imprecision:
serious. Downgraded one level due to imprecision: few events and large CI. Publication bias: no serious.
31. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
370 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
371 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
illness.. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Downgraded one level due to imprecision:
large CI that incorporates the possibility of benefit and harm. Study was powered for a pooled analysis only, country
estimates vary but confidence intervals are wide and consistent with pooled effect.. Publication bias: no serious.
49. Pilot implementation study designed to be analysed using cluster randomized control methodology. Across the three
countries, there were a total of 27,678 admissions to sentinel hospitals in children 1-59 months during the period from
vaccine introduction until 30 April 2021: 4,853 were vaccine-eligible based on their date of birth out of 13,918 total
admissions in areas where the vaccine was provided (implementing areas); 5,141 were vaccine-eligible out of 13,760 total
admissions in comparison areas.
50. [155]. Patients admitted to sentinel hospitals were routinely tested for malaria infection by rapid diagnostic test (RDT)
or microscopy. Out of a total of 27,678 patients admitted, test results were available for 88%. Among children eligible to
have received three vaccine doses, the number of patients admitted with a positive malaria test was 2630-- 1075 from
implementation areas and 1555 from comparison areas. The rate ratio comparing the incidence of hospital admission with a
positive malaria test between implementation and comparison areas was 0.79 (95%CI 0.68 to 0.93), a reduction of 21%
(95%CI 7% to 32%).. Baseline/comparator: .
51. Risk of Bias: no serious. Not downgraded for risk of bias despite being an open-label study because the findings from
the household survey suggest there is no evidence that the introduction of RTS,S/AS01 had a negative effect on uptake of
other childhood vaccines, ITN use, care-seeking behaviour, or health worker behaviour in testing and treating for febrile
illness.; . Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious.
52. 4-dose group = three doses of RTS,S/AS01 at months 0, 1, and 2 and a booster dose at month 20; 3-dose group =
three doses of RTS,S/AS01 at months 0, 1, and 2 and a comparator vaccine at month 20; Control group received =
comparator vaccine at months 0, 1, 2, and 20. Protective efficacy = (1-hazard ratio).
53. [143]. Four dose group: 61 deaths (13 malaria)/2976 children + Three dose group: 51 deaths (17 malaria) / 2972
children vs Control group: 46 deaths (13 malaria) / 2974 children.. Baseline/comparator: .
54. Risk of Bias: no serious. Study was rated as unclear risk of bias due to heavy involvement of the funder in the project;
however, it has not been downgraded for risk of bias as this was the only concern and the study was carefully scrutinized by
independent experts and considered well conducted. Inconsistency: no serious. Indirectness: no serious. Imprecision: very
serious. Downgraded two levels due to imprecision: few events and a very large CI that incorporates the possibility of
benefit and harm; . Publication bias: no serious.
55. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
SMC = combination group).
56. Primary study[145]. In the RTS,S vaccine alone group there were 22 deaths total/1734 participants or 3.97 deaths
(95% CI 2.92 to 6.04) per 1000 PYAR. In the SMC alone group, there were 25 deaths total/1716 participants or 4.59 deaths
(95% CI 3.10 to 6.79) per 1000 PYAR.. Baseline/comparator: .
57. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Downgraded two levels due to
imprecision: few events and a very large confidence interval that incorporates the possibility of benefit and harm; .
Publication bias: no serious.
58. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
SMC = combination group).
59. Primary study[145]. In the RTS,S vaccine + SMC combination group there were 12 deaths total/1740 children or 2.18
deaths (95% CI 1.24 to 3.84) per 1000 PYAR. In the SMC alone group, there were 25 deaths total/1716 children or 4.59
deaths (95% CI 3.10 to 6.79) per 1000 PYAR.. Baseline/comparator: .
60. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Downgraded one level due to imprecision:
few events and large CI.. Publication bias: no serious.
61. mITT analysis; 4-dose group = three doses of RTS,S/AS01 at months 0, 1, and 2 and a booster dose at month 20;
3-dose group = three doses of RTS,S/AS01 at months 0, 1, and 2 and a comparator vaccine at month 20; Control group
received = comparator vaccine at months 0, 1, 2, and 20. Protective efficacy = (1-hazard ratio).
62. [143]. 4-dose group 11/2976 + 3-dose group 10/2972 vs Control group 1/2974. Baseline/comparator: .
63. Risk of Bias: serious. This outcome was not pre-specified in the protocol (post-hoc analysis). Study was rated as unclear
risk of bias due to heavy involvement of the funder within the project.. Inconsistency: no serious. Indirectness: no serious.
Imprecision: serious. Downgraded one level due to imprecision: few events and large confidence interval; . Publication bias:
no serious.
64. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
SMC = combination group).
65. Primary study[145]. Eight cases of clinically suspected meningitis (four in the SMC-alone group, three in the RTS,S
372 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
vaccine-alone group, and one in the RTS,S + SMC combined group) were investigated with the use of lumbar puncture, but
none showed proven meningitis.. Baseline/comparator: .
66. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Downgraded two levels for imprecision:
no events reported in any groups. Publication bias: no serious.
67. Pilot implementation study designed to be analysed using cluster randomized control methodology; to be able to rule
out an association with meningitis of the magnitude seen in the Phase 3 trial it would therefore be necessary to exclude rate
ratios of about 10.5 (4.5 allowing for coverage and contamination) or more. Across the three countries, there was a total of
27,678 admissions to sentinel hospitals in children 1-59 months during the period from vaccine introduction until 30 April
2021: 4,853 were vaccine-eligible based on their date of birth out of 13,918 total admissions in areas where the vaccine was
provided (implementing areas); 5,141 were vaccine-eligible out of 13,760 total admissions in comparison areas
68. Primary study[155]. A total of 4,311 suspected cases of meningitis were investigated. Lumbar punctures were
performed in 2,652 (62%) of these patients, and PCR analysis of samples of cerebrospinal fluid (CSF) was available for 2,249
patients (52%). A total of 51 cases of probable or confirmed meningitis were seen in sentinel hospitals among age groups of
children eligible for the malaria vaccine: 27 from implementation areas and 24 from comparison areas. Among the age
groups that were not eligible for the malaria vaccine, there were 79 probable or confirmed cases of meningitis: 44 from
implementation areas and 35 from comparison areas. The incidence rate ratio comparing rates of admission with meningitis
in implementation and comparison areas, among vaccine-eligible children, was 0.81 (95%CI 0.43 to 1.55). There was
therefore no evidence that introduction of the malaria vaccine led to an increase in the incidence of hospital admission with
meningitis. There were sufficient cases and high coverage of the vaccine to detect an excess of the magnitude observed in
the Phase 3 trial if it had occurred. Of the patients with probable or confirmed meningitis in vaccine-eligible age groups from
implementation areas, 41% (11/27) had received the RTS,S/AS01 vaccine, compared to 53% (2491/4672) of all other
hospital admissions in this age group from implementation areas (odds ratio, adjusted for country and age: 0.73 (95%CI
0.31,1.71). The PCR results showed that only 15% (8/55) of samples from confirmed cases, were of vaccine serotypes
preventable by Hib or pneumococcus vaccines (i.e. Haemophilus influenzae type b, or vaccine serotypes of Streptococcus
pneumoniae).. Baseline/comparator: .
69. Risk of Bias: no serious. Not downgraded for risk of bias despite being an open-label study because the findings from
the household survey suggest there is no evidence that the introduction of RTS,S/AS01 had a negative effect on uptake of
other childhood vaccines, ITN use, care-seeking behaviour, or health worker behaviour in testing and treating for febrile
illness. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Downgraded one level due to imprecision:
large CI that incorporates the possibility of benefit and harm. It was only downgraded by 1 level because the result excludes
an effect of the magnitude observed in the Phase 3 trial (RR = 4.5-10.5), after allowing for vaccine uptake levels in the pilot..
Publication bias: no serious.
70. Unplanned sub-group analysis of participant groups: 4-dose group received three doses of RTS,S/AS01 at months 0, 1,
and 2 and a booster dose at month 20; 3-dose group received three doses of RTS,S/AS01 and a dose of comparator vaccine
at month 20; Control group received a comparator vaccine at months 0, 1, 2, and 20 (control group).
71. [143]. In the context of an overall decrease in severe malaria, in an unplanned subgroup analysis from study months 0
to 20, 13 cases of possible cerebral malaria by record review and expert opinion occurred in the combined 3- and 4-dose
RTS,S/AS01 group compared to 7 in the control group (2:1 randomization). From study month 21 until trial end, there were
7 cerebral malaria cases in the 4-dose RTS,S/AS01 group, 8 cases in the 3-dose RTS,S/AS01 group, and 2 cases in the
control group. Baseline/comparator: .
72. Risk of Bias: very serious. Downgraded two levels for risk of bias: This was a post-hoc analysis based on an imprecise
algorithm, followed by record review and expert panel review. Cerebral malaria is a difficult diagnosis to make in real time,
and more difficult through record review Study was rated as unclear risk of bias due to heavy involvement of the funder in
the project; however, it has not been downgraded for risk of bias for this reason. Inconsistency: no serious. Indirectness: no
serious. Imprecision: serious. Downgraded one level due to imprecision: few events and large CI. Publication bias: no
serious.
73. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
SMC = combination group).
74. Primary study[145]. Due to the absence of cases in the reference group, it was not possible to calculate the incidence
rate ratio in vaccine recipients. There were no cases of cerebral malaria in the SMC alone group, 4 cases in the RTS,S vaccine
alone group (0.723 cases per 1000 PYAR; 95%CI 0.271 to 1.93), and 1 case in the combination of RTS,S vaccine + SMC
group (0.182 cases per 1000 PYAR; 95%CI 0.026 to 1.29).. Baseline/comparator: .
75. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Downgraded two levels due to
imprecision: very few events and 0 events in the control arm; . Publication bias: no serious.
76. Pilot implementation study designed to be analysed using cluster randomized control methodology; to be able to rule
out an association with cerebral malaria of the magnitude seen in the Phase 3 trial it would therefore be necessary to
373 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
exclude rate ratios of about 2.2 (1.6 allowing for 60% coverage and 5% contamination) or more. Across the three countries,
there was a total of 27,678 admissions to sentinel hospitals in children 1-59 months during the period from vaccine
introduction until 30 April 2021: 4,853 were vaccine-eligible based on their date of birth out of 13,918 total admissions in
areas where the vaccine was provided (implementing areas); 5,141 were vaccine-eligible out of 13,760 total admissions in
comparison areas
77. [155]. There were 55 cases of cerebral malaria, in whom lumbar puncture was performed to exclude cases with
probable meningitis): 25 from implementation areas and 30 from comparison areas. Among age groups of children not
eligible to receive the malaria vaccine, there were 241 cases of cerebral malaria, 115 from implementation areas and 126
from comparison areas. The incidence rate ratio comparing rates of admission to hospital with cerebral malaria in
implementation areas relative to comparison areas, among children eligible for the malaria vaccine, was 0.77 (95%CI 0.44 to
1.35). The incidence rate ratio for admission with other forms of severe malaria excluding cerebral malaria was 0.70 (95%CI
0.54 to 0.89). There was no evidence that effectiveness differed between cerebral malaria and other forms of severe malaria
(relative rate ratio 0.94 (95%CI 0.57 to 1.56; and test of interaction p-value: 0.808). When the analysis was broadened to
include cases meeting the criteria for cerebral malaria but in whom lumbar puncture was not performed, there was a total of
103 cases in age-groups eligible to have received at least one dose of the malaria vaccine: 49 from implementation areas and
54 from comparison areas. There were 455 cases in non-eligible age groups: 230 from implementing areas and 225 from
comparison areas. The incidence rate ratio comparing rates of admission to hospital with cerebral malaria (with the broader
case definition) in implementation areas relative to comparison areas, among children eligible for the malaria vaccine, was
0.96 (95%CI 0.61 to 1.52). Again there was no evidence that impact differed between cerebral malaria and other forms of
severe malaria (test of interaction p-value: 0.470). Similar results were obtained when cerebral malaria was limited to cases
defined as U (unresponsive) on the AVPU score. Among children eligible tohave received the vaccine, 20 of the cases from
implementation areas and 25 from comparison areas met this stricter criterion, and the estimate of the rate ratio was 0.66
(95%CI: 0.31 to 1.43). Of the patients with cerebral malaria in vaccine-eligible age groups from implementation areas, 47%
(23/49) had received RTS,S/AS01 vaccine, compared to 53% (2479/4650) of all other admissions in this age group from
implementation areas (odds ratio, adjusted for country and age,1.03, 95%CI 0.56,1.90; the odds ratio among cases meeting
the stricter definition requiring a lumbar puncture was 1.58; 95%CI: 0.66 to 3.80). There was therefore no evidence that
introduction of the malaria vaccine led to an increase in the incidence of hospital admission with cerebral malaria. The
incidence rate ratio excludes an effect of the magnitude observed in the Phase 3 trial (RR = 2.2), after allowing for uptake of
the vaccine in the pilot. Baseline/comparator: .
78. Risk of Bias: no serious. Not downgraded for risk of bias despite being an open-label study because the findings from
the household survey suggest there is no evidence that the introduction of RTS,S/AS01 had a negative effect on uptake of
other childhood vaccines, ITN use, care-seeking behaviour, or health worker behaviour in testing and treating for febrile
illness.. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Downgraded one level due to imprecision:
large CI that incorporates the possibility of benefit and harm. Study was powered for a pooled analysis only; country
estimates vary but CIs are wide and consistent with pooled effect; .
79. All-cause mortality (month 0 to study end) (modified ITT analysis); 4-dose group = three doses of RTS,S/AS01 at
months 0, 1, and 2 and a booster dose at month 20; 3-dose group = three doses of RTS,S/AS01 at months 0, 1, and 2 and a
comparator vaccine at month 20; Control group received = comparator vaccine at months 0, 1, 2, and 20.
80. [143]. Incidence rate ratio (IRR) of 4-dose group + 3-dose group vs Control group: Girls only IRR 2.0 (95% CI: 1.2 - 3.4)
vs Boys only IRR 0.8 (95% CI 0.5 - 1.2). Girls only: 4-dose group 35 deaths (9 malaria)/1467 girls + 3-dose group 32 deaths
(8 malaria) / 1500 girls vs Control group 17 deaths (4 malaria) / 1503 girls. Boys only 4-dose group 26 deaths (4 malaria) /
1509 boys + 3-dose group 19 deaths (9 malaria) / 1472 boys vs Control group 29 deaths (8 malaria) / 1471 boys. Baseline/
comparator: .
81. Risk of Bias: no serious. Study was rated as unclear risk of bias due to heavy involvement of the funder in the project;
however, it has not been downgraded for risk of bias as this was the only concern and the study was carefully scrutinized by
independent experts and considered well conducted.. Inconsistency: no serious. Indirectness: no serious. For this safety
outcome we have reported the combined results for children receiving 3 or 4 doses of the vaccine; however, it has not been
downgraded for indirectness. Imprecision: very serious. Downgraded two levels due to imprecision: few events and a very
large CI that incorporates the possibility of benefit and harm; . Publication bias: no serious.
82. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
SMC = combination group).
83. Primary study[145]. Gender interaction parameter 1.80 (95%CI: 0.56 to 5.79); Girls only RTS,S vs SMC alone hazard
ratio (HR) 1.23 (95% CI: 0.51 to 2.96); there were 11 deaths total or 4.15 deaths per 1000 PYAR (95% CI 2.30 to 7.49)
among girls in the RTS,S alone group compared to 9 deaths total or 3.42 deaths per 1000 PYAR (95% CI 1.78 to 6.57) among
girls in the SMC alone group. Boys only RTS,S vs SMC alone HR 0.68 (95% CI 0.32 to 1.47); there were 11 deaths total or
3.82 deaths per 1000 PYAR (95% CI 2.11 to 6.89) among boys in the RTS,S alone group compared to 16 deaths total or 5.68
374 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
deaths per 1000 PYAR (95% CI 3.48 to 9.27) among boys in the SMC alone group. Baseline/comparator: .
84. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Downgraded two levels due to
imprecision: few events and a very large CI that incorporates the possibility of benefit and harm; . Publication bias: no
serious.
85. Randomly assigned children 5 to 17 months of age to receive sulfadoxine–pyrimethamine and amodiaquine (SMC =
chemoprevention-alone group), RTS,S/AS01E (RTS,S = vaccine-alone group), or chemoprevention and RTS,S/AS01E (RTS,S +
SMC = combination group).
86. Primary study[145]. Gender interaction parameter 0.35 (95%CI 0.06 to 1.98). Girls only RTS,S+SMC combination group
vs SMC alone group hazard ratio (HR) 0.22 (95% CI 0.05 to 1.02); there were 2 deaths total or 0.75 deaths per 1000 PYAR
(95% CI 0.19 - 3.01) among girls in the RTS,S + SMC combination group compared to 9 deaths total or 3.42 deaths per 1000
PYAR (95% CI 1.78 - 6.57) among girls in the SMC alone group. Boys only RTS,S + SMC combination group vs SMC alone
group HR 0.62 (95% CI 0.28 to 1.37); there were 10 deaths total or 3.51 deaths per 1000 PYAR (95% CI 1.89 - 6.52) among
boys in the Combination group compared to 16 deaths total or 5.68 deaths per 1000 PYAR (95% CI 3.48 - 9.27) among boys
in the SMC alone group.. Baseline/comparator: .
87. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Downgraded two levels due to
imprecision: few events and a very large CI that incorporates the possibility of benefit and harm;. Publication bias: no
serious.
88. Pilot implementation study designed to be analysed using cluster randomized control methodology. The evaluation was
not powered at this time point to assess the overall impact of vaccine introduction on mortality but the evaluation was well
powered to detect gender imbalance in all-cause mortality of the magnitude observed in the Phase 3 trial (mortality ratio =
1.4--1.6), in children up to about 2 years of age. A total of 13682 deaths among children 1-59 months of age were reported
via community-based mortality surveillance across the three countries from the start of vaccinations on 23 April 2019 to 31
March 2021 (deaths in April 2021 were excluded because verbal autopsies have not all been completed).
89. [155]. There was no evidence that the effect of RTS,S/AS01 introduction on all-cause mortality differed between girls
and boys in this age group. Excluding deaths due to injury in children eligible to have received three doses of RTS,S/AS01,
there was a total of 2864 deaths reported, 1421 from implementing regions and 1443 from comparison regions. In children
who were not eligible to have received the vaccine there were 4218 deaths in implementing regions and 3874 in comparison
regions. The mortality ratio in the vaccine-eligible age group (eligible for three doses) between implementing and comparison
regions, was 0.93 (95%CI: 0.84 to 1.03), a 7% reduction (95%CI: -3% to 16%). There was no evidence that the mortality ratio
differed between girls and boys, the p-value for this interaction was 0.343. The mortality ratio in girls was 0.98 and in boys
0.90; the relative mortality ratio (girls:boys) was 1.08 (95%CI: 0.92 to 1.28). When analysis was extended to children eligible
to have received at least one dose of the vaccine, similar results were obtained (ratio of mortality ratios: 1.08; 95%CI: 0.93
to 1.25; p-value for the interaction: 0.321). Similar results were also obtained when the analysis was repeated for different
age groups of eligible children (mortality ratio girls:boys in eligible children under 18 months of age was 1.10 [95%CI: 0.94 to
1.29], and in eligible children aged 18 months and over it was 0.95 [95%CI: 0.70 to 1.31]). The vaccination status of vaccine-
eligible children who died in implementation areas was similar in girls and boys (58.9% and 57.0% respectively). According to
the household surveys in 12-23 month olds, coverage of the first dose of RTS,S/AS01 was slightly higher in girls than in boys
(77.6% in girls and 73.0% in boys in Ghana; 75.1% in girls and 70.1% in boys in Malawi; and 79.0% in girls and 78.2% in boys
in Kenya). Coverage was similar for the third dose.. Baseline/comparator: .
90. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Downgraded one level because the evaluation
was not powered at this time point to assess overall impact of vaccine introduction on mortality. However the evaluation
was well powered to detect gender imbalance in all-cause mortality of the magnitude observed in the Phase 3 trial (mortality
ratio = 1.4 - 1.6), in children up to about 2 years of age.. Publication bias: no serious.
Attached Images
References
121. Seasonal malaria chemoprevention with sulfadoxine-pyrimethamine plus amodiaquine in children: A field guide.
Geneva: World Health Organization 2013; Website
143. Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa:
final results of a phase 3, individually randomised, controlled trial. Lancet (London, England) 2015;386(9988):31-45
Pubmed Journal
145. Chandramohan D, Zongo I, Sagara I, Cairns M, Yerbanga R-S, Diarra M, et al. : Seasonal Malaria Vaccination with or
without Seasonal Malaria Chemoprevention. The New England journal of medicine 2021;385(11):1005-1017 Pubmed
375 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Journal
155. Milligan P, Moore K : Statistical report on the results of the RTS,S/AS01 Malaria Vaccine Pilot Evaluation 24 months
after the vaccine was introduced (unpublished evidence). 2021;V1.3 6 Sept 2021 Website
5. CASE MANAGEMENT
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Artemether +
Timeframe measurements sinin + (Quality of summary
lumefantrine
piperaquine evidence)
Treatment 230 78
Relative risk 0.34 per 1000 per 1000
failure - PCR (CI 95% 0.3 — 0.39)
unadjusted 1 Based on data from High
28 days Difference: 152 fewer per
6,200 participants in 9 2
1000
studies. (Randomized ( CI 95% 161
controlled) fewer — 140
fewer )
Treatment 30 13
Relative risk 0.42 per 1000 per 1000
failure - PCR (CI 95% 0.29 — 0.62)
adjusted 3 Based on data from High
28 days Difference: 17 fewer per
5,417 participants in 9 4
1000
studies. (Randomized ( CI 95% 21
controlled) fewer — 11
fewer )
376 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Artemether +
Timeframe measurements sinin + (Quality of summary
lumefantrine
piperaquine evidence)
Treatment
Relative risk 0.72 60 43
failure - PCR (CI 95% 0.5 — 1.04) per 1000 per 1000
adjusted 7 Based on data from High
63 days 2,097 participants in 2 Difference: 17 fewer per 8
1. PCR unadjusted
2. Risk of Bias: no serious. Trials generally have little risk of bias. Exclusion of studies with high or unclear risk for
selection bias or detection bias did not change the result.. Inconsistency: no serious. All the trials had similar results,
and statistical heterogeneity was low.. Indirectness: no serious. The trials were conducted in different transmission
settings in east, west and southern Africa. Most studies were limited to children.. Imprecision: no serious. The 95%
CI implies appreciable benefit, and the meta-analysis is adequately powered to detect this result.. Publication bias:
no serious.
3. PCR adjusted
4. Risk of Bias: no serious. Trials generally have little risk of bias. Exclusion of studies with high or unclear risk for
selection bias or detection bias did not change the result.. Inconsistency: no serious. All the trials had similar results,
and statistical heterogeneity was low.. Indirectness: no serious. The trials were conducted in different transmission
settings in east, west and southern Africa. Most studies were limited to children.. Imprecision: no serious. Although
there is a benefit in favour of dihydroartemisinin + piperaquine, the PCR-adjusted treatment failure rate was < 5%
with both drugs.. Publication bias: no serious.
5. PCR unadjusted
6. Risk of Bias: no serious. Trials generally have little risk of bias. Exclusion of studies with high or unclear risk for
selection bias or detection bias did not change the result.. Inconsistency: no serious. At this time, there is
inconsistency between trials; both show a benefit with dihydroartemisinin + piperaquine, but the size of the benefit
differs.. Indirectness: no serious. The trials were conducted in different transmission settings in east, west and
southern Africa. Most studies were limited to children.. Imprecision: no serious. The 95% CI implies appreciable
benefit, and the meta-analysis is adequately powered to detect this result.. Publication bias: no serious.
7. PCR adjusted
8. Risk of Bias: no serious. Trials generally have little risk of bias. Exclusion of studies with high or unclear risk for
selection bias or detection bias did not change the result.. Inconsistency: no serious. The treatment failure rate with
dihydroartemisinin + piperaquine was < 5% in both trials.. Indirectness: no serious. The trials were conducted in
different transmission settings in east, west and southern Africa. Most studies were limited to children.. Imprecision:
no serious. Both ACTs performed well in these two trials, with low rates of treatment failure.. Publication bias: no
serious.
Attached Images
377 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Artesunate +
Timeframe measurements sinin + (Quality of summary
mefloquine
piperaquine evidence)
Treatment 20 20
Relative risk 1.02 per 1000 per 1000
failure - PCR (CI 95% 0.28 — 3.72)
unadjusted 1 High
Based on data from
28 days Difference: 0 fewer per Due to serious
3,487 participants in 8
1000 inconsistency 2
studies. (Randomized ( CI 95% 14
controlled) fewer — 54 more
)
Treatment
Relative risk 0.41 10 4
failure - PCR (CI 95% 0.21 — 0.8) per 1000 per 1000
adjusted 3 High
Based on data from
28 days Difference: 6 fewer per Due to serious
3,467 participants in 8
1000 inconsistency 4
studies. (Randomized
controlled) ( CI 95% 8 fewer
— 2 fewer )
Treatment
Relative risk 0.84 120 101
failure - PCR (CI 95% 0.69 — 1.03) per 1000 per 1000
unadjusted 5 Moderate
Based on data from
63 days Difference: 19 fewer per Due to serious
2,715 participants in 5
1000 inconsistency 6
studies. (Randomized
controlled) ( CI 95% 37
fewer — 4 more )
Treatment 30 15
Relative risk 0.5 per 1000 per 1000
failure - PCR (CI 95% 0.3 — 0.84)
adjusted 7 High
Based on data from
63 days Difference: 15 fewer per Due to serious
2,500 participants in 5
1000 inconsistency 8
studies. (Randomized ( CI 95% 21
controlled) fewer — 5 fewer
)
1. PCR unadjusted
2. Risk of Bias: no serious. Trials generally have little risk of selection or detection bias. Exclusion of trials with high
or unclear risk of bias did not change the result.. Inconsistency: serious. In six trials, very few recurrences of
parasitaemia were found in both groups. Two trials conducted mainly in areas in Thailand with multi-drug resistance
showed increased risks for recurrent parasitaemia with artesunate + mefloquine.. Indirectness: no serious. The trials
were conducted in adults and children in Cambodia, India, the Lao People’s Democratic Republic, Myanmar, Thailand
and Viet Nam.. Imprecision: no serious. Overall, no significant difference between treatments; however,
dihydroartemisinin + piperaquine may be superior where P. falciparum is resistant to mefloquine.. Publication bias:
no serious.
3. PCR adjusted
4. Risk of Bias: no serious. Trials generally have little risk of selection or detection bias. Exclusion of trials with high
or unclear risk of bias did not change the result.. Inconsistency: serious. In six trials, very few recurrences of
parasitaemia were found in both groups. Two trials conducted mainly in areas in Thailand with multi-drug resistance
showed increased risks for recurrent parasitaemia with artesunate + mefloquine.. Indirectness: no serious. The trials
were conducted in adults and children in Cambodia, India, the Lao People’s Democratic Republic, Myanmar, Thailand
and Viet Nam.. Imprecision: no serious. Overall, a statistically significant benefit with dihydroartemisinin +
piperaquine, although the benefit may be present only where there is resistance to mefloquine.. Publication bias: no
378 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
serious.
5. PCR unadjusted
6. Risk of Bias: no serious. Trials generally have little risk of selection or detection bias. Exclusion of trials with high
or unclear risk of bias did not change the result.. Inconsistency: serious. Of the five trials, one in Thailand in 2005
showed a statistically significant benefit with dihydroartemisinin + piperaquine, one in Myanmar in 2009 showed a
benefit with dihydroartemisinin + piperaquine, and three found no difference.. Indirectness: no serious. The trials
were conducted in adults and children in Cambodia, India, the Lao People’s Democratic Republic, Myanmar and
Thailand.. Imprecision: no serious. Overall, no significant difference between treatments. Although some trials found
statistically significant differences, these may not be clinically important.. Publication bias: no serious.
7. PCR adjusted
8. Risk of Bias: no serious. Trials generally have little risk of selection or detection bias. Exclusion of trials with high
or unclear risk of bias did not change the result.. Inconsistency: serious. Slight variation among trials, only one
showing a statistically significant benefit with dihydroartemisinin + piperaquine.. Indirectness: no serious. The trials
were conducted in adults and children in Cambodia, India, the Lao People’s Democratic Republic, Myanmar and
Thailand.. Imprecision: no serious. Overall, no significant difference between treatments. Although some trials found
statistically significant differences, these may not be clinically important.. Publication bias: no serious.
Attached Images
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Artemether +
Timeframe measurements sinin + (Quality of summary
lumefantrine
piperaquine evidence)
Serious
adverse events
6 10
per 1000 per 1000 Moderate
(including Based on data from
deaths) Due to serious
7,022 participants in 8 Difference: 4 more per 1000
imprecision 1
studies. (Randomized
controlled)
Early vomiting
20 30
per 1000 per 1000 Moderate
Based on data from
Due to serious
2,695 participants in 3
Difference: 10 more per risk of bias 2
studies. (Randomized
controlled) 1000
Vomiting
90 90
per 1000 per 1000 Moderate
Based on data from
Due to serious
6,761 participants in 9
Difference: 0 fewer per risk of bias 3
studies. (Randomized
controlled) 1000
379 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Artemether +
Timeframe measurements sinin + (Quality of summary
lumefantrine
piperaquine evidence)
Nausea
20 20 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
547 participants in 2
Difference: 0 fewer per serious
studies. (Randomized
1000 imprecision 4
controlled)
Diarrhoea
120 120
per 1000 per 1000 Moderate
Based on data from
Due to serious
4,889 participants in 7
Difference: 0 fewer per risk of bias 5
studies. (Randomized
controlled) 1000
Anorexia
150 140
per 1000 per 1000 Moderate
Based on data from
Due to serious
3,834 participants in 5
Difference: 10 fewer per risk of bias 7
studies. (Randomized
controlled) 1000
Headache
270 330 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
309 participants in 2
Difference: 60 more per serious
studies. (Randomized
1000 imprecision 8
controlled)
Sleeplessness
10 30 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
547 participants in 2
Difference: 20 more per serious
studies. (Randomized
1000 imprecision 9
controlled)
Dizziness
30 40 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
547 participants in 2
Difference: 10 more per serious
studies. (Randomized
1000 imprecision 10
controlled)
Sleepiness
0 0 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
384 participants in 1
Difference: 0 fewer per serious
studies. (Randomized
1000 imprecision 11
controlled)
380 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Artemether +
Timeframe measurements sinin + (Quality of summary
lumefantrine
piperaquine evidence)
Weakness
170 180
per 1000 per 1000 Moderate
Based on data from
Due to serious
1,812 participants in 5
Difference: 10 more per risk of bias 12
studies. (Randomized
controlled) 1000
Cough
420 420
per 1000 per 1000 Moderate
Based on data from
Due to serious
4,342 participants in 5
Difference: 0 fewer per risk of bias 13
studies. (Randomized
controlled) 1000
Coryza
680 660
per 1000 per 1000 Low
Based on data from
Due to serious
832 participants in 2
Difference: 20 fewer per imprecision 14
studies. (Randomized
controlled) 1000
Prolonged QT
interval
30 20 Low
per 1000 per 1000
(adverse event) Due to serious
Based on data from
imprecision and
1,548 participants in 1 Difference: 10 fewer per serious risk of
studies. (Randomized 1000
bias 15
controlled)
Prolonged QT
interval (Bazett
70 90 Low
per 1000 per 1000 Due to serious
correction) Based on data from
imprecision and
1,548 participants in 1
Difference: 20 more per serious risk of
studies. (Randomized
1000 bias 16
controlled)
Prolonged QT
interval
0 0 Low
per 1000 per 1000
(Fridericia Due to serious
Based on data from
correction) risk of bias and
1,548 participants in 1 Difference: 0 fewer per serious
studies. (Randomized 1000
imprecision 17
controlled)
Pruritus
20 40
per 1000 per 1000 Moderate
Based on data from
Due to serious
2,033 participants in 5
Difference: 20 more per risk of bias 18
studies. (Randomized
controlled) 1000
0 0 Low
Facial oedema Based on data from per 1000 per 1000 Due to serious
risk of bias and
384 participants in 1
381 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Artemether +
Timeframe measurements sinin + (Quality of summary
lumefantrine
piperaquine evidence)
1. Risk of Bias: no serious. All but one of the trials were open label; however, we did not downgrade for this
outcome.. Inconsistency: no serious. The finding is consistent across all trials. Statistical heterogeneity is low..
Indirectness: no serious. The trials were conducted mainly in children in Africa; few trials in Asia or in adults..
Imprecision: serious. No statistically significant difference was detected between treatments; however the sample
size does not exclude the possibility of rare but clinically important differences..
2. Risk of Bias: serious. The majority of trials were open label.. Inconsistency: no serious. The finding is consistent
across all trials. Statistical heterogeneity is low.. Indirectness: no serious. The trials were conducted mainly in
children in Africa; few trials in Asia or in adults.. Imprecision: no serious. No effect found, and the CIs around the
absolute effects exclude clinically important differences..
3. Risk of Bias: serious. The majority of trials were open label.. Inconsistency: no serious. The finding is consistent
across all trials. Statistical heterogeneity is low.. Indirectness: no serious. The trials were conducted mainly in
children in Africa; few trials in Asia or in adults.. Imprecision: no serious. No effect found, and the CIs around the
absolute effects exclude clinically important differences..
4. Risk of Bias: serious. Downgraded by 1 for risk of bias: The majority of trials were open label.. Inconsistency: no
serious. No serious inconsistency: The finding is consistent across all trials. Statistical heterogeneity is low..
Indirectness: no serious. No serious indirectness: The trials were conducted mainly in children in Africa; few trials in
Asia or in adults.. Imprecision: serious. Downgraded by 1 for serious imprecision: There are limited data..
5. Risk of Bias: serious. The majority of trials were open label.. Inconsistency: no serious. The finding is consistent
across all trials. Statistical heterogeneity is low.. Indirectness: no serious. The trials were conducted mainly in
children in Africa; few trials in Asia or in adults.. Imprecision: no serious. No effect found, and the CIs around the
absolute effects exclude clinically important differences..
6. Risk of Bias: serious. The majority of trials were open label.. Inconsistency: no serious. The finding is consistent
across all trials. Statistical heterogeneity is low.. Indirectness: no serious. The trials were conducted mainly in
children in Africa; few trials in Asia or in adults.. Imprecision: serious. The result does not reach statistical
significance..
7. Risk of Bias: serious. The majority of trials were open label.. Inconsistency: no serious. The finding is consistent
across all trials. Statistical heterogeneity is low.. Indirectness: no serious. The trials were conducted mainly in
children in Africa; few trials in Asia or in adults.. Imprecision: no serious. No effect found, and the CIs around the
absolute effects exclude clinically important differences..
8. Risk of Bias: serious. The majority of trials were open label.. Inconsistency: no serious. The finding is consistent
across all trials. Statistical heterogeneity is low.. Indirectness: no serious. The trials were conducted mainly in
children in Africa; few trials in Asia or in adults.. Imprecision: serious. The result does not reach statistical
significance..
9. Risk of Bias: serious. The majority of trials were open label.. Inconsistency: no serious. The finding is consistent
across all trials. Statistical heterogeneity is low.. Indirectness: no serious. The trials were conducted mainly in
children in Africa; few trials in Asia or in adults.. Imprecision: serious. There are limited data..
10. Risk of Bias: serious. The majority of trials were open label.. Indirectness: no serious. The trials were conducted
mainly in children in Africa; few trials in Asia or in adults.. Imprecision: serious. There are limited data..
11. Risk of Bias: serious. The majority of trials were open label.. Inconsistency: no serious. The finding is consistent
across all trials. Statistical heterogeneity is low.. Indirectness: no serious. The trials were conducted mainly in
children in Africa; few trials in Asia or in adults.. Imprecision: serious. There are limited data..
12. Risk of Bias: serious. The majority of trials were open label.. Inconsistency: no serious. The finding is consistent
across all trials. Statistical heterogeneity is low.. Indirectness: no serious. The trials were conducted mainly in
children in Africa; few trials in Asia or in adults.. Imprecision: no serious. No effect found, and the CIs around the
absolute effects exclude clinically important differences..
13. Risk of Bias: serious. The majority of trials were open label.. Inconsistency: no serious. The finding is consistent
across all trials. Statistical heterogeneity is low.. Indirectness: no serious. The trials were conducted mainly in
children in Africa; few trials in Asia or in adults.. Imprecision: no serious. No effect found, and the CIs around the
382 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Attached Images
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Artesunate +
Timeframe measurements sinin + (Quality of summary
mefloquine
piperaquine evidence)
Serious
adverse events
8 9
per 1000 per 1000 Moderate
(including Based on data from
deaths) Due to serious
3,522 participants in 8 Difference: 1 more per 1000
imprecision 1
studies. (Randomized
controlled)
Nausea
20 14
per 1000 per 1000 Moderate
Based on data from
Due to serious
4,531 participants in 9
Difference: 6 fewer per risk of bias 2
studies. (Randomized
controlled) 1000
383 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Artesunate +
Timeframe measurements sinin + (Quality of summary
mefloquine
piperaquine evidence)
Early vomiting
7 6
per 1000 per 1000 Moderate
Based on data from
Due to serious
4,114 participants in 9
Difference: 1 fewer per risk of bias 3
studies. (Randomized
controlled) 1000
Vomiting
13 8
per 1000 per 1000 Moderate
Based on data from
Due to serious
2,744 participants in 5
Difference: 5 fewer per risk of bias 4
studies. (Randomized
controlled) 1000
Anorexia
15 13 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
3,497 participants in 6
Difference: 2 fewer per serious
studies. (Randomized
1000 imprecision 5
controlled)
Diarrhoea
6 8
per 1000 per 1000 Moderate
Based on data from
Due to serious
2,217 participants in 5
Difference: 2 more per 1000 risk of bias 6
studies. (Randomized
controlled)
Abdominal pain 11 11
per 1000 per 1000 Moderate
Based on data from
Due to serious
3,887 participants in 7
Difference: 0 fewer per risk of bias 7
studies. (Randomized
controlled) 1000
Headache
12 10 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
2,039 participants in 4
Difference: 2 fewer per serious
studies. (Randomized
1000 inconsistency 8
controlled)
Dizziness
36 26
per 1000 per 1000 Moderate
Based on data from
Due to serious
4,531 participants in 9
Difference: 10 fewer per risk of bias 9
studies. (Randomized
controlled) 1000
Sleeplessness
21 10
per 1000 per 1000 Moderate
Based on data from
Due to serious
2,551 participants in 6
Difference: 11 fewer per risk of bias 10
studies. (Randomized
controlled) 1000
384 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Artesunate +
Timeframe measurements sinin + (Quality of summary
mefloquine
piperaquine evidence)
Fatigue
8 3 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
872 participants in 2
Difference: 5 fewer per serious
studies. (Randomized
1000 indirectness 11
controlled)
Nightmares
10 1 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
220 participants in 1
Difference: 9 fewer per serious
studies. (Randomized
1000 indirectness 12
controlled)
Anxiety
11 1 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
522 participants in 1
Difference: 10 fewer per serious
studies. (Randomized
1000 indirectness 13
controlled)
Blurred vision
9 4 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
464 participants in 1
Difference: 5 fewer per serious
studies. (Randomized
1000 indirectness 14
controlled)
Tinnitus
9 4 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
220 participants in 1
Difference: 5 fewer per serious
studies. (Randomized
1000 indirectness 15
controlled)
Palpitations
18 11
per 1000 per 1000 Moderate
Based on data from
Due to serious
1,175 participants in 3
Difference: 7 fewer per risk of bias 16
studies. (Randomized
controlled) 1000
Cough
10 8 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
1,148 participants in 1
Difference: 2 fewer per serious
studies. (Randomized
1000 imprecision 17
controlled)
Dyspnoea
9 3 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
220 participants in 1
Difference: 6 fewer per serious
studies. (Randomized
1000 imprecision 18
controlled)
385 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Artesunate +
Timeframe measurements sinin + (Quality of summary
mefloquine
piperaquine evidence)
Prolonged QT
interval
4 5 Low
per 1000 per 1000
(adverse event) Due to serious
Based on data from
risk of bias and
1,148 participants in 1 Difference: 1 more per 1000 serious
studies. (Randomized
imprecision 19
controlled)
Prolonged QT
interval (Bazett
4 9 Low
per 1000 per 1000 Due to serious
correction) Based on data from
risk of bias and
1,148 participants in 1
Difference: 5 more per 1000 serious
studies. (Randomized
imprecision 20
controlled)
Prolonged QT
interval
5 4 Low
per 1000 per 1000
(Fridericia Due to serious
Based on data from
correction) risk of bias and
1,148 participants in 1 Difference: 1 fewer per serious
studies. (Randomized 1000
imprecision 21
controlled)
Arthralgia
6 5
per 1000 per 1000 Moderate
Based on data from
Due to serious
1,148 participants in 1
Difference: 1 fewer per risk of bias 22
studies. (Randomized
controlled) 1000
Myalgia
6 6
per 1000 per 1000 Moderate
Based on data from
Due to serious
1,148 participants in 1
Difference: 0 fewer per risk of bias 23
studies. (Randomized
controlled) 1000
Urticaria
2 1 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
719 participants in 2
Difference: 1 fewer per serious
studies. (Randomized
1000 imprecision 24
controlled)
Pruritus
3 2 Low
per 1000 per 1000 Due to serious
Based on data from
risk of bias and
872 participants in 2
Difference: 1 fewer per serious
studies. (Randomized
1000 imprecision 25
controlled)
1 0 Low
Rash Based on data from per 1000 per 1000 Due to serious
risk of bias and
220 participants in 1
386 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Artesunate +
Timeframe measurements sinin + (Quality of summary
mefloquine
piperaquine evidence)
1. Risk of Bias: no serious. Only eight of the 11 reports made any comment on serious adverse events. None of
these eight trials was blinded. . Inconsistency: no serious. None of the eight trials found statistically significant
differences.. Indirectness: no serious. These trials included both adults and children and were conducted in Asia and
South America.. Imprecision: serious. These trials do not exclude the possibility of rare but clinically important
adverse effects..
2. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. This finding was consistent across
trials, with no significant statistical heterogeneity.. Indirectness: no serious. These trials included both adults and
children and were conducted in Asia and South America.. Imprecision: no serious. The result is statistically
significant, and the meta-analysis has adequate power to detect this effect..
3. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. None of the eight trials found
statistically significant differences.. Indirectness: no serious. These trials included both adults and children and were
conducted in Asia and South America.. Imprecision: no serious. The 95% CI around the absolute effect is narrow and
excludes clinically important differences..
4. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. This finding was consistent across
trials, with no significant statistical heterogeneity.. Indirectness: no serious. These trials included both adults and
children and were conducted in Asia and South America.. Imprecision: no serious. The result is statistically
significant, and the meta-analysis has adequate power to detect this effect..
5. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. This finding was consistent across
trials, with no significant statistical heterogeneity.. Indirectness: no serious. These trials included both adults and
children and were conducted in Asia and South America.. Imprecision: serious. This result does not reach statistical
significance..
6. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. This finding was consistent across
trials, with no significant statistical heterogeneity.. Indirectness: no serious. These trials included both adults and
children and were conducted in Asia and South America.. Imprecision: no serious. The result is statistically
significant, and the meta-analysis has adequate power to detect this effect..
7. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. This finding was consistent across
trials, with no significant statistical heterogeneity.. Indirectness: no serious. These trials included both adults and
children and were conducted in Asia and South America.. Imprecision: no serious. No difference was found between
treatments, and the sample is large enough for detection of any differences..
8. Risk of Bias: serious. All trials were open label.. Inconsistency: serious. There is moderate heterogeneity among
trials.. Indirectness: no serious. These trials included both adults and children and were conducted in Asia and South
America.. Imprecision: no serious. The result is statistically significant, and the meta-analysis has adequate power to
detect this effect..
9. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. This finding was consistent across
trials, with no significant statistical heterogeneity.. Indirectness: no serious. These trials included both adults and
children and were conducted in Asia and South America.. Imprecision: no serious. The result is statistically
significant, and the meta-analysis has adequate power to detect this effect..
10. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. This finding was consistent across
trials, with no significant statistical heterogeneity.. Indirectness: no serious. These trials included both adults and
children and were conducted in Asia and South America.. Imprecision: no serious. The result is statistically
significant, and the meta-analysis has adequate power to detect this effect..
11. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. This finding was consistent across
trials, with no significant statistical heterogeneity.. Indirectness: serious. Only two trials assessed this outcome..
Imprecision: no serious.
12. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. Indirectness: serious. Only two trials
assessed this outcome.. Imprecision: no serious.
13. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. Indirectness: serious. Only two trials
assessed this outcome.. Imprecision: no serious.
387 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
14. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. Indirectness: serious. Only two trials
assessed this outcome.. Imprecision: no serious.
15. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. Indirectness: serious. Only two trials
assessed this outcome.. Imprecision: no serious.
16. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. This finding was consistent across
trials, with no significant statistical heterogeneity.. Indirectness: no serious. These trials included both adults and
children and were conducted in Asia and South America.. Imprecision: no serious. The result is statistically
significant, and the meta-analysis has adequate power to detect this effect..
17. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. Indirectness: no serious. Imprecision:
serious. This result does not reach statistical significance..
18. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. Indirectness: no serious. Imprecision:
serious. Limited data available, and the result is not statistically significant..
19. Risk of Bias: serious. This trial is unblinded. Only a few of the recorded prolonged QT intervals were registered
as adverse events, which removed the statistical significance. The reasons for this are unclear.. Inconsistency: no
serious. Indirectness: no serious. This single large trial was conducted in adults and children in India, the Lao People’s
Democratic Republic and Thailand.. Imprecision: serious. This result does not reach statistical significance..
20. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. Indirectness: no serious. This single
large trial was conducted in adults and children in India, the Lao People’s Democratic Republic and Thailand..
Imprecision: serious. This result does not reach statistical significance..
21. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. Indirectness: no serious. This single
large trial was conducted in adults and children in India, the Lao People’s Democratic Republic and Thailand..
Imprecision: serious. This result does not reach statistical significance..
22. Risk of Bias: serious. All trials were open label. This trial is unblinded. Only a few of the recorded prolonged QT
intervals were registered as adverse events, which removed the statistical significance. The reasons for this are
unclear. 15 . Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. No difference was found
between treatments, and the sample is large enough for detection of any differences..
23. Risk of Bias: serious. All trials were open label. This trial is unblinded. Only a few of the recorded prolonged QT
intervals were registered as adverse events, which removed the statistical significance. The reasons for this are
unclear.. Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. No difference was found
between treatments, and the sample is large enough for detection of any differences..
24. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. Indirectness: no serious. Imprecision:
serious. Limited data available, and the result is not statistically significant..
25. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. Indirectness: no serious. Imprecision:
serious. Limited data available, and the result is not statistically significant..
26. Risk of Bias: serious. All trials were open label.. Inconsistency: no serious. Indirectness: no serious. Imprecision:
serious. Limited data available, and the result is not statistically significant..
Attached Images
Certainty of
Comparator Intervention
Outcome Study results and the Evidence Plain language
Artemether + Artemisinin +
Timeframe measurements (Quality of summary
lumefantrine naphthoquine
evidence)
388 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Comparator Intervention
Outcome Study results and the Evidence Plain language
Artemether + Artemisinin +
Timeframe measurements (Quality of summary
lumefantrine naphthoquine
evidence)
Treatment
Relative risk 3.25 0 0
failure on day per 1000 per 1000 Very low
(CI 95% 0.13 — 78.69)
28 (PCR- Due to serious
Based on data from
adjusted) Difference: 0 fewer per indirectness and
295 participants in 2
1000 very serious
studies. (Randomized
( CI 95% 0 fewer imprecision 2
controlled)
— 0 fewer )
Parasite 20 3
Relative risk 0.15 per 1000 per 1000
clearance: Very low
(CI 95% 0.01 — 2.92)
parasitaemia Due to serious
Based on data from
on day 2 Difference: 17 fewer per indirectness and
297 participants in 2
1000 very serious
studies. (Randomized ( CI 95% 20 imprecision 4
controlled) fewer — 38 more
)
20 39
Relative risk 1.97 per 1000 per 1000
Gametocytaem Very low
(CI 95% 0.18 — 21.14)
ia on day 7 Due to serious
Based on data from
Difference: 19 more per indirectness and
123 participants in 1
1000 very serious
studies. (Randomized ( CI 95% 16 imprecision 5
controlled) fewer — 403
more )
1. Risk of Bias: no serious. One study adequately concealed allocation and thus had a low risk of selection bias. In
the other study, the process of randomization and allocation concealment was unclear. Inconsistency: no serious.
Statistical heterogeneity was low. Indirectness: serious. Only two studies, in Benin and Cote d’Ivoire, evaluated this
comparison. Further studies in additional settings are required before this result can be generalized. Imprecision:
very serious. Demonstration of non-inferiority at 95% efficacy would require a sample size of 472. Both trials are
significantly underpowered.
2. Risk of Bias: no serious. One study adequately concealed allocation and thus had a low risk of selection bias. In
the other study, the process of randomization and allocation concealment was unclear. Inconsistency: no serious.
Statistical heterogeneity was low. Indirectness: serious. Only two studies, in Benin and Cote d’Ivoire, evaluated this
comparison. Further studies in additional settings are required before this result can be generalized. Imprecision:
very serious. Demonstration of non-inferiority at 95% efficacy would require a sample size of 472. Both trials are
significantly underpowered.
389 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
3. Risk of Bias: no serious. This study adequately concealed allocation and thus had a low risk of selection bias.
Indirectness: serious. Study in Cote d’Ivoire. Further studies in additional settings are required before this result can
be generalized. Imprecision: very serious. This trial was small and the result has a very wide 95% confidence interval,
including appreciable benefit and harm.
4. Risk of Bias: no serious. One study adequately concealed allocation and thus had a low risk of selection bias. In
the other study, the process of randomization and allocation concealment was unclear. Inconsistency: no serious.
Statistical heterogeneity was low. Indirectness: serious. Only two studies, in Benin and Cote d’Ivoire, evaluated this
comparison. Further studies in additional settings are required before this result can be generalized. Imprecision:
very serious. The result has a very wide 95% confidence interval, including appreciable benefit and harm.
5. Risk of Bias: no serious. This study adequately concealed allocation and thus had a low risk of selection bias.
Indirectness: serious. Study in Cote d’Ivoire. Further studies in additional settings are required before this result can
be generalized. Imprecision: very serious. This trial was small and the result has a very wide 95% confidence interval,
including appreciable benefit and harm.
Attached Images
Comparator Certainty of
Intervention
Outcome Study results and Dihydroartemi the Evidence Plain language
Artemisinin +
Timeframe measurements sinin + (Quality of summary
naphthoquine
piperaquine evidence)
Treatment
failure on day
0 0 Very low
per 1000 per 1000
28 (PCR- Due to serious
Based on data from
unadjusted) indirectness and
143 participants in 1
very serious
studies. (Randomized
imprecision 1
controlled)
Treatment
failure on day
0 0 Very low
per 1000 per 1000
28 (PCR- Due to serious
Based on data from
adjusted) indirectness and
143 participants in 1
very serious
studies. (Randomized
imprecision 2
controlled)
Treatment 30 27
Relative risk 0.91 per 1000 per 1000
failure on day Very low
(CI 95% 0.13 — 6.26)
42 (PCR- Due to serious
Based on data from
unadjusted) Difference: 3 fewer per indirectness and
143 participants in 1
1000 very serious
studies. (Randomized ( CI 95% 26 imprecision 3
controlled) fewer — 158
more )
390 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator Certainty of
Intervention
Outcome Study results and Dihydroartemi the Evidence Plain language
Artemisinin +
Timeframe measurements sinin + (Quality of summary
naphthoquine
piperaquine evidence)
Treatment 30 6
Relative risk 0.19 per 1000 per 1000
failure on day Very low
(CI 95% 0.01 — 3.82)
42 (PCR- Due to serious
Based on data from
adjusted) Difference: 24 fewer per indirectness and
141 participants in 1
1000 very serious
studies. (Randomized ( CI 95% 30 imprecision 4
controlled) fewer — 85 more
)
Fever
clearance:
0 0 Very low
per 1000 per 1000 Due to serious
fever on day 2 Based on data from
indirectness and
144 participants in 1
very serious
studies. (Randomized
imprecision 5
controlled)
Parasite
clearance:
Relative risk 6.29
(CI 95% 0.33 —
0 40 Very low
119.69) per 1000 per 1000
parasitaemia Due to serious
on day 2 Based on data from indirectness and
144 participants in 1 very serious
studies. (Randomized imprecision 6
controlled)
80 110
Relative risk 1.38 per 1000 per 1000
Gametocytaem Very low
(CI 95% 0.52 — 3.7)
ia: on day 7 Due to serious
Based on data from
Difference: 30 more per indirectness and
144 participants in 1
1000 very serious
studies. (Randomized ( CI 95% 38 imprecision 7
controlled) fewer — 216
more )
1. Risk of Bias: no serious. Although the description of the randomization procedure is vague, this trial is probably
at low risk of selection bias. Inconsistency: no serious. Indirectness: serious. This comparison has been evaluated in
only a single setting. Further studies in additional settings are required before this result can be generalized.
Imprecision: very serious. Demonstration of non-inferiority at 95% efficacy would require a sample size of 472. This
trial is significantly underpowered.
2. Risk of Bias: no serious. Although the description of the randomization procedure is vague, this trial is probably
at low risk of selection bias. Inconsistency: no serious. Indirectness: serious. This comparison has been evaluated in
only a single setting. Further studies in additional settings are required before this result can be generalized.
Imprecision: very serious. Demonstration of non-inferiority at 95% efficacy would require a sample size of 472. This
trial is significantly underpowered.
3. Risk of Bias: no serious. Although the description of the randomization procedure is vague, this trial is probably
at low risk of selection bias. Inconsistency: no serious. Indirectness: serious. This comparison has been evaluated in
only a single setting. Further studies in additional settings are required before this result can be generalized.
Imprecision: very serious. Demonstration of non-inferiority at 95% efficacy would require a sample size of 472. This
trial is significantly underpowered.
4. Risk of Bias: no serious. Although the description of the randomization procedure is vague, this trial is probably
at low risk of selection bias. Inconsistency: no serious. Indirectness: serious. This comparison has been evaluated in
only a single setting. Further studies in additional settings are required before this result can be generalized.
Imprecision: very serious. Demonstration of non-inferiority at 95% efficacy would require a sample size of 472. This
trial is significantly underpowered.
391 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
5. Risk of Bias: no serious. Although the description of the randomization procedure is vague, this trial is probably
at low risk of selection bias. Inconsistency: no serious. Indirectness: serious. This comparison has been evaluated in
only a single setting. Further studies in additional settings are required before this result can be generalized.
Imprecision: very serious. This trial is small. No participants in either group had fever on day 2.
6. Risk of Bias: no serious. Although the description of the randomization procedure is vague, this trial is probably
at low risk of selection bias. Inconsistency: no serious. Indirectness: serious. This comparison has been evaluated in
only a single setting. Further studies in additional settings are required before this result can be generalized.
Imprecision: very serious. The result has a very wide 95% confidence interval, including appreciable benefit and
harm.
7. Risk of Bias: no serious. Although the description of the randomization procedure is vague, this trial is probably
at low risk of selection bias. Inconsistency: no serious. Indirectness: serious. This comparison has been evaluated in
only a single setting. Further studies in additional settings are required before this result can be generalized.
Imprecision: very serious. The result has a very wide 95% confidence interval, including appreciable benefit and
harm.
Attached Images
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements AL ASPY (Quality of summary
evidence)
Low
15 9 Due to serious
per 1000 per 1000 indirectness,
Due to serious
Difference: 6 fewer per imprecision.
1000 Certainty of the
( CI 95% 11 evidence grade
fewer — 5 more ) differs from the
2014 review
version due to
additional data:
the previous
Total failure: Relative risk 0.59 review reported
day 28 (PCR- (CI 95% 0.26 — 1.31) no substantial Compared to AL, ASPY
adjusted) Based on data from difference may have fewer PCR-
3,068 participants in 4 between ASPY adjusted failures at day
studies. 1 (Randomized and AL in 28.
controlled) reference to this
outcome and
therefore did not
downgrade for
imprecision. In
this update we
report a reduced
rate in the ASPY
arm. Because we
concluded that
there may be a
difference, we
necessarily
392 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements AL ASPY (Quality of summary
evidence)
downgraded for
the imprecision.
2
23 20
Total failure: Relative risk 0.86 per 1000 per 1000 Low There may be little or
day 42 (PCR- (CI 95% 0.49 — 1.51)
Due to serious no difference in PCR-
adjusted) Based on data from
Difference: 3 fewer per indirectness, adjusted failures at day
2,575 participants in 4
1000 Due to serious 42 between ASPY and
studies. 3 (Randomized ( CI 95% 12 imprecision 4 AL.
controlled) fewer — 12 more
)
Low
126 34 Due to serious
per 1000 per 1000 indirectness,
Due to serious
Difference: 92 fewer per inconsistency,
1000 Certainty of the
Total failure: Relative risk 0.27 ( CI 95% 110 evidence grade
day 28 (CI 95% 0.13 — 0.58) fewer — 53 Compared to AL, ASPY
differs from the
(unadjusted) Based on data from fewer ) may have fewer
2014 review
3,149 participants in 4 unadjusted failures at
version due to
studies. 5 (Randomized additional data:
day 28.
controlled) the introduction
of more data
increased the
heterogeneity
between the
included trials. 6
Low
254 155 Due to serious
per 1000 per 1000 inconsistency,
Due to serious
Difference: 99 fewer per indirectness,
1000 Certainty of the
Total failure: Relative risk 0.61 ( CI 95% 137 evidence grade
day 42 (CI 95% 0.46 — 0.82) fewer — 46 Compared to AL, ASPY
differs from the
(unadjusted) Based on data from fewer ) may have fewer
2014 review
3,080 participants in 4 unadjusted failures at
version due to
studies. 7 (Randomized additional data:
day 42.
controlled) the introduction
of more data
increased the
heterogeneity
between the
included trials. 8
393 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements AL ASPY (Quality of summary
evidence)
First treatment,
abnormal ALT
Relative risk 3.34
(CI 95% 1.33 — 8.39)
3 10 Low
Compared to AL, ASPY
may lead to higher
Based on data from per 1000 per 1000
increase (42 Due to serious events of abnormal
days) 3,415 participants in 4 indirectness, ALT increase.
Difference: 7 more per 1000
studies. 11 Due to serious (Aggregate analysis
(Randomized ( CI 95% 1 more imprecision 12 indicates this estimate
controlled) — 22 more ) may be accurate).
First treatment,
abnormal
Relative risk 0.82 6 5
(CI 95% 0.33 — 2.04) per 1000 per 1000 Low
bilirubin Based on data from Due to serious
We do not know if
increase (42 there is a difference in
3,130 participants in 3 Difference: 1 fewer per indirectness,
days) bilirubin between
studies. 15 1000 Due to serious
ASPY and AL.
(Randomized ( CI 95% 4 fewer imprecision 16
controlled) — 6 more )
1. Systematic review [184] with included studies: Sagara 2018 (Bobo-Doiulasso, Burkina Faso), Sagara 2018 (Kolle,
Mali), Tshefu 2010, Sagara 2018 (Bougoula, Mali), Roth 2018a, Kayentao 2012, Sagara 2018 (Sotuba, Mali). Baseline/
comparator: Control arm of reference used for intervention.
2. Inconsistency: no serious. Indirectness: serious. The trials included adults and children and had sites in Africa
and Asia. However, across the trials, only 115 children and 0 adults were randomized to ASPY in Asia. Further
adequately powered studies in adults and children in Asia would be needed to fully apply this result.. Imprecision:
serious. The CIs are wide and include both almost no effect and clinically significant effect..
3. Systematic review [184] with included studies: Sagara 2018 (Kolle, Mali), Roth 2018a, Sagara 2018 (Bobo-
Doiulasso, Burkina Faso), Sagara 2018 (Sotuba, Mali), Kayentao 2012, Sagara 2018 (Bougoula, Mali), Tshefu 2010.
Baseline/comparator: Control arm of reference used for intervention.
4. Inconsistency: no serious. Indirectness: serious. The trials included adults and children and had sites in Africa
and Asia. However, across the trials, only 115 children and 0 adults were randomized to ASPY in Asia. Further
adequately powered studies in adults and children in Asia would be needed to fully apply this result.. Imprecision:
serious. The CIs are wide and include both almost no effect and clinically significant effect..
5. Systematic review [184] with included studies: Tshefu 2010, Roth 2018a, Sagara 2018 (Bougoula, Mali), Sagara
2018 (Kolle, Mali), Sagara 2018 (Bobo-Doiulasso, Burkina Faso), Kayentao 2012, Sagara 2018 (Sotuba, Mali).
Baseline/comparator: Control arm of reference used for intervention.
6. Inconsistency: serious. There was quantitative heterogeneity between studies.. Indirectness: serious. The trials
included adults and children and had sites in Africa and Asia. However, across the trials, only 115 children and 0
adults were randomized to ASPY in Asia. Further adequately powered studies in adults and children in Asia would be
needed to fully apply this result.. Imprecision: no serious.
7. Systematic review [184] with included studies: Sagara 2018 (Bougoula, Mali), Sagara 2018 (Sotuba, Mali), Tshefu
2010, Roth 2018a, Kayentao 2012, Sagara 2018 (Bobo-Doiulasso, Burkina Faso), Sagara 2018 (Kolle, Mali). Baseline/
comparator: Control arm of reference used for intervention.
8. Inconsistency: serious. There was quantitative heterogeneity between studies.. Indirectness: serious. The trials
included adults and children and had sites in Africa and Asia. However, across the trials, only 115 children and 0
adults were randomized to ASPY in Asia. Further adequately powered studies in adults and children in Asia would be
needed to fully apply this result.. Imprecision: no serious.
394 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
9. Systematic review [184] with included studies: Tshefu 2010, Kayentao 2012, Roth 2018a. Baseline/comparator:
Control arm of reference used for intervention.
10. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. The low number of events
recorded in the studies is insufficient for confidently estimating the effect size. .
11. Systematic review [184] with included studies: Sagara 2018 (Bobo-Doiulasso, Burkina Faso), Roth 2018a,
Sagara 2018 (Sotuba, Mali), Tshefu 2010, Sagara 2018 (Kolle, Mali), Sagara 2018 (Bougoula, Mali), Kayentao 2012.
Baseline/comparator: Control arm of reference used for intervention.
12. Inconsistency: no serious. Indirectness: serious. The trials included adults and children and had sites in Africa
and Asia. However, across the trials, only 115 children and 0 adults were randomized to ASPY in Asia. Further
adequately powered studies in adults and children in Asia would be needed to fully apply this result.. Imprecision:
serious. The CIs are wide and include both almost no effect and clinically significant effect..
13. Systematic review [184] with included studies: Sagara 2018 (Sotuba, Mali), Sagara 2018 (Bobo-Doiulasso,
Burkina Faso), Sagara 2018 (Bougoula, Mali), Tshefu 2010, Kayentao 2012, Roth 2018a, Sagara 2018 (Kolle, Mali).
Baseline/comparator: Control arm of reference used for intervention.
14. Inconsistency: no serious. Indirectness: serious. The trials included adults and children and had sites in Africa
and Asia. However, across the trials, only 115 children and 0 adults were randomized to ASPY in Asia. Further
adequately powered studies in adults and children in Asia would be needed to fully apply this result.. Imprecision:
serious. The CIs are wide and include both almost no effect and clinically significant effect..
15. Systematic review [184] with included studies: Sagara 2018 (Bougoula, Mali), Sagara 2018 (Kolle, Mali), Sagara
2018 (Bobo-Doiulasso, Burkina Faso), Kayentao 2012, Tshefu 2010, Sagara 2018 (Sotuba, Mali). Baseline/
comparator: Control arm of reference used for intervention.
16. Inconsistency: no serious. Indirectness: serious. The trials included adults and children and had sites in Africa
and Asia. However, across the trials, only 115 children and 0 adults were randomized to ASPY in Asia. Further
adequately powered studies in adults and children in Asia would be needed to fully apply this result.. Imprecision:
serious. The CIs include both no effect and clinically significant effect.
Attached Images
References
184. Pryce J, Taylor M, Fox T, Hine P : Pyronaridine-artesunate for treating uncomplicated Plasmodium
falciparum malaria. Cochrane Database Syst Rev 2022;6(6): Pubmed Journal
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements AS-AQ ASPY (Quality of summary
evidence)
395 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements AS-AQ ASPY (Quality of summary
evidence)
75 37
Total failure: Relative risk 0.49 per 1000 per 1000
day 28 (CI 95% 0.3 — 0.81) Compared to AS-AQ,
Moderate
(unadjusted) Based on data from ASPY probably has
Difference: 38 fewer per Due to serious
1,257 participants in 1 fewer unadjusted
1000 indirectness 6
studies. 5 (Randomized ( CI 95% 52
failures at day 28.
controlled) fewer — 14
fewer )
195 192
Total failure: Relative risk 0.98 per 1000 per 1000 There is probably little
day 42 (CI 95% 0.78 — 1.23)
Moderate or no difference in
(unadjusted) Based on data from
Difference: 4 fewer per Due to serious unadjusted failures at
1,235 participants in 1
1000 indirectness 8 day 42 between ASPY
studies. 7 (Randomized ( CI 95% 43 and AS-AQ.
controlled) fewer — 45 more
)
First treatment,
Relative risk 1.41 1 1 Compared to AL, ASPY
abnormal ALT per 1000 per 1000 Low may lead to higher
(CI 95% 0.28 — 7.09)
increase (42 Due to serious events of abnormal
Based on data from
days) Difference: 0 fewer per indirectness, ALT increase.
1,317 participants in 1
1000 Due to serious (Aggregate analysis
studies. 9 (Randomized
( CI 95% 1 fewer imprecision 10 indicates this estimate
controlled)
may be accurate).
— 6 more )
First treatment,
abnormal
Relative risk 0.99 1 1 Very low
(CI 95% 0.06 — 15.76) per 1000 per 1000
bilirubin Based on data from
Due to serious We do not know if
increase (42 indirectness, there is a difference in
1,317 participants in 1 Difference: 0 fewer per
days) Due to very bilirubin between
studies. 13 1000 serious ASPY and AS-AQ
(Randomized ( CI 95% 1 fewer
imprecision 14
controlled) — 15 more )
396 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements AS-AQ ASPY (Quality of summary
evidence)
adverse events
15 disaggregated by site
to allow inclusion in
this comparison.
1. Systematic review [184] with included studies: Sagara 2018 (Bougoula, Mali), Sagara 2018 (Mafrinyah, Guinea),
Sagara 2018 (Sotuba, Mali), Sagara 2018 (Djoliba, Mali), Sagara 2018 (Kolle, Mali), Sagara 2018 (Ouagadougou,
Burkina Faso). Baseline/comparator: Control arm of reference used for intervention.
2. Inconsistency: no serious. Indirectness: serious. The data are drawn from one study, conducted in six sites in
three countries in West Africa. Further studies in Asia would be needed to fully apply this result.. Imprecision:
serious. The CI is large and includes both no effect and clinically important effects..
3. Systematic review [184] with included studies: Sagara 2018 (Djoliba, Mali), Sagara 2018 (Kolle, Mali), Sagara
2018 (Sotuba, Mali), Sagara 2018 (Bougoula, Mali), Sagara 2018 (Mafrinyah, Guinea), Sagara 2018 (Ouagadougou,
Burkina Faso). Baseline/comparator: Control arm of reference used for intervention.
4. Inconsistency: no serious. Indirectness: serious. The data are drawn from one study, conducted in six sites in
three countries in West Africa. Further studies in Asia would be needed to fully apply this result.. Imprecision:
serious. The effect estimate is close to no effect, but the CI is wide..
5. Systematic review [184] with included studies: Sagara 2018 (Mafrinyah, Guinea), Sagara 2018 (Ouagadougou,
Burkina Faso), Sagara 2018 (Bougoula, Mali), Sagara 2018 (Kolle, Mali), Sagara 2018 (Sotuba, Mali), Sagara 2018
(Djoliba, Mali). Baseline/comparator: Control arm of reference used for intervention.
6. Inconsistency: no serious. Indirectness: serious. The data are drawn from one study, conducted in six sites in
three countries in West Africa. Further studies in Asia would be needed to fully apply this result.. Imprecision: no
serious.
7. Systematic review [184] with included studies: Sagara 2018 (Djoliba, Mali), Sagara 2018 (Kolle, Mali), Sagara
2018 (Sotuba, Mali), Sagara 2018 (Bougoula, Mali), Sagara 2018 (Mafrinyah, Guinea), Sagara 2018 (Ouagadougou,
Burkina Faso). Baseline/comparator: Control arm of reference used for intervention.
8. Inconsistency: no serious. Indirectness: serious. The data are drawn from one study, conducted in six sites in
three countries in West Africa. Further studies in Asia would be needed to fully apply this result.. Imprecision: no
serious.
9. Systematic review [184] with included studies: Sagara 2018 (Kolle, Mali), Sagara 2018 (Mafrinyah, Guinea),
Sagara 2018 (Djoliba, Mali), Sagara 2018 (Bougoula, Mali), Sagara 2018 (Sotuba, Mali), Sagara 2018 (Ouagadougou,
Burkina Faso). Baseline/comparator: Control arm of reference used for intervention.
10. Inconsistency: no serious. Indirectness: serious. The data are drawn from one study, conducted in six sites in
three countries in West Africa. Further studies in Asia would be needed to fully apply this result.. Imprecision:
serious. The low number of events recorded in the study is insufficient for confidently estimating the effect size.
However, aggregate analysis of ALT increase across different comparator drugs provides indirect evidence that the
point estimate may be accurate..
11. Systematic review [184] with included studies: Sagara 2018 (Djoliba, Mali), Sagara 2018 (Sotuba, Mali), Sagara
2018 (Ouagadougou, Burkina Faso), Sagara 2018 (Mafrinyah, Guinea), Sagara 2018 (Kolle, Mali), Sagara 2018
(Bougoula, Mali). Baseline/comparator: Control arm of reference used for intervention.
12. Inconsistency: no serious. Indirectness: serious. The data are drawn from one study, conducted in six sites in
three countries in West Africa. Further studies in Asia would be needed to fully apply this result.. Imprecision: very
serious. The CI is very large and includes both no effect and clinically important effects..
13. Systematic review [184] with included studies: Sagara 2018 (Mafrinyah, Guinea), Sagara 2018 (Djoliba, Mali),
Sagara 2018 (Bougoula, Mali), Sagara 2018 (Sotuba, Mali), Sagara 2018 (Ouagadougou, Burkina Faso), Sagara 2018
(Kolle, Mali). Baseline/comparator: Control arm of reference used for intervention.
14. Inconsistency: no serious. Indirectness: serious. The data are drawn from one study, conducted in six sites in
three countries in West Africa. Further studies in Asia would be needed to fully apply this result.. Imprecision: very
serious. The CI is very large and includes both no effect and clinically important effects..
15. Serious adverse events data were not available disaggregated by site to allow inclusion in this comparison.
397 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Attached Images
References
184. Pryce J, Taylor M, Fox T, Hine P : Pyronaridine-artesunate for treating uncomplicated Plasmodium
falciparum malaria. Cochrane Database Syst Rev 2022;6(6): Pubmed Journal
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements MQ+AS ASPY (Quality of summary
evidence)
Low
22 8 Due to serious
per 1000 per 1000 indirectness,
Due to serious
Difference: 14 fewer per imprecision,
1000 Certainty of the
( CI 95% 19 evidence grade
fewer — 1 more ) differs from the
Total failure: Relative risk 0.37
2014 review
day 28 (PCR- (CI 95% 0.13 — 1.05) Compared to MQ+AS,
version due to
adjusted) Based on data from ASPY may have fewer
alterations in the
1,117 participants in 1 PCR-adjusted failures
data extraction
studies. 1 (Randomized protocol: the CI
at day 28.
controlled) has become less
precise, and our
decision has
greater
consistency with
other outcome
certainty grades.
2
398 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements MQ+AS ASPY (Quality of summary
evidence)
Low
83 70 Due to serious
per 1000 per 1000 indirectness,
Due to serious
Difference: 13 fewer per imprecision,
1000 Certainty of the
( CI 95% 38 evidence grade
fewer — 26 more differs from the
Total failure: Relative risk 0.84 ) 2014 review There is probably little
day 42 (CI 95% 0.54 — 1.31)
version due to or no difference in
(unadjusted) Based on data from
alterations in the unadjusted failures at
1,059 participants in 1
data extraction day 42 between ASPY
studies. 7 (Randomized protocol: the CI and MQ+AS.
controlled) has become less
precise, and our
decision has
greater
consistency with
other outcome
certainty grades.
8
Adverse events 9 6
Relative risk 0.62 per 1000 per 1000
leading to (CI 95% 0.17 — 2.31)
withdrawal Based on data from
Difference: 3 fewer per
1,271 participants in 1
1000
studies. 11 ( CI 95% 7 fewer
— 12 more )
399 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements MQ+AS ASPY (Quality of summary
evidence)
First treatment,
abnormal
Relative risk 3.49 2 7 Very low
(CI 95% 0.43 — 28.29) per 1000 per 1000
bilirubin Based on data from
Due to serious We do not know if
increase (42 indirectness, there is a difference in
1,271 participants in 1 Difference: 5 more per 1000
days) Due to very bilirubin between
studies. 16 ( CI 95% 1 fewer serious ASPY and MQ+AS.
(Randomized — 55 more )
imprecision 17
controlled)
1. Systematic review [184] with included studies: Rueangweerayut 2012. Baseline/comparator: Control arm of
reference used for intervention.
2. Inconsistency: no serious. Indirectness: serious. Of the 1271 children and adults aged greater than 5 years
enrolled in this trial, 81.3% (1033) were enrolled and treated in trial sites in Asia (Cambodia, India, Thailand, and
Vietnam), and only 18.7% (237) in Africa (Burkina Faso, Ivory Coast, and Tanzania). Further studies in African children
are necessary to fully apply this result.. Imprecision: serious. The CI is large and includes both no effect and clinically
important effects.
3. Systematic review [184] with included studies: Rueangweerayut 2012. Baseline/comparator: Control arm of
reference used for intervention.
4. Inconsistency: no serious. Indirectness: serious. Of the 1271 children and adults aged greater than 5 years
enrolled in this trial, 81.3% (1033) were enrolled and treated in trial sites in Asia (Cambodia, India, Thailand, and
Vietnam), and only 18.7% (237) in Africa (Burkina Faso, Ivory Coast, and Tanzania). Further studies in African children
are necessary to fully apply this result.. Imprecision: serious. The CI is large and includes both no effect and clinically
important effects..
5. Systematic review [184] with included studies: Rueangweerayut 2012. Baseline/comparator: Control arm of
reference used for intervention.
6. Inconsistency: no serious. Indirectness: serious. Of the 1271 children and adults aged greater than 5 years
enrolled in this trial, 81.3% (1033) were enrolled and treated in trial sites in Asia (Cambodia, India, Thailand, and
Vietnam), and only 18.7% (237) in Africa (Burkina Faso, Ivory Coast, and Tanzania). Further studies in African children
are necessary to fully apply this result.. Imprecision: no serious.
7. Systematic review [184] with included studies: Rueangweerayut 2012. Baseline/comparator: Control arm of
reference used for intervention.
8. Inconsistency: no serious. Indirectness: serious. Of the 1271 children and adults aged greater than 5 years
enrolled in this trial, 81.3% (1033) were enrolled and treated in trial sites in Asia (Cambodia, India, Thailand, and
Vietnam), and only 18.7% (237) in Africa (Burkina Faso, Ivory Coast, and Tanzania). Further studies in African children
are necessary to fully apply this result.. Imprecision: serious. The CI is large and includes both no effect and clinically
important effects..
9. Systematic review [184] with included studies: Rueangweerayut 2012. Baseline/comparator: Control arm of
reference used for intervention.
10. Inconsistency: no serious. Indirectness: serious. Of the 1271 children and adults aged greater than 5 years
enrolled in this trial, 81.3% (1033) were enrolled and treated in trial sites in Asia (Cambodia, India, Thailand, and
Vietnam), and only 18.7% (237) in Africa (Burkina Faso, Ivory Coast, and Tanzania). Further studies in African children
are necessary to fully apply this result.. Imprecision: serious. The CI is large and includes both no effect and clinically
important effects..
11. Systematic review [184] with included studies: Rueangweerayut 2012. Baseline/comparator: Control arm of
reference used for intervention.
12. Systematic review [184] with included studies: Rueangweerayut 2012. Baseline/comparator: Control arm of
reference used for intervention.
13. Inconsistency: no serious. Indirectness: serious. Of the 1271 children and adults aged greater than 5 years
enrolled in this trial, 81.3% (1033) were enrolled and treated in trial sites in Asia (Cambodia, India, Thailand, and
Vietnam), and only 18.7% (237) in Africa (Burkina Faso, Ivory Coast, and Tanzania). Further studies in African children
are necessary to fully apply this result.. Imprecision: serious. The low number of events recorded in the study is
insufficient for confidently estimating the effect size. However, aggregate analysis of ALT increase across different
400 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
comparator drugs provides indirect evidence that the point estimate may be accurate..
14. Systematic review [184] with included studies: Rueangweerayut 2012. Baseline/comparator: Control arm of
reference used for intervention.
15. Inconsistency: no serious. Indirectness: serious. Of the 1271 children and adults aged greater than 5 years
enrolled in this trial, 81.3% (1033) were enrolled and treated in trial sites in Asia (Cambodia, India, Thailand, and
Vietnam), and only 18.7% (237) in Africa (Burkina Faso, Ivory Coast, and Tanzania). Further studies in African children
are necessary to fully apply this result.. Imprecision: very serious. The CI is very large and includes both no effect and
clinically important effects..
16. Systematic review [184] with included studies: Rueangweerayut 2012. Baseline/comparator: Control arm of
reference used for intervention.
17. Inconsistency: no serious. Indirectness: serious. Of the 1271 children and adults aged greater than 5 years
enrolled in this trial, 81.3% (1033) were enrolled and treated in trial sites in Asia (Cambodia, India, Thailand, and
Vietnam), and only 18.7% (237) in Africa (Burkina Faso, Ivory Coast, and Tanzania). Further studies in African children
are necessary to fully apply this result.. Imprecision: very serious. The CI is very large and includes both no effect and
clinically important effects..
Attached Images
References
184. Pryce J, Taylor M, Fox T, Hine P : Pyronaridine-artesunate for treating uncomplicated Plasmodium
falciparum malaria. Cochrane Database Syst Rev 2022;6(6): Pubmed Journal
Certainty of
Comparator
Outcome Study results and Intervention the Evidence Plain language
other
Timeframe measurements ASPY (Quality of summary
antimalarials
evidence)
Serious
Relative risk 1.24 5 7 There is probably little
(CI 95% 0.54 — 2.84) per 1000 per 1000 or no difference in the
adverse events Moderate
Based on data from rate of serious adverse
Due to serious
3,941 participants in 7 events with ASPY
Difference: 1 more per 1000 imprecision 2
studies. 1 (Randomized ( CI 95% 2 fewer
compared to other
controlled) antimalarials.
— 9 more )
401 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Comparator
Outcome Study results and Intervention the Evidence Plain language
other
Timeframe measurements ASPY (Quality of summary
antimalarials
evidence)
First treatment,
Relative risk 1.03 4 4
abnormal (CI 95% 0.49 — 2.18) per 1000 per 1000 There is probably little
bilirubin Moderate or no difference for
Based on data from
increase Difference: 0 fewer per Due to serious bilirubin between
6,417 participants in 7
7 1000 imprecision 8 ASPY and other
studies. (Randomized
antimalarials.
controlled) ( CI 95% 2 fewer
— 5 more )
Subsequent
treatment(s),
Relative risk 2.18 4 8 Low
There may be an
(CI 95% 0.76 — 6.27) per 1000 per 1000 increased risk of raised
abnormal ALT Due to serious
Based on data from ALT with subsequent
increase indirectness,
1,649 participants in 1 Difference: 5 more per 1000 treatments with ASPY
Due to serious
studies. 9 (Randomized ( CI 95% 1 fewer compared to other
imprecision 10 antimalarials.
controlled) — 21 more )
Subsequent
treatment(s),
Relative risk 1.82
(CI 95% 0.74 — 4.44)
6 11 Low
There may be an
per 1000 per 1000 increased risk of raised
abnormal AST Based on data from Due to serious
AST with subsequent
increase 1,649 participants in 1 indirectness,
Difference: 5 more per 1000 treatments with ASPY
studies. 11 Due to serious
( CI 95% 2 fewer compared to other
(Randomized imprecision 12 antimalarials.
controlled) — 21 more )
Subsequent
treatment(s),
Relative risk 1.13 8 9
(CI 95% 0.42 — 3.01) per 1000 per 1000 Low There may be little or
abnormal Based on data from Due to serious no difference for
bilirubin 1,649 participants in 1 indirectness, bilirubin between
Difference: 1 more per 1000
increase studies. 13 ( CI 95% 5 fewer Due to serious ASPY and other
(Randomized — 16 more ) imprecision 14 antimalarials.
controlled)
1. Systematic review [184] with included studies: Roth 2018a, Kayentao 2012, Nelwan 2015, Tshefu 2010,
Rueangweerayut 2012, Shin 2011, Poravuth 2011. Baseline/comparator: Control arm of reference used for
intervention.
2. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. The CI includes both no effect and
clinically important effects..
3. Systematic review [184] with included studies: Sagara 2018 (Sotuba, Mali), Sagara 2018 (Mafrinyah, Guinea),
Rueangweerayut 2012, Sagara 2018 (Kolle, Mali), Tshefu 2010, Shin 2011, Nelwan 2015, Kayentao 2012, Sagara
2018 (Bougoula, Mali), Sagara 2018 (Bougoula, Mali), Sagara 2018 (Sotuba, Mali), Sagara 2018 (Ouagadougou,
Burkina Faso), Poravuth 2011, Sagara 2018 (Djoliba, Mali), Sagara 2018 (Bobo-Doiulasso, Burkina Faso), Sagara 2018
(Kolle, Mali), Roth 2018a. Baseline/comparator: Control arm of reference used for intervention.
4. Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious. Although the CI is wide, there were
few events..
5. Systematic review [184] with included studies: Nelwan 2015, Poravuth 2011, Sagara 2018 (Mafrinyah, Guinea),
402 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Sagara 2018 (Kolle, Mali), Roth 2018a, Shin 2011, Kayentao 2012, Sagara 2018 (Bougoula, Mali), Tshefu 2010,
Sagara 2018 (Sotuba, Mali), Sagara 2018 (Bobo-Doiulasso, Burkina Faso), Sagara 2018 (Kolle, Mali), Sagara 2018
(Sotuba, Mali), Sagara 2018 (Ouagadougou, Burkina Faso), Sagara 2018 (Djoliba, Mali), Rueangweerayut 2012, Sagara
2018 (Bougoula, Mali). Baseline/comparator: Control arm of reference used for intervention.
6. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. The CI includes both almost no effect
and clinically important effects.
7. Systematic review [184] with included studies: Tshefu 2010, Sagara 2018 (Kolle, Mali), Shin 2011,
Rueangweerayut 2012, Sagara 2018 (Bobo-Doiulasso, Burkina Faso), Sagara 2018 (Sotuba, Mali), Poravuth 2011,
Nelwan 2015, Sagara 2018 (Bougoula, Mali), Sagara 2018 (Mafrinyah, Guinea), Sagara 2018 (Ouagadougou, Burkina
Faso), Kayentao 2012, Sagara 2018 (Sotuba, Mali), Sagara 2018 (Bougoula, Mali), Sagara 2018 (Djoliba, Mali), Sagara
2018 (Kolle, Mali). Baseline/comparator: Control arm of reference used for intervention.
8. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. The CI includes both no effect and
clinically important effects..
9. Systematic review [184] with included studies: Sagara 2018 (Sotuba, Mali), Sagara 2018 (Sotuba, Mali), Sagara
2018 (Ouagadougou, Burkina Faso), Sagara 2018 (Mafrinyah, Guinea), Sagara 2018 (Djoliba, Mali), Sagara 2018
(Kolle, Mali), Sagara 2018 (Bougoula, Mali), Sagara 2018 (Bobo-Doiulasso, Burkina Faso), Sagara 2018 (Kolle, Mali),
Sagara 2018 (Bougoula, Mali). Baseline/comparator: Control arm of reference used for intervention.
10. Inconsistency: no serious. Indirectness: serious. The CI includes both no effect and clinically important effects..
Imprecision: serious.
11. Systematic review [184] with included studies: Sagara 2018 (Sotuba, Mali), Sagara 2018 (Sotuba, Mali), Sagara
2018 (Bougoula, Mali), Sagara 2018 (Kolle, Mali), Sagara 2018 (Mafrinyah, Guinea), Sagara 2018 (Bobo-Doiulasso,
Burkina Faso), Sagara 2018 (Kolle, Mali), Sagara 2018 (Ouagadougou, Burkina Faso), Sagara 2018 (Bougoula, Mali),
Sagara 2018 (Djoliba, Mali). Baseline/comparator: Control arm of reference used for intervention.
12. Inconsistency: no serious. Indirectness: serious. Imprecision: serious.
13. Systematic review [184] with included studies: Sagara 2018 (Bougoula, Mali), Sagara 2018 (Sotuba, Mali),
Sagara 2018 (Mafrinyah, Guinea), Sagara 2018 (Djoliba, Mali), Sagara 2018 (Kolle, Mali), Sagara 2018 (Bougoula,
Mali), Sagara 2018 (Kolle, Mali), Sagara 2018 (Ouagadougou, Burkina Faso), Sagara 2018 (Sotuba, Mali), Sagara 2018
(Bobo-Doiulasso, Burkina Faso). Baseline/comparator: Control arm of reference used for intervention.
14. Inconsistency: no serious. Indirectness: serious. Imprecision: serious.
Attached Images
References
184. Pryce J, Taylor M, Fox T, Hine P : Pyronaridine-artesunate for treating uncomplicated Plasmodium
falciparum malaria. Cochrane Database Syst Rev 2022;6(6): Pubmed Journal
403 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator Intervention
Artesunate 1 Artesunate 3 Certainty of
Outcome Study results and day plus days plus the Evidence Plain language
Timeframe measurements sulfadoxine- sulfadoxine- (Quality of summary
pyrimethamin pyrimethamin evidence)
e e
19 7
Parasitological Relative risk 0.36 per 1000 per 1000
failure (CI 95% 0.27 — 0.5)
14 days Based on data from
Difference: 12 fewer per High
1,276 participants in 4 1
1000
studies. (Randomized ( CI 95% 14
controlled) fewer — 9 fewer
)
Parasitological 47 29
Relative risk 0.62 per 1000 per 1000
failure - PCR- (CI 95% 0.54 — 0.71) *Corresponding risk
unadjusted Based on data from High calculated is different
28 days Difference: 18 fewer per
1,260 participants in 4 2 than what is reported
1000
studies. (Randomized in WHO document*
( CI 95% 22
controlled) fewer — 14
fewer )
Parasitological 33 15
Relative risk 0.45 per 1000 per 1000
failure - PCR- (CI 95% 0.36 — 0.55) *Corresponding risk
adjusted Based on data from High calculated is different
28 days Difference: 18 fewer per
1,202 participants in 4 3 than what is reported
1000
studies. (Randomized in WHO document*
( CI 95% 21
controlled) fewer — 15
fewer )
20 15
Gametocytae Relative risk 0.74 per 1000 per 1000
mia (CI 95% 0.58 — 0.93)
7 days Based on data from
Difference: 5 fewer per High
1,260 participants in 4 4
1000
studies. (Randomized ( CI 95% 8
controlled) fewer — 1 fewer
)
11 9
Gametocytae Relative risk 0.8 per 1000 per 1000
mia (CI 95% 0.57 — 1.14) *Corresponding risk
14 days Based on data from
Difference: 2 fewer per High calculated is different
1,199 participants in 4 5 than what is reported
1000
studies. (Randomized in WHO document*
( CI 95% 5
controlled) fewer — 2 more
)
404 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator Intervention
Artesunate 1 Artesunate 3 Certainty of
Outcome Study results and day plus days plus the Evidence Plain language
Timeframe measurements sulfadoxine- sulfadoxine- (Quality of summary
pyrimethamin pyrimethamin evidence)
e e
( CI 95% 3
fewer — 0 fewer
)
1. Inconsistency: no serious. All four studies found reductions with 3 days of artesunate, although there was
some variation in the size of this effect. Indirectness: no serious. The four trials were conducted in children with
uncomplicated P. falciparum malaria in the Gambia, Kenya, Malawi and Uganda. The same screening methods and
inclusion criteria were used. Sulfadoxine–pyrimethamine was the partner antimalarial drug in all four trials.
Resistance to sulfadoxine–pyrimethamine was noted at three study sites, parasitological failure with
sulfadoxine–pyrimethamine alone being seen in 10–13% of participants in the Gambia, 27% in Kenya and 25% in
Uganda. Imprecision: no serious. The confidence intervals are narrow, and the intervals comprise clinically
important effects. No serious imprecision: The confidence intervals are narrow and do not include no effect.
2. Inconsistency: no serious. All four studies found reductions with 3 days of artesunate, although there was
some variation in the size of this effect. Indirectness: no serious. The four trials were conducted in children with
uncomplicated P. falciparum malaria in the Gambia, Kenya, Malawi and Uganda. The same screening methods and
inclusion criteria were used. Sulfadoxine–pyrimethamine was the partner antimalarial drug in all four trials.
Resistance to sulfadoxine–pyrimethamine was noted at three study sites, parasitological failure with
sulfadoxine–pyrimethamine alone being seen in 10–13% of participants in the Gambia, 27% in Kenya and 25% in
Uganda. Imprecision: no serious. The confidence intervals are narrow, and the intervals comprise clinically
important effects. No serious imprecision: The confidence intervals are narrow and do not include no effect.
3. Inconsistency: no serious. All four studies found reductions with 3 days of artesunate, although there was
some variation in the size of this effect. Indirectness: no serious. The four trials were conducted in children with
uncomplicated P. falciparum malaria in the Gambia, Kenya, Malawi and Uganda. The same screening methods and
inclusion criteria were used. Sulfadoxine–pyrimethamine was the partner antimalarial drug in all four trials.
Resistance to sulfadoxine–pyrimethamine was noted at three study sites, parasitological failure with
sulfadoxine–pyrimethamine alone being seen in 10–13% of participants in the Gambia, 27% in Kenya and 25% in
Uganda. Imprecision: no serious. The confidence intervals are narrow, and the intervals comprise clinically
important effects. No serious imprecision: The confidence intervals are narrow and do not include no effect.
4. Inconsistency: no serious. All four studies found reductions with 3 days of artesunate, although there was
some variation in the size of this effect. Indirectness: no serious. The four trials were conducted in children with
uncomplicated P. falciparum malaria in the Gambia, Kenya, Malawi and Uganda. The same screening methods and
inclusion criteria were used. Sulfadoxine–pyrimethamine was the partner antimalarial drug in all four trials.
Resistance to sulfadoxine–pyrimethamine was noted at three study sites, parasitological failure with
sulfadoxine–pyrimethamine alone being seen in 10–13% of participants in the Gambia, 27% in Kenya and 25% in
Uganda. Imprecision: no serious. The confidence intervals are narrow, and the intervals comprise clinically
important effects. No serious imprecision: The confidence intervals are narrow and do not include no effect.
5. Inconsistency: no serious. All four studies found reductions with 3 days of artesunate, although there was
some variation in the size of this effect. Imprecision: no serious. The confidence intervals are narrow, and the
intervals comprise clinically important effects. No serious imprecision: The confidence intervals are narrow and
do not include no effect.
6. Inconsistency: no serious. All four studies found reductions with 3 days of artesunate, although there was
some variation in the size of this effect. Imprecision: serious. The confidence intervals are narrow, and the
intervals comprise clinically important effects. Downgraded by 1 for serious imprecision: As gametocytaemia at
this time was rare in both groups, the studies have inadequate power to confidently detect important differences.
Attached Images
405 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
ACT +
Timeframe measurements ACT (Quality of summary
primaquine
evidence)
Malaria
incidence, CI 95%
prevalence or Relative risk
entomological
inoculation Based on data from 0
rate participants in 0
studies.
Limited observational
People CI 95% data from mosquito
Relative risk
infectious to feeding studies
mosquitoes suggests that 0.25 mg/
Based on data from 0
kg bw may rapidly
participants in 0
reduce the infectivity
studies.
of gametocytes to
mosquitoes.
Participants
with
34 23
per 1000 per 1000
gametocytes Relative risk 0.67
on microscopy (CI 95% 0.44 — 1.02) Low
Difference: 11 fewer per
or PCR (day 8) Based on data from Due to very
1000
(dose < 0.4 mg/ 223 participants in 1 serious
( CI 95% 19
kg bw) 1 studies. (Randomized imprecision 2
fewer — 1 more )
controlled)
406 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
ACT +
Timeframe measurements ACT (Quality of summary
primaquine
evidence)
Participants
with
30 9
per 1000 per 1000
gametocytes Relative risk 0.29
on microscopy (CI 95% 0.22 — 0.37)
Difference: 21 fewer per
or PCR (day 8) Based on data from High
1000
(dose > 0.6 mg/ 1,380 participants in 7 6
( CI 95% 23
kg bw) 5 studies. (Randomized
fewer — 19
controlled)
fewer )
1. AUC estimates (log10 AUC for days 1–43) are included as footnotes for each dosing stratum.
2. Risk of Bias: no serious. Includes one trial with no risk of bias detected. Imprecision: very serious. One small trial
with CIs that include 50% reduction and no effect.
3. AUC estimates (log10 AUC for days 1–43) are included as footnotes for each dosing stratum.
4. Risk of Bias: no serious. Includes one trial with no risk of bias detected. Indirectness: serious. This is a single trial
in a single setting. Imprecision: serious. A single trial with few events.
5. AUC estimates (log10 AUC for days 1–43) are included as footnotes for each dosing stratum.
6. Indirectness: no serious. While there is marked quantitative heterogeneity, the studies with no demonstrable
effect had few events. Not downgraded.
7. One trial reported a relative decrease in haemoglobin against baseline in both groups on days 8, 15, 29 and 43 in
all participants irrespective of G6PD status. No difference at any time between participants receiving primaquine and
those that not did not. We present the data for day 43 in this table.
8. Indirectness: very serious. The percentage of people with large drops in haemoglobin, not the mean change in
the population, is the important safety outcome, and the estimates are averages in a small population (N = 99) that
includes people with normal G6PD function. The study is therefore unlikely to detect effects in a small subgroup
with a relatively uncommon adverse event.
Attached Images
407 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
antimalarial
Certainty of
not including Intervention
Outcome Study results and the Evidence Plain language
artemisinin artemisinin
Timeframe measurements (Quality of summary
derivative derivatives
evidence)
and
recommende
71 53
Relative risk 0.74 per 1000 per 1000
Miscarriage (CI 95% 0.47 — 1.17)
Difference: 18 fewer per ABT may reduce
Based on data from Low
1000 miscarriage
1,739 participants in
12 studies. ( CI 95% 37
fewer — 12
more )
16 11
Relative risk 0.71 per 1000 per 1000
Stillbirth (CI 95% 0.32 — 1.57)
Difference: 5 fewer per ABT may reduce
Based on data from Low
1000 stillbirth
1,389 participants in
12 studies. ( CI 95% 11
fewer — 9 more
)
82 58
Relative risk 0.7 per 1000 per 1000
Fetal loss (CI 95% 0.47 — 1.02)
Difference: 24 fewer per ABT may reduce fetal
Based on data from Low
1000 loss
1,810 participants in
12 studies. ( CI 95% 43
fewer — 2 more
)
Major 7 4
Relative risk 0.6 per 1000 per 1000
congenital (CI 95% 0.13 — 2.87) ABT may reduce
anomalies Based on data from Low congenital
Difference: 3 fewer per
1,810 participants in abnormalities
1000
12 studies. ( CI 95% 6
fewer — 14
408 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator
antimalarial
Certainty of
not including Intervention
Outcome Study results and the Evidence Plain language
artemisinin artemisinin
Timeframe measurements (Quality of summary
derivative derivatives
evidence)
and
recommende
more )
Attached Images
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Artemether-
Timeframe measurements Quinine (Quality of summary
lumefantrine
evidence)
92 54
Relative risk 0.58 per 1000 per 1000
Composite (CI 95% 0.36 — 0.92)
Difference: 37 fewer per AL may reduce
Based on data from Low
1000 adverse fetal events
1,439 participants in
12 studies. ( CI 95% 58
fewer — 7 fewer
)
74 51
Relative risk 0.67 per 1000 per 1000
Miscarriage (CI 95% 0.37 — 1.23)
Difference: 24 fewer per AL may reduce
Based on data from Low
1000 miscarriage
1,377 participants in
12 studies. ( CI 95% 46
fewer — 16
more )
20 11
Relative risk 0.53 per 1000 per 1000
Stillbirth (CI 95% 0.22 — 1.24)
Difference: 10 fewer per AL may reduce
Based on data from Low
1000 stillbirth
1,078 participants in
12 studies. ( CI 95% 16
fewer — 5 more
)
409 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Artemether-
Timeframe measurements Quinine (Quality of summary
lumefantrine
evidence)
Major
congenital
4
per 1000 AL may reduce
anomalies Based on data from Low congenital
1,439 participants in abnormalities
12 studies.
Attached Images
410 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements Chloroquine ACT (Quality of summary
evidence)
520 218
Remaining Relative risk 0.42 per 1000 per 1000
parasitaemia at (CI 95% 0.36 — 0.5)
24 h Based on data from
Difference: 302 fewer per High
1,652 participants in 4 1
1000
studies. (Randomized ( CI 95% 333
controlled) fewer — 260
fewer )
290 160
Relative risk 0.55 per 1000 per 1000
Still febrile (CI 95% 0.43 — 0.7)
after 24 h Moderate
Based on data from
Difference: 130 fewer per Due to serious
990 participants in 2
1000 inconsistency 2
studies. (Randomized ( CI 95% 165
controlled) fewer — 87
fewer )
Effective
treatment of
30 17
per 1000 per 1000
blood-stage
Relative risk 0.58
infection as (CI 95% 0.18 — 1.9) Difference: 13 fewer per
assessed by Based on data from 1000 High
recurrent 1,622 participants in 5 ( CI 95% 25 3
parasitaemia studies. (Randomized fewer — 27 more
before day 28 controlled) )
Post-treatment
prophylaxis as
60 16
per 1000 per 1000
assessed by
recurrent Relative risk 0.27
Difference: 44 fewer per Low
parasitaemia (CI 95% 0.08 — 0.94)
1000 Due to serious
between day Based on data from
( CI 95% 55 indirectness and
28 and day 42, 376 participants in 1
fewer — 4 fewer serious
56 or 63 - with studies. (Randomized ) imprecision 4
controlled)
primaquine
Post-treatment Moderate
prophylaxis as
Relative risk 0.57
(CI 95% 0.4 — 0.82)
400 228 Due to serious
per 1000 per 1000
assessed by Based on data from indirectness 5
411 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements Chloroquine ACT (Quality of summary
evidence)
recurrent
parasitaemia Difference: 172 fewer per
between day 1000
28 and day 42, ( CI 95% 240
1,066 participants in 3 fewer — 72
56 or 63 - studies. (Randomized fewer )
without controlled)
primaquine
Relative risk 1 0 0
Serious (CI 95% 0.14 — 7.04) per 1000 per 1000
adverse events Based on data from High
1,775 participants in 5 Difference: 0 fewer per 6
1. Risk of Bias: no serious. Three studies adequately concealed allocation to be at low risk of selection bias.
Removal of the remaining trials did not substantially change the result. Inconsistency: no serious. The findings of all
the trials are consistent. Indirectness: no serious. The findings of these studies can reasonably be applied to other
settings with similar transmission and resistance patterns. Imprecision: no serious. The studies show a clinically and
statistically significant benefit of ACT. Publication bias: no serious.
2. Risk of Bias: no serious. Three studies adequately concealed allocation to be at low risk of selection bias.
Removal of the remaining trials did not substantially change the result. Inconsistency: serious. In one additional trial
which could not be included in the meta-analysis, fever clearance was not significantly different between groups.
Indirectness: no serious. The findings of these studies can reasonably be applied to other settings with similar
transmission and resistance patterns. Imprecision: no serious. The studies show a clinically and statistically
significant benefit of ACT.
3. Risk of Bias: no serious. Three studies adequately concealed allocation to be at low risk of selection bias.
Removal of the remaining trials did not substantially change the result. Inconsistency: no serious. The findings of all
the trials are consistent. Indirectness: no serious. The findings of these studies can reasonably be applied to other
settings with similar transmission and resistance patterns. Imprecision: no serious. No clinically important difference
between ACTs and chloroquine. Although the 95% CI around the relative effect is very wide, recurrent parasitaemia
before day 28 and serious adverse events were very rare; consequently, the 95% CI around the absolute effect is
very narrow.
4. Indirectness: serious. This study delayed primaquine until day 28; therefore, the course was not completed until
day 42, the last day of the trial. The effect might not be present if primaquine is given in the usual way (on
completion of 3 days of ACT). The period of follow-up was not long enough to fully assess this effect; the inevitable
relapse might simply be delayed, rather than a reduction in clinical episodes. Imprecision: serious. Although the
result is statistically significant, the 95% CI is wide and includes the possibility of no appreciable benefit.
5. Inconsistency: no serious. The findings of all the trials are consistent. Indirectness: serious. Both studies were
conducted in Afghanistan where primaquine is not recommended because of a high prevalence of G6PD deficiency.
The period of follow-up was not long enough to fully assess this effect; the inevitable relapse might simply be
delayed, rather than a reduction in clinical episodes. Imprecision: no serious. The studies show a clinically and
statistically significant benefit of ACT.
6. Risk of Bias: no serious. Three studies adequately concealed allocation to be at low risk of selection bias.
Removal of the remaining trials did not substantially change the result. Inconsistency: no serious. The findings of all
the trials are consistent. Indirectness: no serious. The findings of these studies can reasonably be applied to other
settings with similar transmission and resistance patterns. Imprecision: no serious. No clinically important difference
between ACTs and chloroquine. Although the 95% CI around the relative effect is very wide, recurrent parasitaemia
before day 28 and serious adverse events were very rare; consequently, the 95% CI around the absolute effect is
412 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
very narrow.
Attached Images
Intervention Certainty of
Comparator
Outcome Study results and Dihydroartemi the Evidence Plain language
Alternative
Timeframe measurements sinin + (Quality of summary
ACT
piperaquine evidence)
Effective
treatment of
350 70
per 1000 per 1000
blood-stage
Relative risk 0.2
parasites as (CI 95% 0.08 — 0.49) Difference: 280 fewer per
assessed by Moderate
Based on data from 1000
recurrent Due to serious
334 participants in 3 ( CI 95% 322
parasitaemia fewer — 178 inconsistency 1
studies. (Randomized
before day 28 controlled) fewer )
Post-treatment
prophylaxis as
340 71
per 1000 per 1000
assessed by
recurrent Relative risk 0.21
Difference: 269 fewer per Low
parasitaemia (CI 95% 0.1 — 0.46)
1000 Due to serious
between days Based on data from
( CI 95% 306 risk of bias and
28 and 42 - 179 participants in 2
fewer — 184 serious
with studies. (Randomized fewer ) indirectness 2
controlled)
primaquine
Post-treatment
prophylaxis as
330 132
per 1000 per 1000
assessed by
Very low
recurrent Relative risk 0.4
Difference: 198 fewer per Due to serious
parasitaemia (CI 95% 0.14 — 1.1)
1000 risk of bias,
between days Based on data from 66
( CI 95% 284 serious
28 and 42 - participants in 1
fewer — 33 more indirectness and
without studies. (Randomized ) serious
controlled)
primaquine imprecision 3
1. Risk of Bias: no serious. Allocation was adequately concealed in these studies, resulting in a low risk of bias.
413 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Inconsistency: serious. There was some clinical heterogeneity between trials. Dihydroartemisinin + piperaquine did
not perform as well in Papua New Guinea as it has elsewhere; however, it was still superior to artemether +
lumefantrine and artesunate+sulfadoxine–pyrimethamine. Indirectness: no serious. Studies included adults and
children and were conducted in areas where transmission is high and chloroquine resistance is well documented.
Imprecision: no serious. Both limits of the 95% CI suggest an appreciable clinical benefit with dihydroartemisinin +
piperaquine.
2. Risk of Bias: serious. Losses to follow-up were high (> 20% at this time). Inconsistency: no serious. Statistical
heterogeneity was low. Indirectness: serious. One trial delayed administration of primaquine until day 28; therefore,
the course will not have been completed until the last day of the trial. The second trial offered unsupervised
primaquine to all participants on completion of ACT. This reflects normal practice, but it is not clear how many
participants completed their course. The period of follow-up was not long enough to fully assess this effect; the
inevitable relapse might simply be delayed, rather than a reduction in clinical episodes.
3. Risk of Bias: serious. Losses to follow-up were high (> 20% at this time). Indirectness: serious. Only one study
assessed this outcome. Recurrent parasitaemia was higher with all three ACTs than seen elsewhere, and the results
are therefore not easily extrapolated to other sites. Imprecision: serious. The 95% CI of the effect estimate is wide
and includes an important clinical benefit and no difference between treatments.
Attached Images
414 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Comparator Intervention
Outcome Study results and the Evidence Plain language
No Primaquine 14
Timeframe measurements (Quality of summary
primaquine days
evidence)
P. vivax relapse
defined as
80 48
per 1000 per 1000
reappearance Relative risk 0.6
of P. vivax (CI 95% 0.48 — 0.75)
Difference: 32 fewer per
parasitaemia > Based on data from
1000 High
30 days after 1,740 participants in 1
10 studies. ( CI 95% 42
starting fewer — 20
primaquine (Randomized
fewer )
controlled)
1. Risk of Bias: no serious. No serious study limitations: Three studies were at high risk of bias; however, they
contributed only 15.5% weight to the pooled effect estimates, and their removal from the sensitivity analysis did not
alter the results appreciably. Inconsistency: no serious. Results were consistent within subgroups based on duration
of follow-up < 6 months or > 6 months and whether treatment was supervised or not; the I2 value for the pooled
effect estimate from the 10 trials was 30%. Indirectness: no serious. The trials included children and were done in
transmission settings and countries representative of the vivax malaria burden. The outcome used was the best
estimate currently available in the absence of widely available validated molecular techniques to differentiate relapse
from new infections. Imprecision: no serious. The upper and lower limits of the 95% CI of the pooled relative risk
indicate appreciable benefit with chloroquine + primaquine for 14 days. The total number of events was < 300, but
the total sample size was larger than the optimal information size, given the magnitude of risk reduction.
Attached Images
415 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Comparator Intervention
Outcome Study results and the Evidence Plain language
7 days 14 days
Timeframe measurements (Quality of summary
primaquine primaquine
evidence)
P. vivax relapse
defined as
420 189
per 1000 per 1000
reappearance
Relative risk 0.45
of P. vivax Low
(CI 95% 0.25 — 0.81) Difference: 231 fewer per
parasitaemia > Due to serious
Based on data from 1000
30 days after indirectness and
126 participants in 1 ( CI 95% 315
starting serious
studies. (Randomized fewer — 80
primaquine fewer ) imprecision 1
controlled)
1. Indirectness: serious. The trial authors did not include children < 15 years. Another trial in the same area by the
same group of investigators immediately afterwards included children. The results for 3 days of primaquine versus 14
days of primaquine did not differ in children from that in adults. Duration of follow-up was 2 months. While this
ensures detection of early relapse, it does not cover relapses after 2 months. The relapse rates at 6 months showed
that most relapses occur by 2 months. The effects of 7 days of primaquine were assessed in only one trial. We
therefore downgraded the evidence by 1. Imprecision: serious. Although the upper and lower limits of the 95% CI of
the risk ratio in this trial showed statistically significant, clinically appreciable benefit with 14 days of primaquine
over 7 days of primaquine, the total number of events was 38 and the sample size of the trial was 104. This is lower
than the optimal information size. We downgraded the evidence by 1.
Attached Images
416 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention
Comparator Certainty of
0.5 mg/kg/
Outcome Study results and Standard the Evidence Plain language
day
Timeframe measurements 14-day course (Quality of summary
primaquine
of primaquine evidence)
for 7 days
Recurrence of 84 81
P. vivax Relative risk 0.96 per 1000 per 1000 There may be little or
Low
parasitaemia 6 (CI 95% 0.66 — 1.39) no difference between
Due to serious
to 7 months of Based on data from 0.5 mg/kg/day
Difference: 3 fewer per risk of bias, Due
follow-up 1,211 participants in 4 primaquine for 7 days
1000 to serious
studies. 1 (Randomized ( CI 95% 29
and the standard
imprecision 2 14-day course.
controlled) fewer — 33 more
)
Serious
adverse events Not estimable. No
Based on data from
events reported.
1,427 participants in 5
studies. 3
We do not know if
Adverse events 3 3 there is any difference
Relative risk 1.04 per 1000 per 1000
that result in Very low in adverse events that
(CI 95% 0.15 — 7.38)
discontinuation Due to serious result in treatment
Based on data from Difference: 0 fewer per
of treatment risk of bias, Due discontinuation
1,427 participants in 5 1000 to very serious between 0.5 mg/kg/
studies. 4 (Randomized ( CI 95% 3 fewer imprecision 5 day primaquine for 7
controlled) — 19 more ) days and the standard
14-day course.
1. Systematic review [229] with included studies: Pareek 2015 IND, Abdon 2001 BRA, Durand 2014 PER, Rajgor
2014 IND. Baseline/comparator: Control arm of reference used for intervention.
2. Risk of Bias: serious. One study, which contributed the most weight to the meta-analysis, was at high risk of
selection bias due to no allocation concealment and high risk of attrition bias. Although another study was at risk of
selection bias as well as other bias for being funded and carried out by drug company, it only contributed a small
amount of weight to the meta-analysis.. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious.
Wide CIs - may be 34% reduction in malaria recurrences or 40% increase with 0.5 mg/kg/day primaquine for seven
days..
3. Systematic review [229] with included studies: Durand 2014 PER, Abdon 2001 BRA, Solari-Soto 2002 PER,
Rajgor 2014 IND, Pareek 2015 IND. Baseline/comparator: Control arm of reference used for intervention.
4. Systematic review [229] with included studies: Rajgor 2014 IND, Solari-Soto 2002 PER, Durand 2014 PER,
Pareek 2015 IND, Abdon 2001 BRA. Baseline/comparator: Control arm of reference used for intervention.
5. Risk of Bias: serious. One study was at high risk of selection bias due to no allocation concealment and high risk
of attrition bias. Another study was at risk of selection bias as well as other bias for being funded and carried out by
drug company.. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Very few events (only
four events occurring in one trial), very wide CIs..
6. Systematic review [229] with included studies: Pareek 2015 IND. Baseline/comparator: Control arm of reference
used for intervention.
417 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
7. Risk of Bias: serious. One study was at risk of selection bias and other bias (funded and performed by drug
company).. Inconsistency: no serious. Indirectness: serious. Only one study that excluded G6PD-deficient adults
measured this safety outcome.. Imprecision: very serious. Only one event (in the 0.5 mg/kg/day primaquine for
seven days group), very wide CIs..
Attached Images
References
229. Milligan R, Daher A, Villanueva G, Bergman H, Graves P : Primaquine alternative dosing schedules for
preventing malaria relapse in people with Plasmodium vivax. Cochrane Database Syst Rev 2020; Pubmed Journal
Comparator Intervention
Certainty of
high-standard 1.0 mg/kg/
Outcome Study results and the Evidence Plain language
0.5 mg/kg/ day
Timeframe measurements (Quality of summary
day for 14 primaquine
evidence)
days for 7 days
Recurrence of
P. vivax
104 107 There is probably little
Relative risk 1.03 per 1000 per 1000
parasitaemia (CI 95% 0.82 — 1.3)
or no difference
with 12 Moderate between 1.0 mg/kg/
Based on data from Difference: 3 more per 1000
months of Due to serious day primaquine for 7
2,526 participants in 2 ( CI 95% 19
follow-up risk of bias 2 days and the high-
studies. 1 (Randomized fewer — 31 more standard 0.5 mg/kg/
controlled) ) day for 14 days course.
There may be a
Serious 1 13 moderate to large
Relative risk 12.03 per 1000 per 1000
adverse events Low increase in serious
(CI 95% 1.57 — 92.3)
- Up to 42 days Due to serious adverse events in the
Based on data from Difference: 11 more per
follow-up indirectness, 1.0 mg/kg/day
1,872 participants in 1 1000 Due to serious primaquine for 7 days
studies. 3 (Randomized ( CI 95% 1 more imprecision 4 compared with the
controlled) — 91 more ) high-standard 0.5 mg/
kg/day.
We do not know if
Adverse events
Relative risk 2.5 2 5 Very low
there is any difference
that resulted in (CI 95% 0.49 — 12.87) per 1000 per 1000 Due to serious
in adverse events
discontinuation risk of bias, Due
Based on data from resulting in treatment
of treatment Difference: 3 more per 1000 to serious
2,526 participants in 2 discontinuation
( CI 95% 1 fewer indirectness,
studies. 5 (Randomized Due to serious
between 1.0 mg/kg/
controlled) — 24 more ) day primaquine for 7
imprecision 6 days and the high-
418 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Comparator Intervention
Certainty of
high-standard 1.0 mg/kg/
Outcome Study results and the Evidence Plain language
0.5 mg/kg/ day
Timeframe measurements (Quality of summary
day for 14 primaquine
evidence)
days for 7 days
1. Systematic review [229] with included studies: Taylor 2019 MULTI, Chu 2019 THA, Taylor 2019 MULTI, Chu
2019 THA. Baseline/comparator: Control arm of reference used for intervention.
2. Risk of Bias: serious. One study was an open-label trial with high risk of performance and detection bias;
although drop-outs were balanced between groups the proportion of drop-outs after one year was high in both trials
(30-40%).. Inconsistency: no serious. Indirectness: no serious. Imprecision: no serious.
3. Systematic review [229] with included studies: Taylor 2019 MULTI. Baseline/comparator: Control arm of
reference used for intervention.
4. Inconsistency: no serious. Indirectness: serious. G6PD-deficient children and adults were excluded from the two
trials that measured this outcome.. Imprecision: serious. Few events..
5. Systematic review [229] with included studies: Taylor 2019 MULTI, Chu 2019 THA. Baseline/comparator:
Control arm of reference used for intervention.
6. Risk of Bias: serious. One study was an open-label trial with high risk of performance and detection bias;
although drop-outs were balanced between groups the proportion of drop-outs after one year was high in both trials
(30-40%).. Inconsistency: no serious. Indirectness: serious. G6PD-deficient children and adults were excluded from
the two trials that measured this outcome . Imprecision: serious. Few events..
7. Systematic review [229] with included studies: Taylor 2019 MULTI, Chu 2019 THA. Baseline/comparator:
Control arm of reference used for intervention.
8. Risk of Bias: serious. One study was an open-label trial with high risk of performance and detection bias;
although drop-outs were balanced between groups the proportion of drop-outs after one year was high in both trials
(30-40%).. Inconsistency: no serious. Indirectness: serious. G6PD-deficient children and adults were excluded from
the two trials that measured this outcome. Imprecision: serious.
Attached Images
References
229. Milligan R, Daher A, Villanueva G, Bergman H, Graves P : Primaquine alternative dosing schedules for
preventing malaria relapse in people with Plasmodium vivax. Cochrane Database Syst Rev 2020; Pubmed Journal
419 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Chloroquine
Timeframe measurements Placebo (Quality of summary
prophylaxis
evidence)
70 1
Relative risk 0.02 per 1000 per 1000
P. vivax (CI 95% 0 — 0.26)
parasitaemia Moderate
Based on data from
Difference: 69 fewer per Due to serious
951 participants in 1
1000 imprecision 1
studies. (Randomized ( CI 95% 70
controlled) fewer — 52
fewer )
Severe
anaemia in CI 95%
Relative risk
third trimester
Adverse events
Relative risk CI 95%
1. Risk of Bias: no serious. This study had a low risk of bias in all domains. Indirectness: no serious. This study was
conducted in Thailand between 1998 and 2001. Chloroquine was administered as four tablets at enrolment,
followed by two tablets once a week until delivery. Imprecision: serious. Although the intervention appeared to
prevent all episodes of P. vivax malaria, there were few events, even in the control group.
2. Risk of Bias: no serious. This study had a low risk of bias in all domains. Indirectness: no serious. This study was
conducted in Thailand between 1998 and 2001. Chloroquine was administered as four tablets at enrolment,
followed by two tablets once a week until delivery. Imprecision: serious. The finding of a small clinical benefit did not
reach statistical significance.
Attached Images
420 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
5.2.2.1. Artesunate
421 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements Quinine Artesunate (Quality of summary
evidence)
109 83
Relative risk 0.76 per 1000 per 1000
Death (CI 95% 0.65 — 0.9)
Based on data from
Difference: 26 fewer per High
5,765 participants in 4 1
1000
studies. (Randomized ( CI 95% 38
controlled) fewer — 11
fewer )
30 19
Relative risk 0.62 per 1000 per 1000
Hypoglycaemia (CI 95% 0.45 — 0.87)
episodes Based on data from High
Difference: 11 fewer per
5,765 participants in 4 4
1000
studies. (Randomized ( CI 95% 16
controlled) fewer — 4 fewer
)
1. Risk of Bias: no serious. All the trials adequately concealed allocation and can be considered at low risk of bias.
The trials were unblinded, but this is unlikely to have biased this objective outcome. Inconsistency: no serious. There
was no statistical heterogeneity between the trials (I² = 0%). Indirectness: no serious. Most of the data are from the
single multicentre trial with centres in the Democratic Republic of Congo, the Gambia, Ghana, Kenya, Mozambique,
Nigeria, Rwanda, Uganda and the United Republic of Tanzania, where the established, standard doses of artesunate
and quinine (with loading dose) were used. The median age of children in this trial was 2.9 years in the quinine group
and 2.8 in the artesunate group. Imprecision: no serious. Both limits of the 95% CI of the pooled effect imply an
appreciable clinical benefit with artesunate. The number of people who must be treated to prevent one childhood
death is 38.
2. Risk of Bias: serious. 41/170 (24%) patients with neurological sequelae at discharge were not available for
assessment at day 28. Indirectness: no serious. This trial was conducted in 11 centres in Africa, with standard dosing
422 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
of artesunate and quinine. The nature of the neurological sequelae is not described. Imprecision: no serious. The
95% CI around the absolute effect is narrow. The worst-case scenario is a 1.2% increase in neurological sequelae at
day 28.
3. Risk of Bias: no serious. All the trials adequately concealed allocation and can be considered at low risk of bias.
The trials were unblinded, but this is unlikely to have biased this objective outcome. Inconsistency: no serious. There
was no statistical heterogeneity between the trials (I² = 0%). Indirectness: no serious. Most of the data are from the
single multicentre trial with centres in the Democratic Republic of Congo, the Gambia, Ghana, Kenya, Mozambique,
Nigeria, Rwanda, Uganda and the United Republic of Tanzania, where the established, standard doses of artesunate
and quinine (with loading dose) were used. The median age of children in this trial was 2.9 years in the quinine group
and 2.8 in the artesunate group. Imprecision: serious. The effect estimate indicates clinically important harm;
however, the 95% CI includes the possibility of no clinically important difference between the two interventions.
4. Risk of Bias: no serious. All the trials adequately concealed allocation and can be considered at low risk of bias.
The trials were unblinded, but this is unlikely to have biased this objective outcome. Inconsistency: no serious. There
was no statistical heterogeneity between the trials (I² = 0%). Indirectness: no serious. Most of the data are from the
single multicentre trial with centres in the Democratic Republic of Congo, the Gambia, Ghana, Kenya, Mozambique,
Nigeria, Rwanda, Uganda and the United Republic of Tanzania, where the established, standard doses of artesunate
and quinine (with loading dose) were used. The median age of children in this trial was 2.9 years in the quinine group
and 2.8 in the artesunate group. Imprecision: no serious. The result is statistically significantly in favour of
artesunate. The sample size is adequate to detect a 40% risk reduction with 80% power and 95% confidence.
5. Risk of Bias: no serious. All the trials adequately concealed allocation and can be considered at low risk of bias.
The trials were unblinded, but this is unlikely to have biased this objective outcome. Inconsistency: no serious. None
of the trials found evidence of a large difference between the two treatment groups. Indirectness: no serious. Most
of the data are from the single multicentre trial with centres in the Democratic Republic of Congo, the Gambia,
Ghana, Kenya, Mozambique, Nigeria, Rwanda, Uganda and the United Republic of Tanzania, where the established,
standard doses of artesunate and quinine (with loading dose) were used. The median age of children in this trial was
2.9 years in the quinine group and 2.8 in the artesunate group. Imprecision: serious. We were unable to pool the
data as they were reported only as medians and range or intraquartile range. There is no evidence of a clinically
important benefit with artesunate on this outcome.
Attached Images
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements Quinine Artesunate (Quality of summary
evidence)
241 147
Relative risk 0.61 per 1000 per 1000
Death (CI 95% 0.5 — 0.75)
Based on data from
Difference: 94 fewer per High
1,664 participants in 5 1
1000
studies. (Randomized ( CI 95% 120
controlled) fewer — 60
fewer )
423 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements Quinine Artesunate (Quality of summary
evidence)
30 19
Relative risk 0.62 per 1000 per 1000
Hypoglycaemia (CI 95% 0.45 — 0.87)
episodes Based on data from High
Difference: 11 fewer per
5,765 participants in 4 3
1000
studies. (Randomized ( CI 95% 16
controlled) fewer — 4 fewer
)
1. Risk of Bias: no serious. Two of the smaller studies did not conceal allocation, and none of the studies was
blinded; however, most data are from studies in which allocation was concealed, and the lack of blinding is unlikely to
introduce bias for an objective outcome such as death. Inconsistency: no serious. The point estimates of all five trials
favoured artesunate. No significant statistical heterogeneity was detected (I² = 0%). Indirectness: no serious. All five
trials were conducted in Asia but in a variety of settings (Bangladesh, India, Indonesia, Myanmar, Thailand and Viet
Nam), and included age groups > 15–16 years. Of the four small trials, two did not give the loading dose of quinine,
but there was no statistical heterogeneity between these two trials and the large multicentre trial, in which the
loading dose was given. Imprecision: no serious. Both limits of the 95% CI imply a clinically important benefit with
artesunate.
2. Risk of Bias: no serious. This trial was unblinded, but the nature of the sequelae makes observer or reporting bias
unlikely. Inconsistency: no serious. Not applicable, as only one trial. Indirectness: no serious. This trial was
conducted in sites in four countries in Asia with the standard doses of artesunate and quinine (with loading dose). Of
the 10 sequelae that occurred in this trial (the additional two were in children), five were psychiatric sequelae, four
were a persistent problem with balance, and two were hemiparesis. Imprecision: serious. Neurological sequelae
appear to be rare after severe malaria in adults; however, the 95% CI includes the possibility of clinically important
harm with artesunate.
3. Risk of Bias: no serious. The large multicentre study adequately concealed allocation and can be considered at
low risk of bias. The smaller trial did not. Neither trial was blinded. Inconsistency: no serious. There was no statistical
heterogeneity (I² = 0%). Indirectness: no serious. This evidence is from multiple sites in Asia (Bangladesh, India,
Indonesia and Myanmar), and both trials used standard drug doses. Imprecision: no serious. This result is statistically
significantly in favour of artesunate. The sample size was adequate to detect a 75% risk reduction with 80% power
and 95% confidence..
424 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
4. Risk of Bias: no serious. The large multicentre study adequately concealed allocation and can be considered at
low risk of bias. The smaller trial did not. Neither trial was blinded. Inconsistency: no serious. Neither trial found a
statistically significant difference in time to hospital discharge. Indirectness: no serious. This evidence is from
multiple sites in Asia (Bangladesh, India, Indonesia and Myanmar), and both trials used standard drug doses.
Imprecision: serious. We were unable to pool data because of the way in which they were presented, but there is no
evidence of a benefit on this outcome with artesunate.
Attached Images
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements Artesunate Artemether (Quality of summary
evidence)
148 81
Relative risk 0.55 per 1000 per 1000
Death (CI 95% 0.34 — 0.92)
Moderate
Based on data from
Difference: 67 fewer per Due to serious
494 participants in 2
1000 imprecision 1
studies. (Randomized ( CI 95% 98
controlled) fewer — 12
fewer )
Neurological
sequelae at CI 95%
Relative risk
discharge
425 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements Artesunate Artemether (Quality of summary
evidence)
1. Risk of Bias: no serious. The trials were generally well conducted and had a low risk of bias. Inconsistency: no
serious. There is no statistical heterogeneity. Indirectness: no serious. The two studies were conducted in Thailand
and Viet Nam; both compared intramuscular artemether with intravenous artesunate in adults. Imprecision: serious.
These trials and the meta-analysis have inadequate power to detect a difference in mortality or to prove equivalence.
2. Risk of Bias: no serious. The trials were generally well conducted and had a low risk of bias. Inconsistency: no
serious. Both studies suggest an advantage with artesunate, although this was statistically significant only in the
small trial. Indirectness: no serious. The two studies were conducted in Thailand and Viet Nam; both compared
intramuscular artemether with intravenous artesunate in adults. Imprecision: serious. These data could not be
pooled.
3. Risk of Bias: no serious. The trials were generally well conducted and had a low risk of bias. Inconsistency: no
serious. Neither study found a difference between treatments. Indirectness: no serious. The two studies were
conducted in Thailand and Viet Nam; both compared intramuscular artemether with intravenous artesunate in
adults. Imprecision: serious. These data could not be pooled.
4. Risk of Bias: no serious. The trials were generally well conducted and had a low risk of bias. Inconsistency: no
serious. One trial found no statistically significant difference, and the other, small trial found a benefit with
artesunate. Indirectness: no serious. The two studies were conducted in Thailand and Viet Nam; both compared
intramuscular artemether with intravenous artesunate in adults. Imprecision: serious. These data could not be
pooled.
Attached Images
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements Quinine Artemether (Quality of summary
evidence)
426 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements Quinine Artemether (Quality of summary
evidence)
220 185
Neurological Relative risk 0.84 per 1000 per 1000
sequelae at (CI 95% 0.66 — 1.07) Low
discharge Based on data from Due to very
Difference: 35 fewer per
968 participants in 7 serious
1000
studies. (Randomized ( CI 95% 75 imprecision 2
controlled) fewer — 15 more
)
1. Risk of Bias: no serious. Various risks of bias, but exclusion of trials with high or unclear risk of selection bias did
not change this result. Inconsistency: no serious. None of the individual trials found statistically significant effects,
and there was no statistical heterogeneity between trials. Indirectness: no serious. Trials were conducted in East and
West Africa and India. All were in children with severe malaria (aged < 15 years), and most compared the standard
dose of intramuscular artemether with the WHO recommended dose of intravenous quinine. Imprecision: serious.
These trials and the meta-analysis had inadequate power to detect a difference or to prove equivalence.
2. Risk of Bias: no serious. Various risks of bias, but exclusion of trials with high or unclear risk of selection bias did
not change this result. Inconsistency: no serious. None of the individual trials found statistically significant effects,
and there was no statistical heterogeneity between trials. Indirectness: no serious. Trials were conducted in East and
West Africa and India. All were in children with severe malaria (aged < 15 years), and most compared the standard
dose of intramuscular artemether with the WHO recommended dose of intravenous quinine. Imprecision: very
serious. These trials and the meta-analysis have inadequate power to detect a difference or to prove equivalence.
The 95% CI is very wide and includes clinically important differences and no effect.
3. Risk of Bias: very serious. Four of the six trials had unclear risk of selection bias. When these four trials are
excluded, the result becomes nonsignificant. Inconsistency: no serious. Statistically significant differences were seen
in only two of the six trials; however, statistical heterogeneity between trials was low, and the result of the meta-
analysis is significant. Indirectness: no serious. Trials were conducted in East and West Africa and India. All were in
children with severe malaria (aged < 15 years), and most compared the standard dose of intramuscular artemether
with the WHO recommended dose of intravenous quinine. Imprecision: no serious. The result is statistically
significant, and the meta-analysis has adequate power to detect this effect.
4. Risk of Bias: no serious. Various risks of bias, but exclusion of trials with high or unclear risk of selection bias did
not change this result. Inconsistency: serious. The mean difference in parasite clearance time ranged from a 2 h
increase with artemether to a 15 h decrease. Indirectness: no serious. Trials were conducted in East and West Africa
and India. All were in children with severe malaria (aged < 15 years), and most compared the standard dose of
427 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
intramuscular artemether with the WHO recommended dose of intravenous quinine. Imprecision: no serious. The
result is statistically significant, and the meta-analysis has adequate power to detect this effect.
5. Risk of Bias: serious. Four of the seven trials had unclear risks of selection bias. When these four trials are
excluded, the result becomes nonsignificant. Inconsistency: serious. The mean difference in fever clearance time
ranged from a 25 h increase with artemether to an 18 h decrease. Indirectness: no serious. Trials were conducted in
East and West Africa and India. All were in children with severe malaria (aged < 15 years), and most compared the
standard dose of intramuscular artemether with the WHO recommended dose of intravenous quinine. Imprecision:
no serious. The meta-analysis has adequate power to detect this effect. The result is statistically significant but may
not be clinically important.
Attached Images
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements Quinine Artemether (Quality of summary
evidence)
208 123
Relative risk 0.59 per 1000 per 1000
Death (CI 95% 0.42 — 0.83)
Moderate
Based on data from
Difference: 85 fewer per Due to serious
716 participants in 4
1000 imprecision 1
studies. (Randomized ( CI 95% 121
controlled) fewer — 35
fewer )
428 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Outcome Study results and Comparator Intervention the Evidence Plain language
Timeframe measurements Quinine Artemether (Quality of summary
evidence)
1. Risk of Bias: no serious. The trials were generally well conducted and with low risk of bias. Inconsistency: no
serious. Statistically significant differences were seen in only one of the four studies; however, statistical
heterogeneity among the trials was low, and the results of the meta-analysis are statistically significant. Indirectness:
no serious. All four trials compared intramuscular artemether with intravenous quinine in adults: two studies in
Thailand, one each in Papua New Guinea and Viet Nam. Imprecision: serious. These trials and the meta-analysis had
inadequate power to detect a difference in mortality or to prove equivalence.
2. Risk of Bias: no serious. This single trial had a low risk of bias. Imprecision: serious. Neurological sequelae in
adults were uncommon. This trial had inadequate power to detect or exclude clinically important differences.
3. Risk of Bias: no serious. The trials were generally well conducted and with low risk of bias. Inconsistency:
serious. One trial found a shorter median coma resolution time with quinine, and one trial found no difference; the
third trial reported mean coma recovery time incompletely. Imprecision: serious. The data could not be pooled.
4. Risk of Bias: no serious. The trials were generally well conducted and with low risk of bias. Inconsistency: no
serious. The two largest studies both found shorter median clearance times with artemether. Indirectness: no
serious. All four trials compared intramuscular artemether with intravenous quinine in adults: two studies in Thailand,
one each in Papua New Guinea and Viet Nam. Imprecision: serious. The data could not be pooled.
5. Risk of Bias: no serious. The trials were generally well conducted and with low risk of bias. Inconsistency: no
serious. One trial found a shorter median fever clearance time with quinine, and two trials found a shorter time with
artemether. Indirectness: no serious. All four trials compared intramuscular artemether with intravenous quinine in
adults: two studies in Thailand, one each in Papua New Guinea and Viet Nam. Imprecision: serious. The data could
not be pooled.
Attached Images
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Rectal
Timeframe measurements Placebo (Quality of summary
artesunate
evidence)
All-cause
mortality (in
Relative risk 0.44
(CI 95% 0.23 — 0.82)
31 14 Low
Due to serious
per 1000 per 1000
Asia) Based on data from inconsistency
429 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Rectal
Timeframe measurements Placebo (Quality of summary
artesunate
evidence)
All-cause
Relative risk 0.81 44 36
mortality (in per 1000 per 1000 Low
(CI 95% 0.63 — 1.04)
Africa) Due to serious
Based on data from
7-30 days Difference: 8 fewer per inconsistency
6,040 participants in 1
1000 and serious
studies. (Randomized
( CI 95% 16 imprecision 2
controlled)
fewer — 2 more )
All-cause 41 30
Relative risk 0.74 per 1000 per 1000
mortality (CI 95% 0.59 — 0.93)
(overall) Moderate
Based on data from
7-30 days Difference: 11 fewer per Due to serious
8,050 participants in 1
1000 inconsistency 3
studies. (Randomized ( CI 95% 17
controlled) fewer — 3 fewer
)
1. Risk of Bias: no serious. Allocation was concealed, and trial participants and staff were blinded to treatment
allocation. Inconsistency: serious. In Asia, older children and adults were also randomized to artesunate or placebo,
and mortality was significantly higher in those given rectal artesunate; the cause is unclear. Indirectness: no serious.
This trial was conducted in community settings in Bangladesh, Ghana and the United Republic of Tanzania.
Imprecision: serious. The number of events was low.
2. Risk of Bias: no serious. Allocation was concealed, and trial participants and staff were blinded to treatment
allocation. Inconsistency: serious. In Asia, older children and adults were also randomized to artesunate or placebo,
and mortality was significantly higher in those given rectal artesunate; the cause is unclear. Indirectness: no serious.
This trial was conducted in community settings in Bangladesh, Ghana and the United Republic of Tanzania.
Imprecision: serious. The 95% confidence interval is wide and includes no difference.
3. Risk of Bias: no serious. Allocation was concealed, and trial participants and staff were blinded to treatment
allocation. Inconsistency: serious. In Asia, older children and adults were also randomized to artesunate or placebo,
and mortality was significantly higher in those given rectal artesunate; the cause is unclear. Indirectness: no serious.
This trial was conducted in community settings in Bangladesh, Ghana and the United Republic of Tanzania.
Imprecision: no serious. The result is statistically significant, and the study had adequate power to detect this effect.
Attached Images
430 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Rectal
Timeframe measurements Placebo (Quality of summary
artesunate
evidence)
1. Risk of Bias: no serious. Allocation was concealed, and trial participants and staff were blinded to treatment
allocation. Inconsistency: serious. Rectal artesunate appears beneficial in children < 5 years and harmful in older
children and adults. This finding is difficult to explain. Indirectness: no serious. This trial was conducted in a single
setting in Bangladesh. Imprecision: serious. There were few deaths in adults in this trial: 31/2009 in treated and 14/
2009 in controls.
Attached Images
431 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Summary
Seven studies of MTaT were included in the systematic review: four cRCTs, conducted in Kenya, Indonesia, Zambia
and Burkina Faso; and three NRSs in Senegal, Ghana and India (Bhamani et al unpublished evidence).
All four of the cRCTs conducted 2–3 rounds of MTaT over a period of up to one year, with the exception of the study
in Kenya that carried out six rounds of MTaT over two years. The studies in Kenya and Burkina Faso were conducted
in areas of moderate to high transmission while those in Indonesia and Zambia were areas of low transmission. The
overall risk of bias for community-level outcomes in these studies was low. Meta-analyses of the results found little
to no reductions in community-level incidence or prevalence of infection. However, there was a small reduction of
the incidence of clinical malaria found in two studies.
The certainty of evidence from the NRSs was GRADEd as very low.
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Mass testing
Timeframe measurements No MTaT (Quality of summary
and treatment
evidence)
2,331 2,214
0 - 12 months - Rate ratio 0.95 per 1000 per 1000
MTaT does not reduce
Incidence (CI 95% 0.87 — 1.04)
Difference: 117 fewer per incidence of malaria
Based on data from 857 High
1000 infection between 0-12
participants in 1 studies.
months.
(Randomized controlled) ( CI 95% 303
fewer — 93 more
)
233 189
0 - 12 months - Relative risk 0.81 per 1000 per 1000
Incidence of (CI 95% 0.7 — 0.95) MTaT reduces the
clinical malaria Based on data from incidence of clinical
Difference: 44 fewer per High
332,454 participants in malaria between 0-12
1000
2 studies. (Randomized months.
( CI 95% 70
controlled) fewer — 12 fewer
)
432 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Mass testing
Timeframe measurements No MTaT (Quality of summary
and treatment
evidence)
Adverse event
(group targeted
5 Low
per 1000 The evidence is very
by the Due to serious
Based on data from uncertain about the
intervention) indirectness, and
6,373 participants in 1 effect of MTaT on
serious
studies. (Randomized adverse events.
imprecision 2
controlled)
Serious adverse
event (SAE)
Low
(group targeted Based on data from The evidence is very
Due to serious
by the 6,373 participants in 1 uncertain about the
indirectness, and
intervention) studies. (Randomized effect of MTaT on
serious
controlled) serious adverse events.
imprecision 3
Follow up: not
estimable.
9 months -
Prevalence Relative risk 0.91
378 344 MTaT likely does not
per 1000 per 1000
(group targeted (CI 95% 0.82 — 1.01) reduce the prevalence
Moderate
by the Based on data from of infection at nine
Difference: 34 fewer per Due to serious
intervention) 2,838 participants in 1 months among the
1000 imprecision 5 group targeted by the
studies. (Randomized
controlled) ( CI 95% 68 intervention.
fewer — 4 more )
2 months -
Prevalence Odds ratio 0.03
34 1 Very low The evidence is very
per 1000 per 1000 Due to serious uncertain about the
(group targeted (CI 95% 0.02 — 0.07)
inconsistency, effect of MTaT on the
by the Based on data from
serious prevalence of infection
intervention) 8,508 participants in 1
indirectness, and at two months in the
studies. (Observational serious group receiving the
(non-randomized))
imprecision 6 intervention.
12 months -
Prevalence
Odds ratio 0.91
(CI 95% 0.67 — 1.38)
438 415 Very low
Due to serious
The evidence is very
uncertain about the
per 1000 per 1000
(group targeted Based on data from 416 inconsistency, effect of MTaT on the
433 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Mass testing
Timeframe measurements No MTaT (Quality of summary
and treatment
evidence)
1. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Used as a proxy for incidence of infection
at the community level.
2. Inconsistency: no serious. Indirectness: serious. SAEs and AEs are not classified based on intervention and control
arms; unable to calculate control measures in absence of control measure. Imprecision: serious. SAEs and AEs are not
classified based on intervention and control arms; unable to calculate control measures in absence of control measure.
3. Inconsistency: no serious. Indirectness: serious. SAEs and AEs are not classified based on intervention and control
arms; unable to calculate control measures in absence of control measure. Imprecision: serious. SAEs and AEs are not
classified based on intervention and control arms; unable to calculate control measures in absence of control measure.
4. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Used as a proxy for prevalence of infection
at the community level.
5. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Used as a proxy for prevalence of infection
at the community level.
6. Inconsistency: serious. Study did not control for one confounding domain and missing register from health facility in
intervention village - the analysis is unlikely to have removed the risk of bias arising from the missing data. Indirectness:
serious. Study did not control for one confounding domain and missing register from health facility in intervention village
- the analysis is unlikely to have removed the risk of bias arising from the missing data. Imprecision: serious. Study did
not control for one confounding domain and missing register from health facility in intervention village - the analysis is
unlikely to have removed the risk of bias arising from the missing data.
434 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
7. Inconsistency: serious. Critical overall risk of bias due to inherent biases associated with study design. Indirectness:
serious. Critical overall risk of bias due to inherent biases associated with study design. Imprecision: serious. Critical
overall risk of bias due to inherent biases associated with study design.
8. Inconsistency: serious. Used as a proxy for prevalence of infection at the community level; critical overall risk of bias
due to inherent biases associated with study design . Indirectness: serious. Used as a proxy for prevalence of infection at
the community level; critical overall risk of bias due to inherent biases associated with study design. Imprecision: serious.
Used as a proxy for prevalence of infection at the community level; critical overall risk of bias due to inherent biases
associated with study design.
9. Risk of Bias: serious. Common AEs are reported for the whole study, however no break-up is provided for different
arms. Imprecision: serious. Common AEs are reported for the whole study, however no break-up is provided for different
arms.
10. Risk of Bias: serious. Critical overall risk of bias due to inherent biases associated with study design.
Attached Images
Summary
No studies from areas approaching elimination were identified in the systematic review (Tusell et al unpublished
evidence). Two studies conducted in post-elimination settings identified imported infections in migrant workers with
onward transmission to the local population. In both studies, the migrant workers were provided with a full
therapeutic dose of chloroquine and 14 days of primaquine in a single round (the study from Greece conducted one
round per year for three years). No additional infections among the migrant workers or the community were
identified for five months (Sri Lanka) or two years (Greece) after the last round of TDA. Adverse events were
monitored in both studies: a single serious case of haemolysis was identified in the study from Greece due to an
incorrect G6PD test result; the remaining adverse events were relatively minor side effects.
Intervention Certainty of
Outcome Study results and Comparator Targeted drug the Evidence Plain language
Timeframe measurements no TDA administration (Quality of summary
(TDA) evidence)
435 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Targeted drug the Evidence Plain language
Timeframe measurements no TDA administration (Quality of summary
(TDA) evidence)
Very low
Serious Adverse 0 0 Due to very The evidence is very
Events (cRCTs) Based on data from per 1000 per 1000 serious risk of uncertain about the
10,079 participants in 1 bias, Due to very effect of TDA on
studies. (Randomized serious serious adverse events
controlled) imprecision 1
Serious adverse
Relative risk 4.19 2 7
(CI 95% 1.43 — 12.31) per 1000 per 1000 Low
events (cRCTs) TDA may result in little
Based on data from Due to very
to no difference in
4,916 participants in 1 serious
Difference: 6 more per 1000 serious adverse events
studies. (Randomized ( CI 95% 1 more imprecision 2
controlled) — 23 more )
Prevalence 406 61
among those Relative risk 0.15 per 1000 per 1000 TDA probably reduces
targeted by the (CI 95% 0.06 — 0.38)
Moderate the prevalence of
intervention Based on data from
Difference: 345 fewer per Due to serious malaria among those
(cRCTs) 5,970 participants in 2
1000 indirectness 6 targeted by the
studies. (Randomized ( CI 95% 381 intervention
controlled) fewer — 251
fewer )
436 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Targeted drug the Evidence Plain language
Timeframe measurements no TDA administration (Quality of summary
(TDA) evidence)
1. Risk of Bias: very serious. Outcome was collected in intervention arm only. Inconsistency: no serious. Indirectness:
no serious. Imprecision: very serious. Unable to calculate effect measure in absence of control measures. Publication
bias: no serious.
2. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Wide confidence intervals. Publication
bias: no serious.
3. Risk of Bias: serious. Critical overall risk of bias due to inherent biases associated with study design. Inconsistency:
no serious. Indirectness: no serious. Imprecision: serious. Few patients and few events. Publication bias: no serious.
4. Inconsistency: no serious. Indirectness: no serious. Imprecision: very serious. Wide confidence intervals. Publication
bias: no serious.
5. Risk of Bias: serious. Critical overall risk of bias due to inherent biases associated with study design. Inconsistency:
no serious. Indirectness: no serious. Imprecision: no serious. Publication bias: no serious.
6. Inconsistency: no serious. Indirectness: serious. Used as a surrogate for prevalence of infection at the community
level. Imprecision: no serious. Publication bias: no serious.
7. Risk of Bias: serious. Moderate risk of bias due to bias due to confounding, bias due to deviations from intended
interventions, and no information about bias in measurement of outcomes. Inconsistency: no serious. Indirectness:
serious. Used as a surrogate for prevalence of infection at the community level. Imprecision: no serious. Publication bias:
no serious.
8. Risk of Bias: serious. Critical overall risk of bias due to inherent biases associated with study design. Inconsistency:
no serious. Indirectness: serious. Used as a surrogate for prevalence of infection at the community level. Imprecision:
serious. Few patients and few events. Publication bias: no serious.
Attached Images
Summary
The systematic review identified three studies for inclusion: two cRCTs in Ghana and Kenya and one NRS in Malawi
(Allen et al unpublished evidence). No studies were conducted in very low to low transmission or post-elimination
settings. The GDG determined that the TTaT strategy would be most relevant in very low to low transmission or
post-elimination settings and, therefore, decided that the PICO question should be modified and the setting limited
to such areas. As a result, the GDG did not consider any of the studies identified by the systematic review to fit the
revised PICO.
437 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Targeted the Evidence Plain language
Timeframe measurements No TTaT testing and (Quality of summary
treatment evidence)
0 - 24 months -
Adverse events
45 TTaT likely results in
per 1000
(group targeted Based on data from little to no difference in
Moderate
by intervention) 2,030 participants in 1 adverse events among
Due to serious
studies. (Randomized the group targeted by
imprecision 1 intervention between
controlled)
Follow up: not 0-24 months.
estimable.
Mortality
(group targeted
Relative risk 0.73 7 5 TTaT likely results in
(CI 95% 0.08 — 6.95) per 1000 per 1000
by intervention) Moderate little to no difference in
Based on data from
Due to serious severe adverse events
8,222 participants in 2 Difference: 2 fewer per 1000
imprecision 4 among group targeted
studies. (Randomized ( CI 95% 6 fewer by intervention.
controlled) — 42 more )
24 months -
Prevalence Relative risk 1.53
84 129
per 1000 per 1000 TTaT probably results in
(group targeted (CI 95% 0.89 — 2.62)
Moderate little to no difference in
by intervention) Based on data from
Difference: 45 more per Due to serious prevalence in the group
4,140 participants in 1
1000 imprecision 5 targeted by intervention
studies. (Randomized
( CI 95% 9 fewer at 24 months.
controlled)
— 136 more )
1. Risk of Bias: serious. Outcome not measured in control arm. Inconsistency: no serious. Indirectness: no serious.
Imprecision: no serious. Outcome not measured in control arm.
438 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
2. Risk of Bias: serious. High risk of bias for domain 5 of RoB2 assessment - Selection of reported result. Study
assessed incidence of malaria; episodes of malaria and accounted for repeat illnesses, but did not assess number of
children in intervention and control arms that had malaria. Incidence was instead categorized by all episodes, episodes
after first fever and repeat malaria and prevalence or number of clinical cases was not reported. Conducted a multi-level
poisson to calculate incidence and rate ratios for comparison in study arms, but did not perform a generalized model
accounting for potential demographics and confounders to assess risk of malaria infection in study arms. Incidence
among high-risk population within the community used as a surrogate for community level impact. Inconsistency: no
serious. Indirectness: no serious. Incidence among high-risk population within the community used as a surrogate for
community level impact. Imprecision: no serious.
3. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Absolute effect estimates both appreciable
risk and appreciable benefit.
4. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Imprecision due to wide confidence
intervals; crude data used for mortality unadjusted for additional criteria or potential confounders. Absolute effect
estimates both appreciable risk and appreciable benefit.
5. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Absolute effect estimates both appreciable
risk and appreciable benefit.
6. Risk of Bias: serious. Moderate risk of bias in D7 of ROBINS-I, bias in selection of the reported result. Inconsistency:
no serious. Indirectness: no serious. Imprecision: serious. Absolute effect estimates both appreciable risk and
appreciable benefit. Upgrade: large magnitude of effect.
Attached Images
Summary
The systematic review identified seven NRSs in six countries (Cambodia, China, Equatorial Guinea, Greece, Myanmar
and the United Arab Emirates that reported on TTaT at points of entry (Coma-Cros et al unpublished evidence). None
of the studies provided information on the outcome considered critical by the GDG, i.e. the number of positive cases
identified by the strategy as a proportion of all imported cases found in the country during the same period.
Intervention Certainty of
Comparator
Outcome Study results and Testing and the Evidence Plain language
no
Timeframe measurements treatment at (Quality of summary
intervention
points of entry evidence)
439 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Testing and the Evidence Plain language
no
Timeframe measurements treatment at (Quality of summary
intervention
points of entry evidence)
1. Risk of Bias: serious. Observational study. Inconsistency: no serious. Indirectness: no serious. Imprecision: no
serious. Publication bias: no serious.
2. Risk of Bias: serious. Observational study. Inconsistency: serious. Big differences among positivity rates (from 0.0 to
21.0). Indirectness: serious. Outcome expressed in positivity rate no prevalence. Imprecision: no serious. Publication
bias: no serious.
3. Risk of Bias: serious. Observational study. Inconsistency: no serious. Indirectness: no serious. Outcome expressed in
positivity rate no prevalence. Imprecision: no serious. Publication bias: no serious.
Attached Images
440 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Summary
The systematic review identified six cRCTs assessing the impact of RDA in four countries of sub-Saharan Africa
(Eswatini, Gambia, Namibia and Zambia) (Steinhardt et al unpublished evidence (c)). All studies used DP for treatment,
with the exception of the study from Namibia that provided AL. One NRS assessing the impact of RDA was
identified; the study, conducted in Peru, provided chloroquine plus seven days of primaquine at a dosage of 0.5mg/
kg. All studies except for one were from low-transmission settings. Three of the cRCTs compared RDA to no RDA
and three compared RDA to RACDT.
In the cRCTs, the people around the index case included in the RDA programme ranged from household and
compound members (of the index case to people living within 500 meters of the index case.
Evidence of low to moderate certainty from the cRCTs suggested that RDA may reduce malaria prevalence and
incidence slightly but probably results in little to no difference in the incidence of clinical malaria. Adverse events
were often not measured in both arms, which complicated interpretation of the findings, but reported rates of
adverse events or serious adverse events were low.
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Reactive drug
Timeframe measurements No RDA (Quality of summary
administration
evidence)
441 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Certainty of
Intervention
Outcome Study results and Comparator the Evidence Plain language
Reactive drug
Timeframe measurements No RDA (Quality of summary
administration
evidence)
1. Inconsistency: no serious. Indirectness: serious. Two studies (Hsiang 2020 and Okebe 2021) lack a true control
group. The comparison in Hsiang 2020 is RACD and the comparison in Okebe 2021 is a modified version of RACD
(testing and treating symptomatic household members of the index case). Although we rated down for indirectness, any
bias would be towards the null if RACD has an effect on reducing malaria transmission., thus these effect sizes might
underestimate the true effect of RDA. The pooled estimate ranges from averting 9 cases of parasitemia per 1,000 to
having 2 more. Imprecision: serious. Two studies (Hsiang 2020 and Okebe 2021) lack a true control group. The
comparison in Hsiang 2020 is RACD and the comparison in Okebe 2021 is a modified version of RACD (testing and
treating symptomatic household members of the index case). Although we rated down for indirectness, any bias would
be towards the null if RACD has an effect on reducing malaria transmission., thus these effect sizes might underestimate
the true effect of RDA. The pooled estimate ranges from averting 9 cases of parasitemia per 1,000 to having 2 more.
2. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. The pooled effect estimate ranges from
0.36 (substantial benefit of RDA) to 1.47, indicating potential harm of RDA.
3. Inconsistency: no serious. Indirectness: serious. Only two studies (Eisele 2020--LOW and Eisele 2020--HIGH) had a
true control group; three (Bridges 2021, Hsiang 2020, and VIlakati 2021) compared RDA to reactive case detection
(RACD), and the fourth (Okebe 2021) compares RDA to a modified version of RACD (testing and treating symptomatic
household members of the index case). Although we rated down for indirectness, any bias would be towards the null if
RACD has an effect on reducing malaria transmission., thus these effect sizes might underestimate the true effect of
RDA. Imprecision: no serious.
4. Risk of Bias: serious. There is a general lack of information about many aspects of this dissertation using data from
Tumbes, Peru. We rated as 'Some concerns' most aspects of the study and consider the bias overall to be serious.
5. Four randomized trials reported on adverse events (AEs); however, AEs were typically only actively solicited from the
RDA arm and not from the comparison or RACD arm. In the Zambia trial comparing RDA using dihydroartemisinin-
442 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
piperaquine (DP) with RACD using artemether-lumefantrine (AL), 123 (6.9%) mild AEs occurred in 1,775 people treated
with DP (Bridges 2021); all resolved. In the Namibia trial (Hsiang 2020) of RDA with AL compared to RACD with AL, 17
of 4,247 treated participants (0.4%) in the RDA arm experienced an AE versus 1 participant of 98 (1.0%) treated in the
RACD arm; 11 AEs were considered unrelated, 6 possibly related, and 6 probably related. In The Gambia (Okebe 2021),
75 AEs (7.6%) among 979 participants receiving DP in the RDA arm reported AEs; 69 were considered mild and 6
moderate. In Eswati, 68 (3.8%) of 1,776 participants receiving RDA with DP experienced AEs; 54 were rated as mild and
14 as moderate.
Attached Images
Summary
The systematic review identified three cRCTs in three countries of sub-Saharan Africa (Eswatini, Namibia and
Zambia) (Steinhardt et al unpublished evidence (d)). However, all three studies were intended to evaluate the impact of
RDA, and RACDT was the comparator. As RDA is likely to be a more effective strategy than RACDT, no conclusions
could be drawn from these studies. The two NRSs identified from Brazil and Zambia reported on outcomes among
those receiving the intervention, but did not evaluate impact at the community level.
Intervention Certainty of
Outcome Study results and Comparator Reactive case the Evidence Plain language
Timeframe measurements No RACDT detection and (Quality of summary
treatment evidence)
Incidence of
Rate ratio 1.3 9 12 Very low
The evidence is very
(CI 95% 0.94 — 1.79) per 1000 per 1000 Due to very
clinical malaria uncertain about the
Based on data from serious
effect of RACDT on the
215,146 participants in indirectness, and
Difference: 3 more per 1000 incidence of clinical
3 studies. (Randomized serious
( CI 95% 1 fewer malaria
controlled) — 17 more ) imprecision 2
Parasitemia Based on data from Results from a difference-in- Very low The evidence is very
prevalence participants in 2 studies. differences analysis of the Brazil Due to serious uncertain about the
among those (Observational (non- study indicate a slight increase (by risk of bias, and effect or RACDT on
443 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Outcome Study results and Comparator Reactive case the Evidence Plain language
Timeframe measurements No RACDT detection and (Quality of summary
treatment evidence)
1. Inconsistency: no serious. Indirectness: very serious. The study on which this effect estimate is based compared
RACD to reactive drug administration (RDA), which is hypothesized to be a more effective intervention. Thus any effect
favoring RACD (vs. RDA) is likely to be underestimated, and any effect favoring the comparison should not necessarily be
interpreted as evidence that RACD has a harmful effect or no beneficial effect.. Imprecision: serious. The actual odds
ratio for the effect size = 1.85 (95% CI: 0.96, 20.00) and is therefore quite imprecise, spanning no effect to a large
harmful effect. (RevMan can only accommodate balanced confidence intervals but this effect size was calculated by
study authors using non-linear model post-estimation combinations.).
2. Inconsistency: no serious. Indirectness: very serious. The studies on which this effect estimate is based all compared
RACD to reactive drug administration (RDA), which is hypothesized to be a more effective intervention. Thus any effect
favoring RACD (vs. RDA) is likely to be underestimated, and any effect favoring the comparison should not necessarily be
interpreted as evidence that RACD has a harmful effect or no beneficial effect. Imprecision: serious. The pooled rate
ratio spans no effect to a substantial absolute effect in a low-transmission setting.
3. Risk of Bias: serious. These data come from non-randomized studies. One of the studies has a before-and-after
design with no control group. Inconsistency: serious. One study showed a slightly beneficial effect of RACD and the
other study showed a slightly negative effect. Indirectness: no serious. Imprecision: no serious.
4. Risk of Bias: very serious. Two of the three studies included here focused adverse event reporting only in the RDA
arm; in these studies no adverse events were reported from the RACD arm. Indirectness: serious. Two of the three
444 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
studies included here focused adverse event reporting only in the RDA arm; in these studies no adverse events were
reported from the RACD arm. Imprecision: very serious. We are unable to calculate an effect measure for AEs since they
were measured in most studies only in the RDA arm.
Attached Images
Summary
The systematic review identified two cRCTs in Namibia and South Africa (Gimnig et al unpublished evidence). The
study from Namibia (superiority trial design) was conducted as a 2x2 factorial design with RACDT alone, RDA alone,
RACDT plus RIRS, and RDA plus RIRS. The study from South Africa was designed as a non-inferiority trial comparing
RIRS to standard IRS (used in defined priority areas) that reached one third of houses. The results below report the
absolute effects (risk differences) of the intervention, as these were used by the GDG in its judgements; relative
effect sizes are available in the Research evidence.
Beneficial outcomes
• RIRS results in a large reduction in the prevalence of malaria (RD: -27 per 1000 persons; 95% CI: -35 to -8 per
1000 persons; one cRCT [superiority design]; high-certainty evidence).
• RIRS may reduce the incidence of clinical malaria. However, the effects of RIRS on clinical malaria vary and it is
possible that RIRS makes little or no difference (RD: -14 per 1000 p-y; 95% CI: -32 to 4 per 1000 p-y; one cRCT
[superiority design]; moderate-certainty evidence).
• RIRS probably results in little to no difference in incidence of clinical malaria (mean difference: 0.1 per 1000 p-y;
95% CI: -0.38 to 0.59 per 1000 p-y; one cRCT [non-inferiority design]; moderate-certainty evidence).
Adverse events
• RIRS results in little to no difference in reported adverse events (RD: 2 per 1000 persons; 95% CI: -2 to 1 per
1000 persons; one cRCT [superiority design]; high-certainty evidence).
• RIRS results in little to no difference in serious adverse events (deaths) (one cRCT [non-inferiority design]; high-
certainty evidence).
Intervention Certainty of
Comparator
Outcome Study results and Reactive the Evidence Plain language
no Reactive
Timeframe measurements indoor residual (Quality of summary
IRS
spraying evidence)
Prevalence of
malaria
Odds ratio 0.32 41 13
(CI 95% 0.15 — 0.8) per 1000 per 1000
(superiority Based on data from Reactive IRS reduces
trial) 4,082 participants in 1 High
Difference: 28 fewer per malaria prevalence
studies. (Randomized 1000
7 Critical controlled) ( CI 95% 35
445 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
Intervention Certainty of
Comparator
Outcome Study results and Reactive the Evidence Plain language
no Reactive
Timeframe measurements indoor residual (Quality of summary
IRS
spraying evidence)
fewer — 8 fewer )
Incidence of
Relative risk 0.65 39 25
clinical malaria per 1000 per 1000
(CI 95% 0.19 — 1.11)
(superiority Moderate Reactive IRS probably
Based on data from
design) Difference: 14 fewer per Due to serious reduces the incidence
2,000 participants in 1
1000 imprecision 1 of clinical malaria
studies. (Randomized
9 Critical controlled) ( CI 95% 32
fewer — 4 more )
Serious adverse
events (deaths,
Relative risk 0.69 0 0 Reactive IRS results in
(CI 95% 0.29 — 1.6) per 1000 per 1000
non-inferiority little to no difference in
Based on data from
trial) 393,387 participants in High serious adverse events
(deaths) compared with
1 studies. (Randomized
standard IRS
controlled)
446 of 447
WHO Guidelines for malaria - 14 March 2023 - World Health Organization (WHO)
1. Inconsistency: no serious. Indirectness: no serious. Imprecision: serious. Confidence interval of adjusted estimate
overlaps 1. Publication bias: no serious.
2. Inconsistency: no serious. Indirectness: serious. Study was designed as a non-inferiority study compared to standard
IRS. Imprecision: no serious. Publication bias: no serious.
Attached Images
7. SURVEILLANCE
8. METHODS
9. GLOSSARY
10.5. Recommendations for interventions in the final phase of elimination and prevention of
re-establishment
447 of 447