WHO Consolidated On Tuberculosis: Guidelines
WHO Consolidated On Tuberculosis: Guidelines
WHO Consolidated On Tuberculosis: Guidelines
consolidated
guidelines on
tuberculosis
Module 5: Management
of tuberculosis in children
and adolescents
WHO
consolidated
guidelines on
tuberculosis
Module 5: Management
of tuberculosis in children
and adolescents
WHO consolidated guidelines on tuberculosis. Module 5: management of tuberculosis in children and adolescents
Some rights reserved. This work is available under the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 IGO licence
(CC BY-NC-SA 3.0 IGO; https://creativecommons.org/licenses/by-nc-sa/3.0/igo).
Under the terms of this licence, you may copy, redistribute and adapt the work for non-commercial 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 licence. 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 licence shall be conducted in accordance with the mediation rules of the World
Intellectual Property Organization (http://www.wipo.int/amc/en/mediation/rules/).
Suggested citation. WHO consolidated guidelines on tuberculosis. Module 5: management of tuberculosis in children and adolescents.
Geneva: World Health Organization; 2022. Licence: 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 https://www.who.int/copyright.
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.
1. Introduction 1
1.1. Background 1
1.2. Rationale for the development of the 2022 consolidated guidelines 2
1.3. Objectives of the 2022 consolidated guidelines 2
1.4. Target audience 2
1.5. WHO recommendations relevant to the management of TB in children and adolescents 2
1.6. Scope of the guideline update 3
1.7. Publication, dissemination, evaluation and expiry 6
1.8. Document structure 6
iii
4.3. Consolidated recommendations on TB diagnostics and diagnostic approaches relevant
to children and adolescents 26
6. Models of TB care for case detection and provision of TPT in children and
adolescents 57
6.1. Decentralized and family-centred, integrated models of care to deliver child and
adolescent TB services 58
6.2. Consolidated recommendations on models of TB care relevant to children and
adolescents 63
7. Special situations 65
8. Research priorities 69
9. References 73
Annex 1. WHO recommendations incorporated in the guidelines on the
management of TB in children and adolescents 81
Web annexes:
Web annex 1: Methods and Expert Panels
https://apps.who.int/iris/bitstream/handle/10665/352507/9789240046788-eng.pdf
Web annex 2: GRADE Summary of Findings Tables
https://apps.who.int/iris/bitstream/handle/10665/352508/9789240046795-eng.pdf
Web annex 3: GRADE Evidence to Decision Tables
https://apps.who.int/iris/bitstream/handle/10665/352509/9789240046801-eng.pdf
Web annex 4: Summaries of unpublished data
https://apps.who.int/iris/bitstream/handle/10665/352510/9789240046818-eng.pdf
Web annex 5: Overview of consolidated WHO recommendations
https://apps.who.int/iris/bitstream/handle/10665/352512/9789240046825-eng.pdf
iv
Acknowledgements
The production and writing of the WHO consolidated guidelines on tuberculosis. Module 5: management
of tuberculosis in children and adolescents, 2022 was coordinated by Sabine Verkuijl, Annemieke Brands,
Kerri Viney and Tiziana Masini, under the guidance of Farai Mavhunga, head of the TB Vulnerable
Populations, Communities and Comorbidities unit and the overall direction of Tereza Kasaeva, Director
of the World Health Organization (WHO) Global Tuberculosis (TB) Programme. Colleagues from the
Prevention, Diagnosis, Treatment, Care and Innovation Unit, Global TB Programme, WHO, under
the leadership of Matteo Zignol, also contributed to these guidelines. The WHO Global Tuberculosis
Programme gratefully acknowledges the contribution of all experts involved in the development of
these guidelines.1
1
For more information about the areas of expertise, the gender and geographical distribution of participants as well as declarations of
interests and the management of potential conflicts for members of the GDG and External Review Group, see Web annex 1.
Acknowledgements v
International Development (USAID), United States), Rahab Mwaniki (Kenya Aids NGOs Consortium
(KANCO), Kenya), Marc Nicol (University of Western Australia, Australia), Elizabeth Maleche Obimbo
(University of Nairobi, Kenya), Peter Owiti (Wote Youth Development Project, Kenya), Nyan Win Phyo
(Civil Society Taskforce; World Vision, Thailand), Ramatoulaye Sall (Independent Consultant, Senegal),
Rina Triasih (Universitas Gadyah Mada, Indonesia) and Eric Wobudeya (Mulago National Referral
Hospital, Uganda; Makerere University – Johns Hopkins University (MU-JHU) Research Collaboration,
Uganda).
Evidence reviewers
The following persons contributed to the reviews and summarized evidence for the guidelines using
the Population, Intervention, Comparator and Outcomes (PICO) framework (see section 1.3 for more
information about the PICO questions).
PICO question 1 (TB screening in children): Bryan Vonasek (Baylor College of Medicine, United
States; University of Wisconsin, United States); Tara Ness, Alexander W Kay, Anna Mandalakas (Baylor
College of Medicine, United States); Yemisi Takwoingi (University of Birmingham, United Kingdom
of Great Britain and Northern Ireland (United Kingdom)); Susan S van Wyk (Stellenbosch University,
South Africa); Laura Ouellette (Texas Medical Center Library, United States); Ben J Marais (Marie Bashir
Institute for Infectious Diseases; University of Sydney, Australia); Karen R Steingart (Liverpool School
of Tropical Medicine, United Kingdom).
PICO question 2a (Integrated treatment decision algorithms to diagnose pulmonary TB): Ted Cohen
and Kenneth S Gunasekera (Yale School of Public Health, United States); James A Seddon (Imperial
College London, United Kingdom; Stellenbosch University, South Africa).
PICO question 2b (Use of Xpert Ultra in gastric aspirate or stool to diagnose pulmonary TB and
rifampicin resistance): Alexander W Kay and Tara Ness (Baylor College of Medicine, United States);
Yemisi Takwoingi (University of Birmingham, United Kingdom), Karen R Steingart (Liverpool School of
Tropical Medicine, United Kingdom).
PICO question 3 (Treatment shortening in children and adolescents with non-severe drug-susceptible
TB): Anna Turkova, Genevieve H Wills, Louise Choo, Krissy LeBeau, Margaret J Thomason, Angela
M Crook, Diana M Gibb (University College London, United Kingdom); Chishala Chabala (University
Teaching Hospital, Zambia), Helen McIlleron (University of Cape Town, South Africa), Paul Revill, James
Love-Koh (University of York, United Kingdom), Graeme Hoddinott (Stellenbosch University, South
Africa), Hayley Jones (University of Bristol, United Kingdom).
PICO question 4a and 4b (Use of bedaquiline and delamanid in children with MDR/RR-TB, aged
below 6 years (bedaquiline) and below 3 years (delamanid)): Pharmacokinetic (PK) and safety analysis:
Susan M. Abdel-Rahman (Children’s Mercy Research Institute, United States); Paediatric drug-resistant
(DR)-TB individual patient dataset (IPD): Anthony Garcia-Prats (Stellenbosch University, South Africa;
University of Wisconsin, United States); Vivian Cox, Rory Dunbar, Tina Sachs, Jessica Workman, Rose
Brown, Anneke C. Hesseling (Stellenbosch University, South Africa); Maria Garcia-Cremades, Kendra
Radtke, Alexander Floren, Rada Savic (University of California San Francisco, United States), Tamara
Kredo, Funeka Bango (South African Medical Research Council, South Africa).
PICO question 5 (Treatment of TB meningitis in children and adolescents): Giorgia Sulis (McGill
University, Canada), Gamuchirai Tavaziva, Andrea Benedetti and Faiz Ahmad Khan (McGill University,
Canada), Geneviève Gore (McGill University, Canada), Regan Solomons and Ronald van Toorn
(Stellenbosch University, South Africa), Stephanie Thee (Charité-Universitätsmedizin Berlin, Germany),
Jeremy Day (University of Oxford, United Kingdom), Silvia S Chiang (Alpert Medical School of Brown
University, United States; Rhode Island Hospital, United States).
PICO question 6 (Models of care for TB case detection and TB prevention in high TB burden settings):
Yael Hirsch-Moverman (Columbia University, United States), Hamidah Hussain (Interactive Research
vi WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
and Development (IRD) Global, Singapore), Daria Szkwarko (Brown University, United States), Courtney
Yuen (Harvard Medical School, United States).
Background question 1 (Socioeconomic impact of TB on affected families): Salla Atkins (Karolinska
Institutet, Sweden; Tampere University, Finland), Kristi Sidney-Annerstedt and Knut Lönnroth (Karolinska
Institutet, Sweden), Lauri Heimo, Maria Ribas Closa (Tampere University, Finland), Lieve Vanleeuw (South
African Medical Research Council, South Africa), Peter Wambi (Uganda Tuberculosis Implementation
Research Consortium, Uganda), Louisa Chenciner (Royal Free London NHS Foundation Trust, United
Kingdom), Uzochukwu Egere (Liverpool School of Tropical Medicine, United Kingdom), Daniel J Carter
and Delia Boccia (London School of Hygiene and Tropical Medicine, United Kingdom), Tom Wingfield
(Liverpool School of Tropical Medicine, United Kingdom; Liverpool University Hospital NHS Foundation
Trust, United Kingdom; Karolinska Institutet, Sweden). (Patient cost surveys): Nobuyuki Nishikiori
(WHO, Switzerland).
Background question 2 (Engaging adolescents with TB, or at risk of TB, in their care): Silvia S.
Chiang (Alpert Medical School of Brown University, United States; Rhode Island Hospital, United
States), Patricia Moscibrodzki (London School of Hygiene and Tropical Medicine, United Kingdom),
Leslie A Enane (Indiana University School of Medicine, United States). Contributors: Margaux Amara,
Meredith B Brooks, Virginia Byron, Jennifer Furin (Harvard Medical School, United States), Sarah
Bernays (University of Sydney, Australia; London School of Hygiene and Tropical Medicine, United
Kingdom), Yaroslava Bondarenko (Bogomolets National Medical University, Ukraine), Márcia Cortez
Bellotti de Oliveria (Universidade Federal do Rio de Janeiro, Brazil), Andrea T. Cruz (Baylor College of
Medicine, United States), Hernán Del Castillo Barrientos (Instituto Nacional de Salud del Niño-Breña,
Peru), Anthony Enimil (Kwame Nkrumah University of Science and Technology, Ghana; Komfo Anokye
Teaching Hospital, Ghana), Vivian Faith (Network of TB Champions in Kenya, Kenya), Gabriella Ferlazzo
(Médecins Sans Frontières, South Africa), Rashida Abbas Ferrand (Rhode Island Hospital, United States;
Biomedical Research and Training Institute, Zimbabwe), Graeme Hoddinott (Stellenbosch University,
South Africa), Petros Isaakidis (Médecins Sans Frontières, South Africa), Evgenia Karayeva (Brown
School of Public Health, United States), Katharina Kranzer (Rhode Island Hospital, United States;
Biomedical Research and Training Institute, Zimbabwe), Homa Mansoor (Médecins Sans Frontières,
India), Ben J Marais (Marie Bashir Institute for Infectious Diseases; University of Sydney, Australia), Lily
Meyersohn (Rhode Island Hospital, United States), Victoria Oliva Rapoport (Alpert Medical School
of Brown University, United States), Erika Mohr-Holland (Médecins Sans Frontières, South Africa),
Anh Phuong Nguyen (TB Patients Community of Vietnam, Hanoi, Vietnam), Joshua Ochieng Oliyo
(Committee of African Youth Advisors, Kenya), Clemax Couto Sant’Anna (Universidade Federal do
Rio de Janeiro, Brazil), Saning’o Saruni (Haydom Lutheran Hospital, Tanzania), Susan M Sawyer (Royal
Children’s Hospital and Murdoch Children’s Research Institute, Australia; University of Melbourne,
Australia), H. Simon Schaaf (Stellenbosch University, South Africa), James A Seddon (Imperial College
London, United Kingdom; Stellenbosch University, South Africa), Sangeeta Sharma (National Institute
of Tuberculosis and Respiratory Diseases, India), Alena Skrahina (The Republican Research and Practica
Centre for Pulmonology and TB, Belarus), Jeffrey R Starke (Baylor College of Medicine, United States),
Tania A Thomas (University of Virginia, United States), Rina Triasih (Universitas Gadjah Mada and Dr
Sardjito Hospital, Indonesia), Bazarragchaa Tsogt (Mongolian Tuberculosis Coalition, Mongolia), Henry
Welch (Baylor College of Medicine, United States; The University of Papua New Guinea, Papua New
Guinea), Olga Zvonareva (Maastricht University, the Netherlands).
Evidence reviewers for contextual factors: Olivier Marcy (Université de Bordeaux, France); Maryline
Bonnet and Manon Lounnas (Institut de Recherche pour le Développement, France), Eric Wobudeya
(Mulago National Referral Hospital, Uganda; Makerere University – Johns Hopkins University (MU-JHU)
Research Collaboration, Uganda); Pamela Nabeta (FIND, Switzerland), Claudia M Denkinger and
Mary Gaeddert (University of Heidelberg, Germany); Sushant Mukherjee, Mario JP Songane, Jean-
François Lemaire and Martina Casenghi (Elizabeth Glaser Pediatric AIDS Foundation, Switzerland);
Nyashadzaishe Mafirakureva and Peter J Dodd (University of Sheffield, United Kingdom); Nancy Medley
and Melissa Taylor (Liverpool School of Tropical Medicine, United Kingdom), Susanna S van Wyk
Acknowledgements vii
(Stellenbosch University, South Africa), Sandy Oliver (University College London, United Kingdom; and
University of Johannesburg, South Africa), Joanna Orne-Gliemann (Université de Bordeaux, France).
Other contributors
Pete Dodd (Sheffield University, United Kingdom), Anneke Hesseling (Stellenbosch University, South
Africa), Oliver Marcy (University of Bordeaux, France), Nicole Salazar-Austin (Johns Hopkins University,
United States), James Seddon (Imperial College London, England) served as technical resource persons
during the GDG meeting.
The following persons participated as observers during the GDG meeting: Draurio Barreira Cravo
Neto (Unitaid, Switzerland), Charlotte Colvin (USAID, United States), Anne Detjen (UNICEF, United
States), Thomas Gradel (Unitaid, Switzerland), Brian Kaiser (Stop TB Partnership Global Drug Facility,
Switzerland), Michael McCaul (Stellenbosch University, South Africa), Lawrence Mbuagbaw (St Joseph’s
Healthcare, Canada), Celeste Naude (Stellenbosch University, South Africa), Oxana Rucsineanu (TB
Community Advisory Board), Anna Scardigli (Global Fund to Fight AIDS, Tuberculosis and Malaria,
Switzerland), Cherise Scott (Unitaid).
The Global Tuberculosis Programme also thanks the WHO Guidelines Review Committee for their
review and approval of the guidelines.
Funder
This update was funded by grants provided to WHO by Unitaid and the United States Agency for
International Development.
viii WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Abbreviations
AIDS acquired immunodeficiency syndrome
ANC antenatal care
ART antiretroviral treatment
BCG bacille Calmette-Guérin
C(A)LHIV children (and adolescents) living with HIV infection
CHW community health worker
CXR chest X-ray or chest radiography
DR-TB drug-resistant tuberculosis
DS-TB drug-susceptible tuberculosis
DST drug susceptibility testing
DTG dolutegravir
E ethambutol
EPTB extrapulmonary TB
Eto ethionamide
FDC fixed-dose combination (medicines)
GDF Stop TB Partnership Global Drug Facility
GDG Guideline Development Group
GRADE Grading of Recommendations Assessment, Development and Evaluation
GUV germicidal ultraviolet
H isoniazid
HEPA high-efficiency particulate air
HIV human immunodeficiency virus
KSP Knowledge Sharing Platform
iCCM integrated community case management
IGRA interferon-gamma release assay
IMCI integrated management of childhood illness
IPD individual patient data (or dataset)
IPT isoniazid preventive therapy
LAMP loop-mediated isothermal amplification
LF-LAM lateral flow lipoarabinomannan assay
LPA line-probe assay
M moxifloxacin
MDR-TB multidrug-resistant tuberculosis
NAATs nucleic acid amplification tests
Abbreviations ix
NPA nasopharyngeal aspirate
NRTIs nucleoside reverse transcriptase inhibitors
NTLP National TB and Leprosy Control Programme
NTP National Tuberculosis Programme
OSF optimized sucrose flotation
PHC primary health care
PICO Population, Intervention, Comparator and Outcomes
PK pharmacokinetic(s)
PTB pulmonary tuberculosis
R rifampicin
RAL raltegravir
RR-TB rifampicin-resistant tuberculosis
SAGE Strategic Advisory Group of Experts on Immunisation
SAM severe acute malnutrition
SDGs Sustainable Development Goals
SL-LPA second-line line-probe assay
SOS simple one step (stool processing method)
SPK stool processing kit
TB tuberculosis
TBM tuberculous meningitis
TPT TB preventive treatment
TST tuberculin skin test
UNGA United Nations General Assembly
US FDA United States Food and Drug Administration
WHO World Health Organization
XDR-TB extensively drug-resistant tuberculosis
Z pyrazinamide
x WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Definitions
Unless otherwise specified, the terms defined here apply as used in this document. They may have
different meanings in other contexts.
Active (tuberculosis) case-finding: Provider-initiated screening and testing in communities by
mobile teams, often using mobile X-ray and rapid molecular tests. The term is sometimes used
synonymously with “systematic screening”.
Adherence: Extent to which a person’s behaviour (e.g. taking medicines, following a particular diet,
changing lifestyle) corresponds with agreed recommendations from a health care provider.
Advanced HIV disease: For adolescents and children aged 5 years and over, this is defined as a
CD4 cell count below 200 cells/mm3 or a WHO clinical stage 3 or 4 event at presentation for care.
All children aged under 5 years living with HIV should be considered as having advanced disease
at presentation.
Adverse event: Any untoward medical occurrence that may present in a person with TB during
treatment with a pharmaceutical product but that does not necessarily have a causal relationship
with the treatment.
Age groups: Unless stated otherwise in the text, the following definitions apply to the terms used
in this document:
• Infant: aged under 1 year (12 months).
• Child: aged under 10 years.
– Young child: aged under 5 years.
• Adolescent: aged 10–19 years (inclusive).
– Young adolescent: aged 10–14 years.
– Older adolescent: aged 15–19 years.
• Adult: aged 20 years or over.
Background HIV and tuberculosis drug resistance prevalence: Settings with high HIV prevalence
are defined as those in which the HIV prevalence is 1% or higher among adult pregnant women, or
5% or higher among people with TB. WHO does not intend to establish thresholds for low, moderate
or high levels of prevalence of isoniazid resistance. National TB programmes will establish definitions
for their own countries.
Bacteriologically confirmed tuberculosis: TB diagnosed in a biological specimen by a WHO-
approved rapid test such as Xpert® MTB/RIF or LF-LAM, smear microscopy or culture.
Contact: Any person exposed to a person with TB.
Contact investigation: Systematic identification of people, including children and adolescents, with
previously undiagnosed TB disease and TB infection among the contacts of an index TB patient in the
household and in comparable settings in which transmission occurs. It consists of identification, clinical
evaluation and/or testing and provision of appropriate TB treatment (for people with confirmed TB)
or TB preventive treatment (for people without TB disease).
Decentralization: Depending on the standard in the research settings used for the comparator, this
includes provision of, access to or capacity for child and adolescent TB services at a lower level of
Definitions xi
the health system than the lowest level where this is currently routinely provided. In most settings,
decentralization applies to the district hospital (first referral level hospital) level and/or primary health
care level and/or community level. Interventions for decentralization include capacity-building of
various cadres of health care workers, expanding access to diagnostic services, ensuring availability
of TB medicines for children and adolescents, and follow-up of children and adolescents with TB or
on TB preventive treatment.
Differentiated HIV service delivery model: Person-centred approach to simplify provision of HIV
services across the cascade in ways that better serve the needs of people living with HIV and reduce
unnecessary burdens on the health system.
Drug susceptibility testing (DST): In vitro testing using either molecular genotypic techniques
to detect resistance-conferring mutations, or phenotypic methods to determine susceptibility to a
medicine.2
Extensive (or advanced) pulmonary tuberculosis disease: Presence of bilateral cavitary disease
or extensive parenchymal damage on chest radiography (CXR). In children aged under 15 years,
advanced disease is usually defined by the presence of cavities or bilateral disease on CXR.
Extensively drug-resistant tuberculosis (XDR-TB):3
• Pre-XDR-TB: TB caused by Mycobacterium tuberculosis strains that fulfil the definition of multidrug-
resistant TB (MDR-TB) or rifampicin-resistant TB (RR-TB) and that are also resistant to any
fluoroquinolone.4
• XDR-TB: TB caused by M. tuberculosis strains that fulfil the definition of MDR/RR-TB and that are
also resistant to any fluoroquinolone and at least one additional Group A medicine.5
Extrapulmonary tuberculosis (EPTB) (classification): Any bacteriologically confirmed or clinically
diagnosed case of TB involving organs other than the lungs (e.g. pleura, peripheral lymph nodes,
abdomen, genitourinary tract, skin, joints and bones, meninges).6
Family-centred, integrated care: Family-centred models of care refer to interventions selected on the
basis of the needs, values and preferences of the child or adolescent and their family or caregiver. This
can include health education, communication, material or psychological support. Integrated services
refer to approaches to strengthen collaboration, coordination, integration and harmonization of child
and adolescent TB services with other child health-related programmes and services. This can include
integration of models of care for TB screening, prevention, diagnosis and treatment with other existing
service delivery platforms for maternal and child health (e.g. antenatal care, integrated community
case management, integrated management of childhood illnesses) and other related services (e.g.
HIV, nutrition, immunization). Other examples include evaluation of children and adolescents with
common comorbidities (e.g. meningitis, malnutrition, pneumonia, chronic lung disease, diabetes, HIV)
for TB and community health strategies integrating child and adolescent TB awareness, education,
screening, prevention and case-finding into training and service delivery activities.
2
Implementing tuberculosis diagnostics: a policy framework. Geneva: World Health Organization; 2015 (https://apps.who.int/iris/
handle/10665/162712, accessed 11 March 2022).
3
Meeting report of the WHO expert consultation on the definition of extensively drug-resistant tuberculosis, 27–29 October 2020.
Geneva: World Health Organization; 2021 (https://www.who.int/publications/i/item/meeting-report-of-the-who-expert-consultation-
on-the-definition-of-extensively-drug-resistant-tuberculosis, accessed 11 March 2022).
4
The fluoroquinolones include levofloxacin and moxifloxacin as currently recommended by WHO for inclusion in shorter and
longer regimens.
5
Group A medicines are currently levofloxacin or moxifloxacin, bedaquiline and linezolid; therefore, XDR-TB is MDR/RR-TB that is
resistant to a fluoroquinolone and either bedaquiline or linezolid (or both). Group A medicines could change in the future. Therefore,
the terminology “Group A” is appropriate here and will apply to any Group A medicines in the future.
6
Following a WHO expert consultation in September 2021, intrathoracic lymph node TB is now classified as pulmonary TB in children.
xii WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Grading of Recommendations Assessment, Development and Evaluation (GRADE): System for
rating quality of evidence and strength of recommendations. This approach is explicit, comprehensive,
transparent and pragmatic.7
High tuberculosis transmission setting: Setting with a high frequency of people with undetected
or undiagnosed TB disease, or where people with infectious TB are present and there is a high risk
of TB transmission. People with TB are most infectious when they are untreated or inadequately
treated. Spread is increased by aerosol-generating procedures and by the presence of highly
susceptible people.
Household contact: Person who shared the same enclosed living space as the index case for one
or more nights or for frequent or extended daytime periods during the 3 months before the start of
current treatment.
Index case (index patient) of tuberculosis: Initially identified person of any age with new or
recurrent TB in a specific household or other comparable setting in which others may have been
exposed. An index case is the person on which a contact investigation is centred but is not necessarily
the source case.
Inpatient health care setting: Health care facility where people are admitted and assigned a bed
while undergoing diagnosis and receiving treatment and care, for at least one overnight stay.
Integrated treatment decision algorithm: Flowchart allocating evidence-based scores to
microbiological, clinical and radiological features that allow clinicians to make decisions regarding
starting TB treatment in children.
Interferon-gamma release assay (IGRA): Blood test used to test for Mycobacterium tuberculosis
infection by measuring the body’s immune response to TB bacteria.
Multidrug-resistant tuberculosis (MDR-TB): TB caused by Mycobacterium tuberculosis strains that
are resistant to at least both rifampicin and isoniazid.
New case: Newly registered episode of TB in a person who has never been treated for TB or has
taken TB medicines for less than 1 month.
Non-severe pulmonary tuberculosis for the purpose of determining treatment duration
for drug-susceptible tuberculosis: Intrathoracic lymph node TB without airway obstruction;
uncomplicated TB pleural effusion or paucibacillary, non-cavitary disease confined to one lobe of
the lungs and without a miliary pattern.
Number needed to screen: Number of people who need to undergo screening in order to diagnose
one person with TB disease.
Operational research or implementation research: In the context of this document, applied
research that aims to develop the critical evidence base that informs the effective, sustained and
embedded adoption of interventions within a health system to improve health or patient outcomes.
Such research deals with the knowledge gap between efficacy, effectiveness and current practice to
produce the greatest gains in disease control.8 Operational research also provides decision-makers
with information to enable them to improve the performance of their health programmes.9
7
GRADE is a transparent framework for developing and presenting summaries of evidence. It provides a systematic approach for making
clinical and public health practice recommendations. Guyatt GH, Oxman AD, Vist GE, et al. GRADE: an emerging consensus on rating
quality of evidence and strength of recommendations. BMJ. 2008;336:924.
8
Guide to operational research in programs supported by the Global Fund. Geneva: Global Fund to Fight AIDS, Tuberculosis and Malaria;
2007 (https://www.who.int/hiv/pub/operational/or_guide_gf.pdf, accessed 11 March 2022).
9
Expanding capacity for operations research in reproductive health: summary report of a consultative meeting. Geneva: World Health
Organization; 2003 (https://apps.who.int/iris/bitstream/handle/10665/67936/WHO_RHR_02.18.pdf?sequence=1&isAllowed=y, accessed
11 March 2022).
Definitions xiii
Outpatient health care setting: Health care facility where people are undergoing diagnosis
and receiving treatment and care but are not admitted for overnight stays (e.g. ambulatory clinic,
dispensary).
Passive case-finding: Patient-initiated pathway to TB diagnosis involving a person with TB disease
who experiences symptoms that they recognize as serious; the person having access to and seeking
care, and presenting spontaneously at an appropriate health facility; a health worker correctly assessing
that the person fulfils the criteria for presumptive TB; and successful use of a diagnostic algorithm
with sufficient sensitivity and specificity to diagnose TB.
People who use drugs: People who engage in the harmful or hazardous use of psychoactive
substances that could impact negatively on their health, social life, resources or legal situation.
Presumptive tuberculosis: Person who presents with symptoms or signs suggestive of TB.
Previously treated: People who have previously received 1 month or more of TB medicines. Previously
treated people may have been treated with a first-line regimen for drug-susceptible TB or a second-
line regimen for drug-resistant forms.
Programmatic management of tuberculosis preventive treatment: All coordinated activities by
public and private health caregivers and the community aimed at scaling up TB preventive treatment
to people who need it.
Pulmonary tuberculosis (PTB) (classification): Any bacteriologically confirmed or clinically
diagnosed case of TB involving the lung parenchyma or the tracheobronchial tree, including tuberculous
intrathoracic lymphadenopathy (mediastinal and/or hilar), without radiographic abnormalities in the
lungs.10 Miliary TB is classified as PTB because there are lesions in the lungs. A person with both PTB
and extrapulmonary TB should be classified as having PTB.
Rifampicin-resistant tuberculosis (RR-TB): TB caused by Mycobacterium tuberculosis strains
resistant to rifampicin. These strains may be susceptible or resistant to isoniazid (i.e. MDR-TB) or
resistant to other first-line or second-line TB medicines. In these guidelines and elsewhere, MDR-TB
and RR-TB cases are often grouped together as MDR/RR-TB and are eligible for treatment with an
MDR-TB regimen.
Rifampicin-susceptible, isoniazid-resistant tuberculosis: TB caused by Mycobacterium tuberculosis
strains resistant to isoniazid and susceptible to rifampicin.
Serious adverse event: Adverse event that can lead to death or a life-threatening experience,
to hospitalization or prolongation of hospitalization, to persistent or significant disability, or to a
congenital anomaly. Serious adverse events that do not immediately result in one of these outcomes
but that require an intervention to prevent such an outcome from happening are included. Serious
adverse events may require a drastic intervention, such as termination of the medicine suspected of
having caused the event.
Severe acute malnutrition: Presence of oedema of both feet or severe wasting (weight-for-height/
length less than −3 standard deviations/Z-scores or mid-upper arm circumference less than 115 mm).11
Severe extrapulmonary tuberculosis: Presence of miliary (disseminated) TB or TB meningitis.
In children and young adolescents aged under 15 years, extrapulmonary forms of disease other
than lymphadenopathy (peripheral nodes or isolated mediastinal mass without compression) are
considered to be severe.
10
Following a WHO expert consultation in September 2021, intrathoracic lymph node TB is now classified as pulmonary TB in children.
11
Pocket book of hospital care for children: guidelines for the management of common childhood illnesses, 2nd edition. Geneva:
World Health Organization; 2013 (https://apps.who.int/iris/bitstream/handle/10665/81170/9789241548373_eng.pdf, accessed
27 September 2021).
xiv WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Severe pneumonia: Cough or difficulty in breathing plus at least one of the following:
• central cyanosis or oxygen saturation <90% on pulse oximetry;
• severe respiratory distress (e.g. grunting, nasal flaring, very severe chest indrawing);
• signs of pneumonia with a general danger sign (inability to breastfeed or drink, persistent vomiting,
lethargy or unconscious, convulsions, stridor in a calm child, severe malnutrition).8
Source case: Person with TB disease who infected others in a new setting. This could be the index
patient or another person who was not identified.
Systematic screening for tuberculosis disease: Systematic identification of people at risk for TB
disease in a predetermined target group by assessing symptoms and using tests, examinations or
other procedures that can be applied rapidly. For those who screen positive, the diagnosis needs
to be established by one or several diagnostic tests and additional clinical assessments. This term is
sometimes used interchangeably with “active tuberculosis case-finding”. It should be distinguished
from testing for TB infection (with a TB skin test or interferon-gamma release assay).
Treatment outcomes and relapse: Categories for treatment outcomes used in this document and
the term “relapse” were applied according to the definitions agreed for use by TB programmes, unless
otherwise specified.12,13
Tuberculin skin test (TST): Intradermal injection of a combination of mycobacterial antigens that
elicit an immune response (delayed-type hypersensitivity), represented by induration, which can be
measured in millimetres. TST is used to diagnose TB infection.
Tuberculosis (TB): Disease state due to Mycobacterium tuberculosis. In this document, it is commonly
referred to as “TB disease” to distinguish it from “TB infection”.
Tuberculosis infection: State of persistent immune response to stimulation by Mycobacterium
tuberculosis antigens with no evidence of clinically manifest TB disease. This is referred to as “TB
infection” as distinct from “TB disease”. There is no gold standard test for direct identification of M.
tuberculosis infection in humans. Most infected people have no signs or symptoms of TB but are at
risk for TB disease. The term “latent TB infection” has been replaced by the term “TB infection”.
Tuberculosis preventive treatment (TPT): Treatment offered to people considered at risk of TB
disease to reduce that risk. Also referred to as “treatment of TB infection” or “TB preventive therapy”.
Underweight: Among adolescents, this usually refers to a body mass index below 18.5. Among
children aged under 10 years, it usually refers to a weight-for-age Z-score below −2 standard deviations.
12
Definitions and reporting framework for tuberculosis – 2013 revision. Geneva: World Health Organization; 2013 (WHO/ HTM/TB/2013.2;
http://apps.who.int/iris/bitstream/10665/79199/1/9789241505345_eng.pdf, accessed 11 March 2022).
13
Meeting report of the WHO expert consultation on drug-resistant tuberculosis treatment outcome definitions, 17–19 November 2020.
Geneva: World Health Organization; 2021 (https://apps.who.int/iris/rest/bitstreams/1336957/retrieve, accessed 11 March 2022).
Definitions xv
xvi WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Executive summary
Introduction
Children and young adolescents (aged below 15 years) represent about 11% of all people with
tuberculosis (TB) globally. This means that 1.1 million children become ill with TB every year, almost
half of them below five years of age. National TB programmes (NTPs) only notify less than half of
these children, meaning that there is a large case detection gap (1). The reasons for this gap include
challenges with specimen collection and bacteriological confirmation of TB in young children, due
to the paucibacillary nature of TB disease in this age group and the lack of highly sensitive point-of-
care tests. In 2020, the COVID-19 pandemic had an additional negative impact on TB notifications
in children. In addition to the case detection gap, only one third of child contacts below five years of
age eligible for TB preventive treatment (TPT) received it in 2020. Young children are at higher risk of
developing TB disease, including severe forms of TB, after TB infection, and the majority do so within
a few months following exposure and infection (2, 3). In addition to children and young adolescents,
over half a million older adolescents (aged 15–19 years) are estimated to develop TB every year (4).
The United Nations Sustainable Development Goal (SDGs) (5) and the World Health Organization
(WHO) End TB Strategy (6) include targets to reduce TB incidence by 80% and TB deaths by 90% to
be achieved by the year 2030, relative to baseline levels in 2015. In addition, to accelerate progress
towards these global targets, the Resolution adopted by the United Nations General Assembly at the
High-Level Meeting on the fight against tuberculosis in September 2018 commits to diagnosing and
treating 40 million people with TB (including 3.5 million children), and 1.5 million people with drug-
resistant TB (DR-TB) (including 115 000 children) by 2022. It also commits to providing at least 30
million people (including 4 million child contacts under five years of age), 20 million other household
contacts (including children aged five years and above) and 6 million people living with HIV (including
children) with TPT by 2022 (7).
Rationale
To support countries in preventing and managing TB in children and adolescents, WHO’s Global
Tuberculosis Programme published the WHO Guidance for national tuberculosis programmes on the
management of tuberculosis in children (second edition) in 2014 (8). Since the publication of the second
edition, new evidence related to diagnostic approaches for TB, treatment for drug-susceptible TB,
DR-TB and TB meningitis, as well as models of care relevant to children and adolescents has become
available. The WHO consolidated guidelines on tuberculosis. Module 5: management of tuberculosis
in children and adolescents (2022) is a consolidated guideline of new and existing recommendations
(see annex 1), and replaces the 2014 guidance. It complements existing WHO guidelines on the
management of TB, recognizing the unique characteristics and needs of these groups, as well as those
of their parents, caregivers and families. The guidelines are complemented by the WHO operational
handbook on tuberculosis. Module 5: Management of tuberculosis in children and adolescents, which
provides guidance on how to implement the recommendations in the guidelines.
Target audience
The target audience for these consolidated guidelines consists primarily of NTPs, primary health
care (PHC) programmes, maternal and child health programmes, national AIDS programmes (or
their equivalents in health ministries) and other health policy-makers. They also target generalist
and specialist paediatricians, clinicians and health practitioners working on TB, HIV and/or infectious
diseases in public and private sectors, the educational sector, nongovernmental, civil society and
community-based organizations, as well as technical and implementing partners.
Diagnostic approaches
1 In children with signs and symptoms of pulmonary TB, Xpert Ultra should be used
as the initial diagnostic test for TB and detection of rifampicin resistance on sputum,
nasopharyngeal aspirate, gastric aspirate or stool, rather than smear microscopy/culture
and phenotypic drug susceptibility testing (DST).
(UPDATED: strong recommendation, moderate certainty of evidence for test accuracy in
stool and gastric aspirate; low certainty of evidence for test accuracy in sputum; very low
certainty of evidence for test accuracy in nasopharyngeal aspirate)
2 In children with presumptive pulmonary TB attending health care facilities, integrated
treatment decision algorithms may be used to diagnose pulmonary TB.
(NEW: interim, conditional recommendation, very low certainty of evidence)
Treatment regimens
3 In children and adolescents between 3 months and 16 years of age with non-severe TB
(without suspicion or evidence of multidrug- or rifampicin-resistant TB (MDR/RR-TB), a
4-month treatment regimen (2HRZ(E)/2HR) should be used.
(NEW: strong recommendation, moderate certainty of evidence)
xviii WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
4 In children with MDR/RR-TB aged below 6 years, an all-oral treatment regimen containing
bedaquiline may be used.
(NEW: conditional recommendation, very low certainty of evidence)14
5 In children with MDR/RR-TB aged below 3 years delamanid may be used as part of longer
regimens.
(NEW: conditional recommendation, very low certainty of evidence)15
6 In children and adolescents with bacteriologically confirmed or clinically diagnosed TB
meningitis (without suspicion or evidence of MDR/RR-TB), a 6-month intensive regimen
(6HRZEto) may be used as an alternative option to the 12-month regimen (2HRZE/10HR).
(NEW: conditional recommendation, very low certainty of evidence)
Models of TB care
7 In high TB burden settings, decentralized TB services may be used in children and
adolescents with signs and symptoms of TB and/or in those exposed to TB.
(NEW: conditional recommendation, very low certainty of evidence)
8 Family-centred, integrated services in addition to standard TB services may be used in
children and adolescents with signs and symptoms of TB and/or those exposed to TB.
(NEW: conditional recommendation, very low certainty of evidence)
Chapter Topic
Screening Screening for TB in targeted populations
and contact
investigation Tools for screening for TB
HIV counselling and testing for household and close contacts of people with TB
Prevention TB infection prevention and control: administrative controls
TB infection prevention and control: environmental controls
TB infection prevention and control: respiratory protection
BCG vaccination: BCG vaccination at birth vs at six weeks
BCG vaccination: Selective BCG vaccination
BCG vaccination: Need for revaccination
BCG vaccination: BCG vaccination for HIV-infected infants
14
This recommendation applies to and complements the 2020 WHO recommendations on shorter and longer regimens that contain
bedaquiline: A shorter all-oral bedaquiline-containing regimen of 9–12 months duration is recommended in eligible patients with
confirmed multidrug- or rifampicin-resistant tuberculosis (MDR/RR-TB) who have not been exposed to treatment with second-line TB
medicines used in this regimen for more than one month, and in whom resistance to fluoroquinolones has been excluded (Conditional
recommendation, very low certainty in the evidence); bedaquiline should be included in longer multidrug-resistant TB (MDR-TB) regimens
for patients aged 18 years or more (Strong recommendation, moderate certainty in the estimates of effect); bedaquiline may also be
included in longer MDR-TB regimens for patients aged 6–17 years; (Conditional recommendation, very low certainty in the estimates
of effect) (9).
15
This recommendation complements the 2020 WHO recommendation on longer regimens that contain delamanid: Delamanid may be
included in the treatment of MDR/RR-TB patients aged 3 years or more on longer regimens (Conditional recommendation, moderate
certainty in the estimates of effect) (9).
xx WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Treatment Treatment of severe forms of pulmonary TB in children
Treatment of infants aged 0–3 months with TB
Treatment of osteoarticular TB in children
Use of thrice weekly dosing and fixed dose combination tablets
Use of corticosteroids in TB meningitis
A 4-month treatment regimen composed of isoniazid, rifapentine, moxifloxacin
and pyrazinamide for treatment of drug-susceptible pulmonary TB
Regimen for rifampicin-susceptible and isoniazid-resistant TB
Shorter all-oral bedaquiline-containing regimen for multidrug- or rifampicin-
resistant TB (MDR/RR-TB)
Longer regimens for MDR/RR-TB
The bedaquiline, pretomanid and linezolid (BPaL) regimen for MDR-TB with
additional fluoroquinolone resistance
Monitoring patient response to MDR-TB treatment using culture
Models of TB Health education and counselling
care
Treatment adherence interventions
Treatment administration options
Ambulatory care for persons with MDR-TB
Decentralized model of care for persons with MDR-TB
Special Routine HIV testing for persons with presumptive and diagnosed TB
situations
Co-trimoxazole prophylaxis for infants, children and adolescents living with HIV
General recommendations on eligibility for ART
Timing of ART for children and adolescents with TB
First-line ART regimens
Second-line ART regimens
Management of severe acute malnutrition (SAM)
Management of moderate undernutrition
Contact investigation
Based on these guidelines, tools to support implementation have been developed, including new
integrated treatment decision algorithms and an updated table on dosing of second-line TB medicines.
In accordance with the process for updating WHO guidelines, a systematic and continuous process
of identifying and bridging evidence gaps following guideline dissemination will be employed. If new
evidence that could potentially impact the current evidence base for any of the recommendations
is identified, it will be reviewed with a view to updating the recommendation. WHO welcomes
suggestions regarding additional questions for inclusion in future updates of the guideline.16
16
The WHO Global Tuberculosis Programme can be contacted at gtbprogramme@who.int.
xxii WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
1. Introduction
1.1. Background
Children and young adolescents (aged below 15 years) represent about 11% of all people with TB
globally. This means that close to 1.1 million children become ill with TB every year, almost half of them
below five years of age. National TB programmes (NTPs) only notify less than half of these children,
meaning that there is a large case detection gap (1). The reasons for this gap include challenges with
specimen collection and bacteriological confirmation of TB in young children, due to the paucibacillary
nature of TB disease in this age group and the lack of highly sensitive point-of-care tests (10). In 2020,
the COVID-19 pandemic had an additional negative impact on TB notifications in children, with a 24%
decrease in notifications compared to 2019 (in comparison, notifications in people aged 15 years
and above decreased by 18%). In addition to the case detection gap, only one third of child contacts
below five years of age eligible for TB preventive treatment (TPT) actually received it in 2020 (1).
Young children are at higher risk of developing TB disease, including severe forms of TB, than older
age groups, after TB infection, and the majority do so within a few months following exposure and
infection (2, 3). In addition to children and young adolescents, over half a million older adolescents
(15–19 years) are estimated to develop TB every year (4).
The United Nations Sustainable Development Goal (SDGs) (5) and the World Health Organization
(WHO) End TB Strategy (6) include targets to reduce TB incidence by 80% and TB deaths by 90%
to be achieved by the year 2030, relative to baseline levels in 2015. In addition, the Resolution
adopted by the United Nations General Assembly (UNGA) at the High-Level Meeting on the fight
against tuberculosis in September 2018 commits to diagnosing and treating 40 million people with
TB (including 3.5 million children), and 1.5 million people with DR-TB (including 115 000 children) by
2022. It also commits to providing at least 30 million people (including 4 million child contacts under
five years of age), 20 million other household contacts (including children over the age of five years)
and 6 million people living with HIV (including children) with TPT by 2022 (7).
To support countries in preventing and managing TB in children and adolescents, WHO’s Global
Tuberculosis Programme published the WHO Guidance for national tuberculosis programmes on
the management of tuberculosis in children (second edition) in 2014. Since these guidelines were
published, new recommendations and guidance on the management of TB have been published
in WHO guidelines and other policy documents on TB prevention, screening, diagnosis, treatment,
management and models of care. Many of these recommendations are also applicable to children and
adolescents. In addition, new evidence related to the management of TB in children and adolescents
became available to WHO in 2021. Some of these data were received in response to a specific request
from WHO for data on the management of TB in children and adolescents, issued as an Expression
of Interest in July 2020,17 which was developed in consultation with the core team of the Child and
Adolescent TB Working group.18 Data from a randomized controlled trial on treatment shortening
for children with non-severe TB were made available to the WHO in 2021. Therefore, in 2021, the
WHO convened a Guideline Development Group (GDG) to review new evidence on the management
of TB in children and adolescents. This guideline update includes new recommendations that were
17
WHO public call for data on the management of TB in children and adolescents. 24 July 2020. WHO [website] (https://www.who.int/
news-room/articles-detail/who-public-call-for-data-on-the-management-of-tb-in-children-and-adolescents, accessed 20 January 2022).
18
Child and Adolescent TB Working Group (https://www.stoptb.org/wg/dots_expansion/childhoodtb/, accessed 20 January 2022).
1. Introduction 1
issued at the GDG meeting in May-June 2021 as well as current recommendations from other WHO
guidelines that are relevant to the management of TB in children and adolescents (including those
that are in the previous guidelines on the management of TB in children that have been validated).
This update consolidates all recommendations in one guideline.
2 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
the 2014 guidance that remain unchanged. These latter recommendations cover key components
of high-quality TB care for which a review of the evidence was either not done (such as HIV testing
for persons with presumptive TB and TB disease), or for which no new evidence was available. A
summary of the changes to the 2014 guidance is provided in the supplementary table in annex 2.
The full details of the new recommendations, including evidence and justification, subgroup and
implementation considerations and monitoring and evaluation, are provided in this guideline. Other
WHO recommendations relevant to the management of TB in children and adolescents have been
consolidated in tables in the relevant chapters. It is important to note that the original wording of
the recommendations based on the source guidelines has been included. In some source guidelines,
age groups have been defined differently compared to the 2022 consolidated guidelines on the
management of TB in children and adolescents, and the adult age group may include adolescents
aged 15 years and above. Where this is the case, it has been indicated in the tables. Users of this
guideline update are advised to refer to the original guideline for full information related to the
recommendation. In addition, all WHO recommendations on TB are now included in the WHO TB
Knowledge Sharing Platform (KSP)19 which can be searched by population (e.g. children or people
living with HIV) or by topic (e.g. diagnosis or treatment). Refer to section 1.7 for further details.
For ease of reference, a full overview of all new and consolidated recommendations is included in
web annex 5.
19
WHO TB Knowledge Sharing Platform (https://extranet.who.int/tbknowledge)
1. Introduction 3
Figure 1: Cascade of care in children and adolescents exposed to and with TB, with
broad topics of PICO questions with corresponding numbers as per section 1.3.1
Diagnostic approaches Treatment of
(PICO 2 a and b) DS-TB, DR-TB
and TBM
Accessed (PICO 3, 4a
health care and 4b, and 5)
Feeling ill system Diagnosed Treated
Diseased
Did NOT
access health
care system
Source: Roadmap towards ending TB in children and adolescents. Second edition. Geneva: World Health Organization; 2018.
DS/DR-TB: drug-susceptible/drug-resistant TB; TBM: TB meningitis.
Based on this understanding of the cascade of TB care in children and adolescents, the potential
contribution of interventions targeting different steps in this cascade is summarized in the following
logic model (which was the model used to frame these guidelines) with the potential contribution of
the evidence to short-term and long-term outcomes (Figure 2).
Figure 2: The logic model used for the guidelines on the management of TB in children
and adolescents
*The numbers under short- and long-term outcomes refer to the PICO questions in section 1.6.1.
DS/DR-TB: drug-susceptible/drug-resistant TB.
4 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
A WHO Steering Group drafted a series of PICO (questions, which were discussed and agreed
upon by the GDG during a preparatory webinar. Systematic reviews or other studies were sought or
commissioned for each of the PICO questions. In addition, two priority background questions were
formulated to provide additional insight into implementation considerations related to TB care for
children and adolescents. After a review of preliminary data, the GDG recommended that one PICO
question (PICO question 1 on TB screening) should not be further pursued as the evidence for this
question overlapped directly with the evidence reviewed for the WHO consolidated guidelines on
tuberculosis. Module 2: screening – systematic screening for tuberculosis disease, published in 2021 (11).
Therefore, this PICO question was not considered by the GDG at their meeting in May-June 2021.
1. Introduction 5
PICO question 5: Treatment of paediatric TB meningitis
In children and adolescents with presumed or bacteriologically confirmed drug-susceptible TB
meningitis, should a 6-month intensive regimen, compared to the 12-month regimen that conforms
to current WHO guidelines be used?
PICO question 6: Models of care for TB case detection and TB prevention settings with a
prevalence of TB in the general population of 100 per 100 000 or more:
a. In children and adolescents with signs and symptoms of TB, should decentralization of child and
adolescent TB services versus centralized child and adolescent TB services (at referral or tertiary
hospital level) be used?
b. In children and adolescents exposed to TB, should decentralization of child and adolescent TB
prevention and care services versus centralized prevention and care services (at referral or tertiary
hospital level) be used to increase coverage of TPT in eligible children and adolescents?
c. In children and adolescents with signs and symptoms of TB, should family-centred, integrated
services versus standard, non-family-centred, non-integrated services be used?
d. In children and adolescents exposed to TB, should family-centred, integrated services versus
standard, non-family-centred, non-integrated services be used to increase coverage of TPT in
eligible children and adolescents?
6 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Box 1: Structure of the 2022 consolidated guidelines
1. Introduction 7
8 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
2. TB screening and contact
investigation
This chapter includes current WHO recommendations that apply to children and adolescents on TB
screening and contact investigation. They have been consolidated from current WHO guidelines
on systematic screening for TB disease and contact investigation, namely the WHO consolidated
guidelines on tuberculosis. Module 2: screening – systematic screening for tuberculosis disease (11)
and Guidance for national tuberculosis programmes on the management of tuberculosis in children
(second edition) (8). For more information on each recommendation including the remarks, source
of evidence, justification, subgroup, implementation and monitoring and evaluation considerations,
the source guidelines or WHO TB KSP should be consulted.
10 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
3. Prevention of TB
This chapter includes current WHO recommendations that apply to children and adolescents on TB
prevention. They have been consolidated from current WHO guidelines on TB infection, prevention
and control, a BCG position paper and guidelines on TPT, namely the WHO guidelines on tuberculosis
infection prevention and control, 2019 update (12), the BCG vaccines: WHO position paper (published in
the Weekly Epidemiological Record) (13) and the WHO consolidated guidelines on tuberculosis. Module 1:
prevention – tuberculosis preventive treatment (14). For more information on each recommendation
including the remarks, source of evidence, justification, subgroup, implementation and monitoring
and evaluation considerations, the source guidelines or WHO TB KSP should be consulted.
3. Prevention of TB 11
Ventilation systems (including natural, mixed-mode, mechanical ventilation and recirculated
air through high-efficiency particulate air (HEPA) filters) are recommended to reduce M.
tuberculosis transmission to health workers, persons attending health care facilities or other
persons in settings with a high risk of transmission.
(Conditional recommendation, very low certainty in the estimates of effects)
Respiratory protection
Particulate respirators, within the framework of a respiratory protection programme, are
recommended to reduce M. tuberculosis transmission to health workers, persons attending
health care facilities or other persons in settings with a high risk of transmission.
(Conditional recommendation, very low certainty in the estimates of effects)
BCG vaccination: BCG vaccines: WHO position paper, 2018 (13)
Bacillus Calmette-Guérin (BCG) vaccination at birth vs at 6 weeks
In countries or settings with a high incidence of TB and/or leprosy, a single dose of BCG vaccine
should be given to neonates at birth, or as soon as possible thereafter, for prevention of TB
and leprosy disease. If it cannot be given at birth, it should be given at the earliest opportunity
thereafter and should not be delayed. Any delay in vaccination may lead to opportunities for
known or unknown exposure to TB or leprosy-infected contacts.
Co-administration of BCG with the hepatitis B birth dose is safe and strongly recommended. In
order to avoid missed opportunities for neonatal vaccination, BCG multi-dose vials should be
opened and used despite any wastage of unused vaccine.
If the birth dose was missed, catch-up vaccination of unvaccinated older infants and children
is recommended since evidence shows it is beneficial. Catch-up vaccination should be done at
the earliest convenient encounter with the health care system to minimize known or unknown
exposure to TB or leprosy infected contacts.
Selective BCG vaccination
Countries with a low incidence of TB or leprosy may choose to selectively vaccinate neonates
in recognized risk groups for developing disease. High-risk groups to be considered for BCG
vaccination include the following:
• Neonates to parents (or other close contacts/relatives) with previous TB or leprosy
• Neonates in households with contacts to countries with high incidence of TB and/or leprosy
• Neonates in any other locally identified risk group for TB and/or leprosy
In a few countries with low TB incidence, BCG vaccination is largely replaced by intensified case
detection, contact tracing and supervised early treatment.
Need for revaccination
Studies show minimal or no evidence of any additional benefit of repeat BCG vaccination
against TB or leprosy. Therefore, revaccination is not recommended even if the TST reaction or
result of an IGRA is negative. The absence of a BCG scar after vaccination is not indicative of a
lack of protection and is not an indication for revaccination.
12 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
BCG vaccination for HIV-infected infants
Children who are HIV-infected when vaccinated with BCG at birth are at increased risk of
developing disseminated BCG disease. However, if HIV-infected individuals including children,
are receiving ART, are clinically well and immunologically stable (CD4% >25% for children aged
<5 years or CD4 count ≥200 if aged >5 years) they should be vaccinated with BCG.
• In general, populations with high prevalence of HIV infection also have the greatest burden of
TB; in such populations the benefits of potentially preventing severe TB through vaccination
at birth are outweighed by the risks associated with the use of BCG vaccine. Therefore, it is
recommended that in such populations:
– Neonates born to women of unknown HIV status should be vaccinated as the benefits of
BCG vaccination outweigh the risks.
– Neonates of unknown HIV status born to HIV-infected women should be vaccinated if they
have no clinical evidence suggestive of HIV infection, regardless of whether the mother is
receiving ART.
– Although evidence is limited, for neonates with HIV infection confirmed by early virological
testing, BCG vaccination should be delayed until ART has been started and the infant
confirmed to be clinically and immunologically stable (CD4% >25%).
TB preventive treatment: WHO consolidated guidelines on tuberculosis. Module 1 –
prevention – tuberculosis preventive treatment, 2020 (14)
Identifying populations for TB infection testing and TPT – people living with HIV
Adults and adolescents living with HIV who are unlikely to have TB disease should receive TB
preventive treatment as part of a comprehensive package of HIV care. Treatment should also
be given to those on antiretroviral treatment, to pregnant women and to those who have
previously been treated for TB, irrespective of the degree of immunosuppression and even if TB
infection testing is unavailable.
(Strong recommendation, high certainty in the estimates of effect)
Infants aged <12 months living with HIV who are in contact with a person with TB and who
are unlikely to have TB disease on an appropriate clinical evaluation or according to national
guidelines should receive TB preventive treatment.
(Strong recommendation, moderate certainty in the estimates of effect)
Children aged ≥12 months living with HIV who are considered unlikely to have TB disease on
an appropriate clinical evaluation or according to national guidelines should be offered TB
preventive treatment as part of a comprehensive package of HIV prevention and care if they
live in a setting with high TB transmission, regardless of contact with TB.
(Strong recommendation, low certainty in the estimates of effect)
All children living with HIV who have successfully completed treatment for TB disease may
receive TB preventive treatment.
(Conditional recommendation, low certainty in the estimates of effect)
Identifying populations for TB infection testing and TB preventive treatment – household
contacts (regardless of HIV status)
Children aged <5 years who are household contacts of people with bacteriologically confirmed
pulmonary TB and who are found not to have TB disease on an appropriate clinical evaluation
or according to national guidelines should be given TB preventive treatment even if TB infection
testing is unavailable.
(Strong recommendation, high certainty in the estimates of effect)
3. Prevention of TB 13
Children aged ≥5 years adolescents and adults who are household contacts of people with
bacteriologically confirmed pulmonary TB who are found not to have TB disease by an
appropriate clinical evaluation or according to national guidelines may be given TB preventive
treatment.
(Conditional recommendation, low certainty in the estimates of effect)
In selected high-risk household contacts of patients with multidrug-resistant tuberculosis,
preventive treatment may be considered based on individualized risk assessment and a sound
clinical justification.
(Conditional recommendation, very low certainty in the estimates of effect)
Identifying populations for TB infection testing and TB preventive treatment – other
people at risk
(these populations may include children and adolescents)
People who are initiating anti-TNF20 treatment, or receiving dialysis, or preparing for an organ
or haematological transplant, or who have silicosis should be systematically tested and treated
for TB infection.
(Strong recommendation, low to very low certainty in the estimates of effect)
Systematic TB infection testing and treatment may be considered for prisoners, health workers,
immigrants from countries with a high TB burden, homeless people and people who use drugs.
(Conditional recommendation, low to very low certainty in the estimates of effect)
Systematic TB infection testing and treatment is not recommended for people with diabetes,
people who engage in the harmful use of alcohol, tobacco smokers and underweight people
unless they also belong to other risk groups included in the above recommendations.
(Conditional recommendation, very low certainty in the estimates of effect)
Algorithms to rule out TB disease
Adults and adolescents living with HIV should be screened for TB according to a clinical
algorithm. Those who do not report any of the symptoms of current cough, fever, weight loss
or night sweats are unlikely to have TB disease and should be offered preventive treatment,
regardless of their antiretroviral treatment (ART) status.
(Strong recommendation, moderate certainty in the estimates of effect)
Adults and adolescents living with HIV who are screened for TB according to a clinical algorithm
and who report any of the symptoms of current cough, fever, weight loss or night sweats may
have TB disease and should be evaluated for TB and other diseases and offered preventive
treatment if TB disease is excluded.
(Strong recommendation, moderate certainty in the estimates of effect)
Chest radiography may be offered to people living with HIV on ART and preventive treatment
given to those with no abnormal radiographic findings.
(Conditional recommendation, low certainty in the estimates of effect)
Infants and children living with HIV who have poor weight gain, fever or current cough or who
have a history of contact with a person with TB should be evaluated for TB and other diseases
that cause such symptoms. If TB disease is excluded after an appropriate clinical evaluation
or according to national guidelines, these children should be offered TB preventive treatment,
regardless of their age.
(Strong recommendation, low certainty in the estimates of effect)
20
TNF: tumour necrosis factor
14 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
The absence of any symptoms of TB and the absence of abnormal chest radiographic findings
may be used to rule out TB disease among HIV-negative household contacts aged ≥5 years
and other risk groups before preventive treatment.
(Conditional recommendation, very low certainty in the estimates of effect)
Testing for TB infection
Either a tuberculin skin test (TST) or interferon-gamma release assay (IGRA) can be used to test
for TB infection.
(Strong recommendation, very low certainty in the estimates of effect)
TB preventive treatment options
The following options are recommended for the treatment of TB infection regardless of HIV
status: 6 or 9 months of daily isoniazid, or a 3-month regimen of weekly rifapentine plus
isoniazid,21 or a 3-month regimen of daily isoniazid plus rifampicin.
(strong recommendation, moderate to high certainty in the estimates of effect).
A 1-month regimen of daily rifapentine plus isoniazid22 or 4 months of daily rifampicin alone
may also be offered as alternatives.
(Conditional recommendation, low to moderate certainty in the estimates of effect)
In settings with high TB transmission, adults and adolescents living with HIV who have an
unknown or a positive TB infection test, and are unlikely to have TB disease, should receive
at least 36 months of daily isoniazid preventive therapy (IPT). Daily IPT for 36 months
should be given whether or not the person is on ART, and irrespective of the degree of
immunosuppression, history of previous TB treatment and pregnancy in settings considered to
have a high TB transmission as defined by national authorities.
(Conditional recommendation, low certainty in the estimates of effect)
21
In children 2 years and above.
22
In children 13 years and above.
3. Prevention of TB 15
16 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
4. Diagnostic approaches for TB
in children and adolescents
Of the estimated 1.1 million children who developed TB annually, only 399 000 (36.5%) were notified
to NTPs in 2020. Under-notification is worst among children below 5 years of age, with only 27.5% of
children with TB being notified (1). These ‘missing’ children are not diagnosed and/or not reported.
TB-related mortality among children below 15 years of age was estimated at 226 000 for 2020 (1).
Modelling has shown that 80% of TB-related deaths are among children under 5 years of age, and
that 96% of children who die of TB, did not access treatment (15).
The low case detection rate among (young) children is due to several factors including: the fact that
young children have paucibacillary TB and do not excrete enough bacilli to be detectable by available
bacteriological tests; the lack of a sensitive point-of-care diagnostic test; difficulties in collecting suitable
respiratory samples for bacteriological confirmation; and misdiagnosis due to the overlap of non-
specific symptoms of TB with other common childhood diseases. Children often first access care at
the PHC level, where there may be little or no awareness and capacity to diagnose and manage
children with TB (this includes capacity for sample collection, access to bacteriological tests and CXR,
as well as capacity and confidence in making a clinical diagnosis when bacteriological testing is not
available or is negative). In addition, paediatric TB services are often highly centralized at secondary
or tertiary levels of the health system and managed in a vertical, non-integrated way. TB screening
is often not systematically incorporated into clinical algorithms for child health (10).
This chapter contains two new recommendations relevant to the diagnosis of TB in children and
adolescents as well as other valid WHO recommendations that apply to children and adolescents
on TB diagnosis. The two new recommendations relate to the use of the Xpert Ultra assay in gastric
aspirate and stool specimens for the diagnosis of PTB and detection of rifampicin resistance among
children, and the use of integrated treatment decision algorithms23 for the diagnosis of PTB in children.
Prior to 2022, Xpert Ultra had already been recommended for use in nasopharyngeal aspirate (NPA)
and sputum specimens; hence, the 2022 recommendation widens the number of specimens that can
be used and aligns this with the WHO recommendation on Xpert MTB/RIF (which WHO recommends
for use in gastric aspirate, NPA, sputum and stool specimens to diagnose PTB and detect rifampicin
resistance) (16). The two new recommendations are described in detail in this chapter.
Section 4.3 consolidates recommendations from current WHO guidelines on rapid diagnostic tests and
the use of commercial serodiagnostic tests, namely the WHO consolidated guidelines on tuberculosis.
Module 3: diagnosis – rapid diagnostics for tuberculosis detection, 2021 update (16) and the Commercial
serodiagnostic tests for diagnosis of tuberculosis: policy statement (17). For more information on each
recommendation including the remarks, source of evidence, justification, subgroup, implementation
and monitoring and evaluation considerations, the source guidelines or WHO’s TB KSP should be
consulted.
The WHO convened an expert consultation on the classification of intrathoracic TB disease among
children in September 2021. The experts reviewed evidence from the point of view of pathophysiology,
23
Treatment decision algorithms are defined as: A flow chart allocating evidence-based scores to microbiological, clinical and radiological
features that allow clinicians to make a decision regarding starting TB treatment in children.
18 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Evidence: In preparation for the GDG meeting on the management of TB in children and adolescents,
an update of the review on the diagnostic accuracy of Xpert MTB/RIF and Xpert Ultra for TB disease
in children (18) was performed. A systematic search of the literature was carried out in January 2021.
This update focused specifically on the diagnostic accuracy of Xpert Ultra in gastric aspirate or
lavage specimens and stool specimens for the diagnosis of PTB and rifampicin resistance in children
below 10 years of age. Nine studies that evaluated the diagnostic accuracy of Xpert Ultra in gastric
aspirate and/or stool specimens in children were identified. For the meta-analysis, six studies (653
participants) provided data for gastric specimens (19–24) and six studies (1278 participants) for stool
specimens (20, 23–27). The review found that for gastric aspirate, Xpert Ultra sensitivity was 64% in
children 0–9 years, against a microbiological reference standard and specificity was 95%. For stool,
Xpert Ultra sensitivity was 53% in children 0–9 years, against the microbiological reference standard
and specificity was 98%. Sensitivity estimates against a composite reference standard were lower for
both specimen types. There were no studies that evaluated the diagnostic accuracy of Xpert Ultra
for detection of rifampicin resistance using gastric aspirate or stool specimens.
Of the nine studies, eight (89%) reported the proportion of Xpert Ultra positive results that were trace
results. In these eight studies, of the total Xpert Ultra positive results, the proportion (expressed as a
percentage) of Ultra trace results ranged from 0% to 66% (median 52%) in studies evaluating gastric
specimens and from 0% to 84% (median 52%) in studies evaluating stool specimens.
GDG considerations: The GDG members discussed that all TB tests, including the microbiological
reference standard (usually culture or Xpert Ultra on respiratory samples) have suboptimal sensitivity
in children, due to the paucibacillary nature of TB disease in this age group. Therefore, the panel
highlighted that a positive test accurately determines a case of TB disease, but a negative test does not
exclude TB disease. As no studies assessed the diagnostic accuracy for Xpert Ultra for the detection
of rifampicin resistance in children, conclusions related to the use of Xpert Ultra for the diagnosis of
rifampicin resistance were based on data in adults, evaluated for the 2020 rapid diagnostic guidelines.
The GDG members agreed that the desirable effects from the test in both specimens are moderate,
but large for the detection of rifampicin resistance, because of the large impact on the choice of
effective treatment when rifampicin resistance is detected. The panel noted that false-negative results
should be interpreted within the context of the clinical picture, and clinicians should not solely use a
negative test to rule out disease. The GDG judged that the balance of effects favours the intervention
(considering both the moderate desirable effects and small undesirable effects), based on moderate
certainty evidence for both gastric aspirate and stool samples.
The GDG judged that more time and training-related resources would be required to collect gastric
aspirate specimens, and that the procedure would also often require hospital admission. Stool
processing may result in some additional time requirement to prepare the sample, depending on
the processing method. The panel decided upon comparing the cost of doing the tests on the sample
types versus not doing the test. The panel then concluded that the costs for both specimen types vary.
While there were no studies on the cost-effectiveness of gastric aspirate, a cost-effectiveness study
compared stool testing against clinical diagnosis at PHCs in Uganda, assuming that no respiratory
samples would be collected without referral to the hospital or without invasive sampling (see web
annex 4). Compared to that standard of care, stool testing was more effective, but also more costly.
The GDG members discussed that an Xpert Ultra test on stool samples becomes cost-effective even at
a TB prevalence level of 3% at a zero discount rate (the practice of discounting future health effects).
Implementation aspects such as the importance of obtaining microbiological confirmation and the
detection of rifampicin resistance would further favour cost-effectiveness of stool testing. The panel
therefore concluded that the evidence probably favours Xpert Ultra testing on stool samples.
In terms of acceptability, although the gastric aspirate sample collection was found to be an invasive
procedure (that could be uncomfortable to children), the panel felt that the procedure is probably
acceptable to children, caregivers and health care workers (HCW), considering the role of this procedure
to obtain bacteriological confirmation at higher level facilities. The GDG noted the importance of
20 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
systems need to be functional to ensure that children who need to be tested using gastric samples
reach the appropriate level of care.
Stool samples: Stool sampling has the advantage of being non-invasive, with the collection generally
perceived as easy and feasible by HCWs and caregivers, irrespective of the clinical condition of the
child. The possible drawback is that children may not be able to pass stool on command causing delay
in sample collection. Enhancing awareness and sensitizing caregivers on stool as a suitable sample
for TB testing is important, to enhance acceptability and implementation. Stool processing is also
an acceptable procedure for laboratory technicians and regarded as a good alternative to sputum
samples. Relatively high rates of non-determinate Xpert Ultra results (including error, invalid, or no
results) were reported in the studies included in the systematic review. These rates varied from less
than 1% to 10% and may depend on the stool processing method used.
Stool processing methods: An analysis of preliminary results of a head-to-head comparison of
three centrifuge-free stool processing methods combined with Xpert Ultra was conducted to inform
implementation considerations on the use of Xpert Ultra in stool samples. The three processing
methods were optimized sucrose flotation (OSF) (28), simple one step stool (SOS) (29) and stool
processing kit (SPK) (25). The results at the time of the GDG meeting showed a similar performance
of Xpert Ultra in combination with the three stool processing methods, in terms of sensitivity and
specificity. All methods were found to be easy to process by laboratory staff at the reference laboratory
level and had a high ease-of-use score. However, most users considered that these methods cannot
be performed by non-laboratory personnel (such as nurses or HCWs) in PHC settings without access
to a laboratory. Overall, the SOS method appeared to be the preferred method as it does not require
additional equipment and is comparable to sputum processing using Xpert Ultra. The SPK is still at
prototype stage but will not be commercialized; the optimized sucrose flotation is still under validation,
and its development into a kit format to simplify some of the steps is envisaged in the near future.
Therefore, either of the available centrifuge-free stool processing methods may be used, depending
on local preference and laboratory infrastructure.
Xpert Ultra trace results: Trace results are common with the use of Xpert Ultra in all paediatric
specimen types, reflecting the paucibacillary nature of TB disease in children. For children as well as
people living with HIV who are being evaluated for PTB, and for persons being evaluated for EPTB,
the “M. tuberculosis complex (MTBC) detected trace” Ultra result is considered as bacteriological
confirmation of TB (30). This is an important implementation consideration, in view of the risk of
morbidity and mortality in these populations. Trace results will have an indeterminate result for
rifampicin resistance; therefore, alternative specimens may need to be collected for Xpert Ultra
processing in persons with a high likelihood of drug resistance.
24
Definitions and reporting framework for tuberculosis – 2013 revision (updated December 2014 and January 2020). Geneva: World
Health Organization; 2013.
25
WHO operational handbook on tuberculosis. Module 5: management of tuberculosis in children and adolescents (https://apps.who.
int/iris/bitstream/handle/10665/352523/9789240046832-eng.pdf ).
22 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
was developed in an attempt to operationalize the 2014 WHO guidance (8) by outlining the steps
that a health care worker should take in evaluating a child with presumptive PTB.
To make full use of available data for comparing the performance of these algorithms in different
settings, missing variables were imputed, heterogeneous definitions of variables were collapsed, and
slight modifications were made to the algorithms or scores to enable the use of the variables available
in the IPD. A total of 14 studies, comprising 5494 records were included, of which 4811 records were
included in this analysis (26, 33–47). Studies from 13 countries (including 12 high TB, TB/HIV and/or
MDR-TB burden countries) within 5 of the 6 WHO regions were included. The cohort in the meta-
analysis used to inform this recommendation had a median age of 26 months (interquartile range
13.4–58.3); 38% of the children had TB, of which 30% was bacteriologically confirmed; 20% of the
children were living with HIV infection; and 14% had SAM.
Seven algorithms or scoring systems were identified for evaluation, comprising the Uganda National
TB/Leprosy Control Program (NTLP) algorithm (48), the Brazilian Ministry of Health Child PTB Scoring
System (49), the Gunasekera et al. 2021 algorithm (50), the Keith Edward score (51), the Marcy et al.
2019 algorithm (52), the Stegen-Toledo score (53), and the Marais et al. 2006 criteria (54). The pooled
estimates of the sensitivity and specificity of each algorithm/score were compared to the standard of
care algorithm (i.e. The Union Desk Guide Algorithm, which constituted the reference standard (32))
for children aged below 10 years, for children living with HIV, for children with SAM and for children
aged below 1 year.
For the overall population of children under the age of 10 years, the pooled sensitivity of the seven
algorithms or scoring systems ranged from 16% (Marais et al. criteria) to 95% (Gunasekera et al.
algorithm), while the pooled specificity ranged from 9% (Gunasekera et al. algorithm) to 89% (Marais
et al. criteria) (see web annex 3).
GDG considerations: The GDG felt that algorithms with clinical criteria have an important role to
play in making decisions on starting children on TB treatment, particularly at peripheral levels of the
health care system. There was strong consensus among the GDG members about the need and
importance of working on treatment decision algorithms to improve the gaps in TB case detection in
children. An important advantage of evidence-based algorithms (as it allocates a modelled weight to
features of clinical evaluation), is that this modelling process allows for specification of the weight of
certain clinical features, rather than being based solely on expert opinion. The panel highlighted that
data in the IPD were mainly from tertiary settings where the proportion of children with confirmed
TB is higher than at district hospital or PHC level. It was acknowledged that the IPD had a high level
of heterogeneity. Conducting a meta-regression analysis by level of the health care system was not
possible because of the limited number of studies.
The GDG concluded that none of the evaluated algorithms were optimal in terms of either sensitivity
or specificity, combined with the very low certainty of evidence. The GDG also noted that algorithms
with a high sensitivity (i.e. low number of false negatives) generally have a low specificity (i.e. a high
number of false positives) and vice versa. The panel reflected on the consequences of false negative
and false positive conclusions based on integrated treatment decision algorithms and agreed that
it was most important to avoid missing a TB diagnosis in a child who has TB, considering the large
case detection gap and the consequences of a missed diagnosis of TB.
During the GDG deliberations, the following options that emerged from the evidence review were
discussed: (i) choose one of the algorithms reviewed for a possible recommendation; (ii) make a
generic recommendation on the use of integrated treatment decisions algorithms and present new
evidence-based algorithms in the operational handbook; (iii) make a statement about the need for
further research on treatment decision algorithms, affirming the need for such algorithms.
The GDG judged the available evidence as inappropriate to support a recommendation for any specific
algorithm; and instead decided to make a generic recommendation on the use of treatment decision
24 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
The decision to start treatment is linked to other recommendations in these guidelines, such as
shortening of the treatment duration for children with non-severe forms of TB and on decentralization
of TB services. Once a decision to start TB treatment has been made, the severity of disease needs
to be assessed to inform the duration of treatment. Detailed criteria for assessing severity of disease
are described in the operational handbook.
Referral: Defining the criteria for referral of children evaluated for PTB at peripheral levels of the
health care system using the algorithms is important. Examples of subgroups in need of referral
include infants, children with presumptive severe forms of EPTB (such as TBM, disseminated TB and
osteoarticular TB) and children with presumptive DR-TB in regions with a high prevalence of DR-TB.
Children presenting with severe acute pneumonia need referral to the appropriate level of care for
oxygen supplementation, while children with SAM need to be provided with appropriate nutritional
support. A high index of suspicion is important among infants with acute symptoms who are contacts
of people with bacteriologically confirmed TB, to make a treatment decision as soon as possible rather
than wait for symptoms to persist. This is due to the potential for rapid deterioration in the clinical
condition of infants and development of severe TB disease.
Clinical monitoring of children started on TB treatment: It is important to acknowledge that the
preference for sufficient sensitivity of the algorithms to detect and treat children with TB will mean
that some children who do not have TB will be treated with TB treatment. The risk of severe drug-
related toxicity in children is very low, and shorter regimens for non-severe TB (see chapter 5) will
further reduce the risks related to treatment. However, it will be critical to monitor children started on
TB treatment and to refer them for evaluation for other diseases and appropriate treatment if they
fail to respond to TB treatment within 1 month.
Implementation in high DR-TB burden settings: Integrated treatment decision algorithms may
be implemented in settings with a high burden of DR-TB. Seeking bacteriological confirmation using
appropriate paediatric samples and WHO recommended rapid diagnostic tests (such as Xpert MTB/
RIF or Ultra) is critical among children who have a history of contact with a source case with confirmed
or highly likely DR-TB (including a TB patient not responding to treatment, or a source case who died
of TB while on treatment). Once a decision to treat a child without bacteriological confirmation for TB
has been made based on the algorithm, risk factors for the child having DR-TB need to be assessed.
Clinicians need to keep a high index of suspicion for DR-TB in these children and ensure they are
tested and managed for DR-TB as appropriate (see chapter 5).
26 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
In adults and children with signs and symptoms of extrapulmonary TB, Xpert MTB/RIF may
be used in lymph node aspirate, lymph node biopsy, pleural fluid, peritoneal fluid, pericardial
fluid, synovial fluid or urine specimens as the initial diagnostic test for respective form of
extrapulmonary TB rather than smear microscopy/culture.
(Conditional recommendation, moderate certainty of evidence for test accuracy for pleural fluid;
low certainty for lymph node aspirate, peritoneal fluid, synovial fluid, urine; very low certainty for
pericardial fluid, lymph nodes biopsy)
In adults and children with signs and symptoms of extrapulmonary TB, Xpert Ultra may be used
in lymph node aspirate and lymph node biopsy as the initial diagnostic test for lymph nodes TB
rather than smear microscopy/culture.
(Conditional recommendation, low certainty of evidence)
In adults and children with signs and symptoms of extrapulmonary TB, Xpert MTB/RIF or Xpert
Ultra should be used for rifampicin-resistance detection rather than culture and phenotypic DST.
(Strong recommendation, high certainty of evidence for test accuracy for Xpert MTB/RIF; low
certainty of evidence for Xpert Ultra)
In HIV-positive adults and children with signs and symptoms of disseminated TB, Xpert MTB/RIF
may be used in blood, as an initial diagnostic test for disseminated TB.
(Conditional recommendation, very low certainty of evidence for test accuracy)
Recommendations on Xpert MTB/RIF and Xpert Ultra repeated testing in children and
adolescents with signs and symptoms of pulmonary TB
In adults* with signs and symptoms of pulmonary TB who have an Xpert Ultra trace positive
result on the initial test, repeated testing with Xpert Ultra may not be used.
(Conditional recommendation, very low certainty of evidence for test accuracy)
Remark: Xpert Ultra trace results in adolescents will require follow-up, including reassessing
clinical symptoms and information on prior history of TB. In the case of suspected rifampicin
resistance, repeated testing may provide additional benefit for detection as well as an initial
attempt to assess rifampicin resistance. For interpretation of trace results in children, see
section 4.1.3.
* Adults and adolescents from 15 years
In children with signs and symptoms of pulmonary TB in settings with pretest probability below
5% and an Xpert MTB/RIF negative result on the initial test, repeated testing with Xpert MTB/RIF
in sputum, gastric fluid, nasopharyngeal aspirate or stool specimens may not be used.
(Conditional recommendation, low certainty of evidence for test accuracy for sputum; and very
low for other specimen types)
In children with signs and symptoms of pulmonary TB in settings with pretest probability 5% or
more and an Xpert MTB/RIF negative result on the initial test, repeated testing with Xpert MTB/
RIF (for total of two tests) in sputum, gastric fluid, nasopharyngeal aspirate and stool specimens
may be used.
(Conditional recommendation, low certainty of evidence for test accuracy for sputum; and very
low for other specimen types)
In children with signs and symptoms of pulmonary TB in settings with pretest probability below
5% and an Xpert Ultra negative result on the initial test, repeated testing with Xpert Ultra in
sputum or nasopharyngeal aspirate specimens may not be used.
(Conditional recommendation, very low certainty of evidence for test accuracy)
In adults* in the general population who had either a positive TB symptom screen or chest
radiograph with lung abnormalities or both, one Xpert Ultra test may be used rather than two
Xpert Ultra tests as the initial test for pulmonary TB.
(Conditional recommendation, very low certainty of evidence for test accuracy).
* Adults and adolescents from 15 years
Truenat MTB, MTB Plus and Truenat MTB-RIF Dx in adults and children with signs and
symptoms of pulmonary TB (specimen type: sputum)
In children and adults* with signs and symptoms of pulmonary TB, the Truenat MTB or MTB
Plus may be used as an initial diagnostic test for TB rather than smear microscopy/culture.
(Conditional recommendation, moderate certainty of evidence for test accuracy)
* Adults and adolescents from 15 years
In children and adults* with signs and symptoms of pulmonary TB and a Truenat MTB or MTB
Plus positive result, Truenat MTB-RIF Dx may be used as an initial test for rifampicin resistance
rather than culture and phenotypic DST.
(Conditional recommendation, very low certainty of evidence for test accuracy)
* Adults and adolescents from 15 years
Moderate complexity automated nucleic acid amplification tests (NAATs) for detection of
TB and resistance to rifampicin and isoniazid
In people with signs and symptoms of pulmonary TB, moderate complexity automated NAATs
may be used on respiratory samples for the detection of pulmonary TB, and of rifampicin and
isoniazid resistance, rather than culture and phenotypic DST.
(Conditional recommendation, moderate certainty of evidence for diagnostic accuracy)
Loop-mediated isothermal amplification
TB-LAMP may be used as a replacement test for sputum-smear microscopy for diagnosing
pulmonary TB in adults* with signs and symptoms consistent with TB.
(Conditional recommendation, very low certainty evidence)
* Adults and adolescents from 15 years
28 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
TB-LAMP may be used as a follow-on test to smear microscopy in adults* with signs and
symptoms consistent with pulmonary TB, especially when further testing of sputum smear-
negative specimens is necessary.
(Conditional recommendation, very low certainty evidence)
* Adults and adolescents from 15 years
Lateral flow urine lipoarabinomannan assay
In inpatient settings, WHO strongly recommends using LF-LAM to assist in the diagnosis of TB
disease in HIV-positive adults and children:
a. with signs and symptoms of TB (pulmonary and/or extrapulmonary) (strong recommendation,
moderate certainty in the evidence about the intervention effects) or
b. with advanced HIV disease or who are seriously ill (strong recommendation, moderate
certainty in the evidence about the intervention effects); or
c. irrespective of signs and symptoms of TB and with a CD4 cell count of less than 200 cells/
mm3 (strong recommendation, moderate certainty in the evidence about the intervention effects).
In outpatient settings, WHO suggests using LF-LAM to assist in the diagnosis of TB disease in
HIV-positive adults and children:
a. with signs and symptoms of TB (pulmonary and/or extrapulmonary) or seriously ill
(conditional recommendation, low certainty in the evidence about test accuracy); and
b. irrespective of signs and symptoms of TB and with a CD4 cell count of less than
100 cells/mm3 (conditional recommendation, very low certainty in the evidence about
test accuracy)
In outpatient settings, WHO recommends against using LF-LAM to assist in the diagnosis of TB
disease in HIV-positive adults and children:
a. without assessing TB symptoms (strong recommendation, very low certainty in the evidence
about test accuracy);
b. without TB symptoms and unknown CD4 cell count or without TB symptoms and CD4 cell
count greater than or equal to 200 cells/mm3 (strong recommendation, very low certainty in the
evidence about test accuracy); and
c. without TB symptoms and with a CD4 cell count of 100–200 cells/mm3 (conditional
recommendation, very low certainty in the evidence about test accuracy).
Low complexity NAATs for detection of resistance to isoniazid and second-line TB agents
In people with bacteriologically confirmed pulmonary TB, low complexity automated NAATs
may be used on sputum for the initial detection of resistance to isoniazid and fluoroquinolones,
rather than culture-based phenotypic DST.
(Conditional recommendation, moderate certainty of evidence for diagnostic accuracy)
In people with bacteriologically confirmed pulmonary TB and resistance to rifampicin, low
complexity automated NAATs may be used on sputum for the initial detection of resistance to
ethionamide, rather than DNA sequencing of the inhA promoter.
(Conditional recommendation, very low certainty of evidence for diagnostic accuracy)
In people with bacteriologically confirmed pulmonary TB and resistance to rifampicin, low
complexity automated NAATs may be used on sputum for the initial detection of resistance to
amikacin, rather than culture-based phenotypic DST.
(Conditional recommendation, low certainty of evidence for diagnostic accuracy)
30 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
5. Treatment of TB disease in
children and adolescents
This chapter contains four new recommendations (published for the first time and described in full
detail here) relevant to the treatment of TB disease in children and adolescents as well as other valid
WHO recommendations that apply to the treatment of TB in children and adolescents (section 5.4).
The four new recommendations concern drug-susceptible TB (DS-TB): (i) a four-month regimen for
children and adolescents below 16 years of age with non-severe drug-susceptible TB (section 5.1);
(ii) a six-month intensive regimen to treat TBM composed of isoniazid, rifampicin, pyrazinamide and
ethionamide (section 5.2); (iii) use of bedaquiline as part of shorter or longer regimens for children
of all ages to treat MDR/RR-TB (section 5.3.1); and (iv) use of delamanid as part of longer regimens
for children of all ages to treat MDR/RR-TB (section 5.3.2).
The recommendations in this chapter have been consolidated from current WHO guidelines on
TB treatment, namely the WHO consolidated guidelines on tuberculosis. Module 4: treatment –
drug-susceptible tuberculosis treatment, 2022 update (55) and the WHO consolidated guidelines on
tuberculosis. Module 4: treatment – drug-resistant tuberculosis treatment, 2020 update (9). Where
appropriate, details on recommendations from the 2014 Guidance for national tuberculosis programmes
on the management of tuberculosis in children (second edition) (8), and Rapid advice: treatment of
tuberculosis in children (56), which remain relevant have been included. For more information on each
recommendation including the remarks, source of evidence, justification, subgroup, implementation
and monitoring and evaluation considerations, the source guidelines or WHO TB KSP should be
consulted.
26
Defined as countries, subnational administrative units, or selected facilities, where the HIV prevalence among adult pregnant women is
≥1% or among TB patients is ≥5% in the 2014 Guidance for national tuberculosis programmes on the management of tuberculosis in
children (second edition) (8).
27
WHO does not intend to establish thresholds for low, moderate or high levels of prevalence of isoniazid resistance; NTPs will establish
definitions for their own countries.
32 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Evidence: In the SHINE trial, the primary efficacy outcome was a composite of treatment failure
(including an extension of treatment beyond the replacement of missed doses, TB treatment drug
changes or restarts due to suspected treatment failure), on-treatment loss-to-follow-up, TB recurrence
or death by 72 weeks (from randomization), excluding children not reaching 16 weeks follow-up
(modified-intention-to-treat). The non-inferiority margin for the primary efficacy outcome was 6%.
The primary safety outcome was grade 3–5 adverse events recorded while on TB treatment.
The SHINE trial definition of non-severe TB was: peripheral lymph node TB or respiratory TB (including
uncomplicated intrathoracic lymph node disease) confined to one lobe without cavities, no significant
airway obstruction, uncomplicated pleural effusion, and no miliary TB.
The SHINE trial inclusion criteria were: children and young adolescents aged <16 years; weight ≥3 kg;
no known drug resistance; symptomatic but non-severe TB; smear negative on gastric aspirate or other
respiratory sample (an Xpert MTB/RIF positive, rifampicin susceptible result was allowed);28 clinician’s
decision to treat with a standard first-line regimen; not treated for TB in the previous two years; known
HIV status (positive or negative). Trial exclusion criteria were: respiratory sample acid fast bacilli smear-
positive (a smear-positive peripheral lymph node sample was allowed); premature birth (<37 weeks)
and aged under 3 months; miliary TB, spinal TB, TBM, osteoarticular TB, abdominal TB, congenital TB;
pre-existing, non-tuberculous disease likely to prejudice the response to, or assessment of, treatment
(such as liver or kidney disease, peripheral neuropathy or cavitation); any known contraindication to
taking TB drugs; known contact with a drug-resistant adult source case (including mono-resistant
TB); known drug-resistance in the child; being severely ill; pregnancy.
A total of 1204 children were enrolled in the trial between July 2016 and July 2018. The median age of
enrolled children was 3.5 years (range: 2 months – 15 years), 52% were male, 11% had HIV-infection,
and 14% had bacteriologically confirmed TB. Retention in the trial by 72 weeks and adherence29 to
allocated TB treatment were 95% and 94%, respectively. Sixteen (2.8%) versus 18 (3.1%) children
reached the primary efficacy outcome (treatment failure) in the 16- versus 24-week arms respectively,
with an unadjusted difference of -0.3% (95% CI: -2.3, 1.6). Treatment success was reported in 97.1%
of participants receiving the 16-week regimen versus 96.9% in those receiving the 24-week regimen
(relative risk (RR): 1.00, 95% CI: 0.98–1.02). Non-inferiority of the 16-week regimen was consistent
across all intention-to-treat, per-protocol and key secondary analyses. This included restricting the
analysis to the 958 (80%) children that were independently adjudicated to have TB at baseline by
the trial Endpoint Review Committee. A total of 7.8% of children experienced a grade 3–5 adverse
event in the 16-week arm, versus 8.0% in the 24-week arm (RR: 0.98, 95% CI: 0.67–1.44). There were
115 on-treatment grade ≥3 adverse events in 95 (8%) children, 47 (8%) in the 16-week and 48 (8%)
in the 24-week arm, most common being pneumonia or other chest infections (29 (25%)) or liver-
related events (11 (10%)) across both arms. There were 17 grade 3 or 4 adverse reactions (considered
possibly, probably or definitely) related to trial drugs, including 11 hepatic events; all adverse reactions
except three occurred in the first eight weeks of treatment.
GDG considerations: The GDG judged that while the desirable effects related to this PICO question
are related to treatment outcomes, shortening the duration of treatment is also important and
desirable (as reducing the length of treatment could make treatment easier for children and caregivers
as well as reduce cost for families and the health system). The GDG discussed that since the SHINE
trial was a non-inferiority trial, no difference in unfavourable outcomes between the two arms is what
the trial aimed to detect. Therefore, both desirable and undesirable effects were judged by most GDG
members as trivial. Since non-inferiority of the 4-month regimen was demonstrated in the trial, the
balance of effects was judged to not favour either the shorter or the longer duration of treatment.
28
In the SHINE trial, children with Xpert MTB/RIF results had very low or low semi-quantitative results, or a negative result. Xpert Ultra
was not used in the SHINE trial.
29
In the SHINE trial, adherence was defined as the proportion of children who received an adequate amount of treatment (as defined in
the statistical analysis plan for both the intervention and control regimens; generally, a cut off of 80% of the allocated doses was used,
within a certain time frame of starting each phase of treatment (i.e. intensive phase versus continuation phase).
34 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
disease was received following a call for data. Therefore, infants aged 0–3 months with suspected or
confirmed PTB or tuberculous peripheral lymphadenitis should be promptly treated with the 6-month
treatment regimen (2HRZ(E)/4HR), as per the existing recommendation from the 2014 Guidance for
national tuberculosis programmes on the management of tuberculosis in children (second edition) (8).
Treatment may require dose adjustment to reconcile the effect of age and possible toxicity in young
infants. The decision to adjust doses should be taken by a clinician experienced in the management
of paediatric TB.
Children treated for TB in the past two years: Given the increased risk of treatment failure and
of drug resistance, children and adolescents treated in the preceding two years were not eligible for
inclusion in the SHINE trial; they should be treated with the 6-month treatment regimen (2HRZ(E)/4HR).
30
This level of resistance was defined as countries, subnational administrative units, or selected facilities, where the HIV prevalence
among adult pregnant women is ≥1% or among TB patients is ≥5% in the 2014 Guidance for national tuberculosis programmes on the
management of tuberculosis in children (second edition) (8).
31
WHO does not intend to establish thresholds for low, moderate or high levels of prevalence of isoniazid resistance; instead NTPs will
establish definitions for their own countries.
36 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
5.2. Treatment regimens for TB meningitis in children
and adolescents
Recommendation
In children and adolescents with bacteriologically confirmed or clinically diagnosed TB meningitis
(without suspicion or evidence of MDR/RR-TB), a 6-month intensive regimen (6HRZEto) may be
used as an alternative option to the 12-month regimen (2HRZE/10HR)
(Conditional recommendation, very low certainty of the evidence).
Remarks
• The shorter intensive regimen is suitable for children and adolescents who have no evidence of
drug resistance and in children and adolescents who have a low likelihood of drug-resistant TB,
e.g. those without risk factors for any form of drug-resistant TB.
• The recommendation from the Guidance for national tuberculosis programmes on the
management of tuberculosis in children (second edition, 2014) remains an option for the treatment
of children and adolescents with suspected or confirmed TB meningitis (TBM): Children and
adolescents with suspected or confirmed tuberculous meningitis should be treated with a four-
drug regimen (HRZE) for 2 months, followed by a two-drug regimen (HR) for 10 months, the total
duration of treatment being 12 months (Strong recommendation, low certainty of evidence).
• Due to a lack of data, the shorter intensive treatment regimen recommendation should not be
used in children and adolescents living with HIV who are diagnosed with TB meningitis.
38 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
treatment success were 78.5%–100.0%, with a random effects pooled proportion of 95.0% (95% CI:
74.0–99.0) among studies of the intervention regimen, and 70%–85.7%, with a random effects pooled
proportion of 75.0% (95% CI: 69.0–81.0) among studies of the comparator regimen. Neurological
sequelae were defined and assessed differently across studies. Among patients treated with the
intervention regimen, 50.0%–66.7% had neurological sequelae, mostly categorized as mild. The
vast majority of these were among patients diagnosed at stage 2 or 3. Among patients treated with
the comparator regimen, 31.9%–50.0% had neurological sequelae. In one study on the comparator
regimen from India, 17 of 29 (58.6%) cases were categorized as mild. The random effects pooled
proportions of neurological sequelae among survivors were 66.0% (95% CI: 55.0–75.0) and 36.0%
(95% CI: 30.0–43.0) for the intervention and comparator regimens, respectively. Among the 135 HIV-
negative and 13 HIV-positive patients who received the intervention regimen in South Africa, none
relapsed within two years of post-treatment. Other studies did not report on relapse.
Because of the non-comparative nature of the studies, which were all observational, narrative
descriptions reporting pooled proportions were provided rather than estimated measures of effect.
The certainty of evidence was deemed to be very low for all outcomes due to very serious risk of
bias, serious or very serious inconsistency within regimens, and very serious indirectness. Imprecision
could not be assessed due to the lack of comparative data.
The reviewers concluded that pooled estimates need to be interpreted with caution considering
the small number of studies, the potential for confounding by indication, other potential residual
confounding and between-study heterogeneity regarding the assessment of neurological sequelae.
GDG considerations: While the GDG members discussed evidence of benefits of the intervention
regimen and biological plausibility, they noted the very low certainty of the data. The GDG members
decided to choose that the balance of effects ‘does not favour either the intervention or the
comparison’. This decision reflects that both the desirable and undesirable effects varied, as well as
the very low certainty of the evidence overall. Recognizing how quickly the condition of children with
TBM can deteriorate, GDG members were not comfortable favouring one regimen over the other,
based on the very low certainty of the data.
The GDG members also noted that the feasibility of introducing the shorter intensive regimen
is dependent on the setting. Acceptability, affordability and access to the component medicines
(including the child-friendly ethionamide formulation, a 125 mg dispersible tablet) are important
contextual factors as well as any additional implementation considerations such as the necessity and
availability of monitoring tests that are needed with the shorter regimen. GDG members thought that
the short intensive regimen would probably be acceptable as it includes medicines that have been in
use for many years, including first line medicines and ethionamide. The intervention regimen is more
costly than the comparator regimen, but refers to the costs of medicines only; GDG members stated
that other costs (including to patients, families and the health system) were important.
The GDG acknowledged the limited data on alternative regimens to treat TBM and the need
for continued efforts to optimize and better understand treatment options for TBM, including
implementation considerations, such as dosing.
40 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
recommendation, moderate certainty of evidence for test accuracy for Xpert MTB/RIF; low certainty
of evidence for test accuracy for Xpert Ultra (16).
42 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
certainty in the evidence) (9). This recommendation on the shorter all-oral bedaquiline-containing
regimen replaced earlier recommendations on shorter regimens which contained an injectable agent.
In a new IPD meta-analysis used as evidence for the 2020 WHO guidelines, bedaquiline use resulted
in significantly fewer episodes of treatment failure, relapse and death (84). At the time, there was
limited experience in the use of bedaquiline in children aged under 6 years, but experience of
its use in adolescents, patients with EPTB disease and people living with HIV was growing. Earlier
recommendations on the composition of longer regimens indicated that bedaquiline could also be
included in such regimens for patients aged 6–17 years; hence, the all-oral bedaquiline-containing
regimen was also conditionally recommended for use in eligible children and adolescents aged 6
years and above.
Gap: The recommendations that apply to children were based on extrapolation of efficacy data in
adults, in combination with PK and safety data from phase II trials in children aged 6–17 years. However,
a recommendation on the use of bedaquiline in children aged less than 6 years was not possible
in the past, due to a lack of evidence, particularly on PK, safety and tolerability. The medicines that
compose the shorter all-oral bedaquiline-containing regimen have been part of MDR-TB regimens
for many years, in similar combinations, for both adults and children. The associated adverse drug
reactions have been widely described and the drug dosages established. This however did not apply
to bedaquiline at the time of the 2019 GDG on DR-TB, and the use of bedaquiline for young children
in both shorter and longer regimens was therefore identified as a gap to be addressed as part of the
2021 update of the child and adolescent TB guidelines.
Evidence: To answer the PICO question on the use of bedaquiline in children under the age of 6
years, data from two phase II trials (TMC207-C211 and IMPAACT P1108) were reviewed by the GDG.
TMC207-C211 is a phase II, open-label, single-arm study to evaluate the PK, safety, tolerability and
anti-mycobacterial activity of bedaquiline in combination with a background regimen of MDR-TB
medications for the treatment of children and adolescents 0–17 years of age who have bacteriologically
confirmed or clinically diagnosed pulmonary and selected forms of extrapulmonary MDR-TB.32
IMPAACT P1108 is a phase I/II dose finding modified age de-escalation study to evaluate the PK,
safety and tolerability, of bedaquiline in combination with optimized individualized MDR-TB regimens
in HIV-infected and HIV-uninfected children with clinically diagnosed or confirmed pulmonary (intra-
thoracic) and selected forms of extrapulmonary MDR-TB.33
As data reviewed from TMC207-C211 corresponded to children aged 5–18 years and that from
IMPAACT P1108 included children aged 0–6 years, the review of PK and safety data focused mainly
on data from IMPAACT P1108. Although the sample size of the available interim data for review was
small (N= 12), the GDG concluded that in children 0–6 years of age, cardiac safety signals were not
distinct from those reported in adults. Population PK models from both studies suggest that drug
exposures observed in adults can be reached in most children receiving bedaquiline, although some
dose modification may be necessary depending on the age and weight of the child.
In addition, data from a paediatric MDR/RR-TB IPD were analysed descriptively (24 231 records from
all six WHO regions, the majority from India and South Africa). The search was conducted in April
2020. Just under 20 000 of these records were used for a matched analysis of treatment outcomes in
children being treated for DR-TB. The analysis included 40 children aged below 6 years and 68 children
aged 6–12 years who received bedaquiline. In the matched analysis, bedaquiline was significantly
associated with shorter treatment duration and a lower adjusted odds ratio of injectable TB drug use.
There was no statistically significant difference in successful treatment outcomes between children
aged less than 6 years receiving an all-oral bedaquiline-based regimen versus those not receiving
32
Pharmacokinetic study to evaluate anti-mycobacterial activity of TMC207 in combination with background regimen (BR) of multidrug
resistant tuberculosis (MDR-TB) medications for treatment of children/adolescents pulmonary MDR-TB (https://clinicaltrials.gov/ct2/
show/NCT02354014, accessed 21 January 2022).
33
P1108. A Phase I/II, open-label, single arm study to evaluate the pharmacokinetics, safety and tolerability of bedaquiline (BDQ) in
combination with optimized individualized multidrug-resistant tuberculosis (MDR-TB) therapy in HIV-infected and HIV-uninfected
infants, children and adolescents with MDR-TB disease (https://www.impaactnetwork.org/studies/p1108, accessed 21 January 2022).
Subgroup considerations
Children living with HIV: While trial TMC207-C211 has not yet enrolled children living with HIV, the
IMPAACT study P1108 study enrols HIV-infected and HIV-uninfected infants, children and adolescents
with MDR-TB disease (of the nine children aged below 6 years enrolled and with PK data at the time
of the GDG report, one was HIV-infected). In the paediatric IPD, 12 of the 40 children (30%) aged
below 6 years treated with a bedaquiline-containing regimen were HIV positive, as compared to 364
out of 1992 (20%) treated with DR-TB regimens not including bedaquiline.
The composition of treatment regimens for MDR/RR-TB does not usually differ substantially for people
living with HIV; bedaquiline may be used in all children, irrespective of HIV status. Known drug-drug
interactions, such as between bedaquiline and efavirenz, need to be avoided.
Extrapulmonary TB: The shorter, all oral bedaquiline-containing regimen is contraindicated in
children with extrapulmonary forms of TB other than TB lymphadenopathy (see implementation
considerations for the full eligibility criteria). Children with these forms of EPTB should be treated
with longer regimens, composed of medicines from groups A, B and C (9). Data on the penetration
of bedaquiline across the blood-brain barrier are sparse.
Implementation considerations
Eligibility for the shorter, all-oral, bedaquiline containing regimen: The removal of the age
restriction for the use of bedaquiline means that children of all ages with confirmed MDR/RR-TB and
without fluoroquinolone resistance may be offered the shorter, all-oral regimen with bedaquiline,
44 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
if they meet the eligibility criteria. This regimen is composed of bedaquiline (used for 6 months),
in combination with levofloxacin/moxifloxacin, ethionamide, ethambutol, isoniazid (high-dose),
pyrazinamide and clofazimine for four months (with the possibility of extending to 6 months if the
patient remains sputum smear positive at the end of four months); followed by 5 months of treatment
with levofloxacin/moxifloxacin, clofazimine, ethambutol and pyrazinamide. The eligibility criteria are:
• No extensive TB disease
• No severe EPTB (any forms other than TB lymphadenopathy)
• No resistance or suspected ineffectiveness of a medicine in the shorter regimen (except
isoniazid resistance)
• No exposure to previous treatment with second-line medicines in the regimen for more than one
month (unless susceptibility to these medicines is confirmed).
Extensive (or advanced) TB disease refers to the presence of bilateral cavitary disease or extensive
parenchymal damage on CXR in adults. In children aged under 15 years, advanced disease is usually
defined by the presence of cavities or bilateral disease on CXR. Severe EPTB refers to the presence of
miliary TB or TBM (i.e. disseminated disease) (61). In children aged under 15 years, extrapulmonary
forms of disease other than lymphadenopathy (peripheral nodes or isolated mediastinal mass without
compression) are considered as severe (86). The shorter regimen should not be used in the presence
of mutations in both the inhA promoter and katG on first-line LPA (MTBDRplus) in the child as this
suggests that both isoniazid at high dose and thioamides are not effective (87).
Promoting the use of bedaquiline in all-oral regimens for children will allow the construction of
regimens that are more child- and family-friendly and shorter, which may reduce the use of second-line
drugs with potentially more severe adverse events compared to bedaquiline (including, but not limited
to injectables). Implementation of shorter, all-oral regimens may also facilitate the implementation
of DR-TB treatment at peripheral levels of the health care system. NTPs and clinicians are therefore
discouraged from using injectable agents as part of treatment regimens for MDR/RR-TB in children
of all ages.
Regimen building for children not eligible for the shorter all-oral bedaquiline-containing
regimen: Children who do not have bacteriological confirmation of TB and/or resistance patterns but
who have a high likelihood of MDR/RR-TB (based on clinical signs and symptoms of TB, in combination
with a history of contact with a patient with confirmed MDR/RR-TB) are eligible to receive bedaquiline
as part of their treatment regimen. However, they do not meet the eligibility criteria for the shorter
all-oral bedaquiline-containing regimen, as this regimen may only be used in people with TB with at
least confirmation of rifampicin resistance in whom resistance to fluoroquinolones has been ruled out
(84). These children will benefit from an individualized longer treatment regimen, taking into account
the drug susceptibility (and/or mutation) pattern of the most likely source case, if this information is
available. Treatment duration will depend on the extent and severity of disease, as well as the response
to treatment. Shortening the total treatment duration to less than 18 months may be considered in
children without extensive disease (61). Children with a diagnosis of rifampicin resistance only without
further DST (such as a child diagnosed with Xpert MTB/RIF or Xpert Ultra testing on a stool sample
but no further DST on respiratory samples) could be treated with available bedaquiline-containing
regimens at the discretion of the treating clinician.
Guidance on how to construct optimal all-oral treatment regimens in children with MDR/RR-TB who
are not eligible for the shorter, all-oral bedaquiline-containing regimen is provided in the operational
handbook, considering their age, resistance patterns in the child or most likely source case and extent
of TB disease. The construction of these treatment regimens is aligned to the WHO consolidated
guidelines on tuberculosis. Module 4: treatment – drug-resistant tuberculosis treatment, the priority
drug groupings in groups A, B and C as well as the minimum number of effective drugs required (61).
Duration of bedaquiline use: Evidence assessed by a GDG in November 2019 supported the
safe use of bedaquiline beyond 6 months in people with TB who received appropriate schedules
46 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Clinical monitoring: The risk of emergence of bedaquiline resistance should be a key consideration
when the drug is being used. Due to the difficulties in obtaining a suitable specimen from children
aged below 6 years, performing DST may be challenging. However, if there are concerns about
acquired drug resistance, every effort should be made to obtain a suitable sample, such as gastric
aspirate, sputum induction or NPA (stool is not a suitable sample for conducting DST).
In the IMPAACT P1108 study, none of the children aged below 6 years had QT prolongation in any
of the categories of 60 milliseconds and above. Almost all children also received clofazimine. Three
children (of 11; 27%) experienced QT prolongation of 30–60 milliseconds, which was described as mild
and insignificant. In general, the use of bedaquiline in populations with underlying cardiac conduction
abnormalities and concomitant medications that prolong QTc needs to be carefully considered. A
major implementation consideration includes the need for safety monitoring, as well as the need to
build staff capacity (training, capacity building) to ensure that appropriate safety monitoring is put in
place. Additional costs associated with wider implementation of bedaquiline (in addition to the higher
cost of the drug), include availability of ECG machines, appropriate monitoring and trained staff to
perform these activities. However, the GDG felt that savings related to potential shorter treatment
duration as well as the avoidance of additional resource requirements related to injectable use (such
as for monitoring with audiometry and implications for child development), outweigh the costs of
bedaquiline introduction and implementation.
Adaptation of the recommendation to the local context will require staff capacity building and training,
followed by supportive supervision and mentoring. Implementing all-oral treatment regimens will
facilitate decentralization of MDR-TB treatment services and may therefore improve access to patient-
centred care.
48 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
past GDGs convened by the WHO had already recommended the use of delamanid. Additionally,
based on the laboratory and cardiac data provided, no safety signals distinct from those reported
in adults were observed in children aged 3–5 years. Based on this, the WHO recommended that
delamanid may be included in the treatment of MDR/RR-TB for children aged 3 years or more on
longer regimens (conditional recommendation, moderate certainty in the estimates of effect); and
in the priority grouping of medicines for use in longer regimens it was classified as a group C drug
(83). The 2019 GDG nonetheless had concerns about the feasibility of administering the correct
dose to children aged 3–5 years, given that the only tablet available at the time was the one used
for adults (i.e. 50 mg), which presents challenges in manipulating its contents without compromising
its effectiveness. Subsequent reviews of the WHO guidelines in 2020 did not prompt any change in
the 2019 recommendation, which was the one in place at the time of the GDG meeting on child and
adolescent TB during May-June 2021.
Gap: The recommendations that apply to children were based on extrapolation of efficacy data
in adults, in combination with PK and safety data from phase II trials for children aged 3–17 years.
However, a recommendation on the use of delamanid in children aged less than 3 years has not been
possible in the past, due to a lack of evidence, particularly on PK, safety and tolerability. This has made
it challenging for clinicians to design all-oral regimens for children under the age of 3 years, especially
for children with (or a source case with) fluoroquinolone resistance, where choices are limited among
group A and B drugs. The use of delamanid in children below 3 years of age was therefore identified
as a gap to be addressed as part of the 2021 update of the child and adolescent TB guidelines.
Evidence: To answer the PICO question on the use of delamanid in children under the age of 3
years, data were reviewed by the GDG from a phase I, open-label, age de-escalation trial designed
to assess the PK, safety and tolerability of delamanid administered twice daily for 10 days in children
with MDR/RR-TB on treatment with an optimized background regimen (protocol 242–12–232)34 and
from the corresponding open-label extension study (protocol 242–12–233).35 Data from cohorts 1
(age 12–17 years), 2 (age 6–11 years), 3 (age 3–5 years) and 4 (age 0–2 years) for both protocols
were reviewed. Exposures in the 0–2 year age group were lower than those of children aged 3 years
and older, necessitating a modelling/simulation approach to dosing. No cardiac safety signals distinct
from those reported in adults were observed in children 0–2 years of age. However, these findings
should be considered knowing that children had lower drug exposures compared to adults. However,
pharmacodynamic simulations suggested that clinically meaningful changes in QT (i.e. prolongation)
would be unlikely in children under 3 years of age, even if higher doses were used to reach drug
exposures comparable to those achieved in adults.
Central nervous system effects were included in the delamanid label for both adults and children
(paraesthesia, tremors, anxiety, depression and insomnia) as important potential safety concerns for
the drug. In March 2021, the study sponsor released a statement of intent to modify the labelling to
include hallucinations as an adverse reaction. This new safety signal has been more prevalent among
children (versus adults) with 15 reports in 14 children 2–16 years of age in India, Philippines, South
Africa, Tajikistan and Ukraine. Children experiencing this safety signal included some with extensively
resistant forms of TB (MDR/XDR-TB) treated with delamanid under programmatic conditions (12
reports) as well as children enrolled in a clinical trial studying delamanid for TB prevention (three
reports). Seven of the 15 reports were for children also receiving cycloserine (under programmatic
conditions). The GDG noted the importance of side-effects involving the central nervous system in
young children, considering their dynamic brain development.
In addition to data from the trials, data from a paediatric DR-TB IPD were analysed descriptively (24
231 records from all six WHO regions, the majority from India and South Africa). The search was
34
Pharmacokinetic and safety trial to determine the appropriate dose for pediatric patients with multidrug resistant tuberculosis (https://
clinicaltrials.gov/ct2/show/NCT01856634, accessed 21 January 2022).
35
A 6-month safety, efficacy, and pharmacokinetic (PK) trial of delamanid in pediatric participants with multidrug resistant tuberculosis
(MDR-TB) (https://clinicaltrials.gov/ct2/show/NCT01859923, accessed 21 January 2022).
Subgroup considerations
Extrapulmonary TB: The use of delamanid in children with extrapulmonary MDR/RR-TB may be
considered (as part of longer regimens used for children with extrapulmonary MDR/RR-TB) by
extrapolating data from those with PTB; the delamanid trials, however, have studied PK and safety
among children with pulmonary MDR/RR-TB.
Children living with HIV: Children living with HIV were not enrolled in trials 242–12–232 and 233.
Although evaluated in healthy adult volunteers, reports from antiretroviral drug-drug interaction
studies suggest that the CYP3A4 inhibitor lopinavir/ritonavir increases delamanid total body
exposure up to 25% [GMR: 1.22 (90% CI 1.06,1.40)] (90). This increase is not clinically relevant and
does not necessitate any dose adjustments. No change in delamanid exposure was observed with
co-administration of either tenofovir [GMR: 0.96 (90% CI 0.84,1.10)], a CYP1A2 inhibitor, or efavirenz
[GMR: 0.94 (90% CI 0.72,1.23)], a weak CYP3A4 inducer. Delamanid does not affect plasma exposure
of the antiretroviral drugs tenofovir, lopinavir/ritonavir or efavirenz (94). No drug interaction studies
of delamanid together with integrase inhibitors have been performed, but based on knowledge
of metabolic pathways, the risk of metabolic drug interaction potential is expected to be low (95).
Therefore, based on available evidence, delamanid can be given to CLHIV with MDR/RR-TB on ART
regimen without dose adjustments.
Implementation considerations
Delamanid has been in use for adults and adolescents since 2014 and for children since 2016 (from
6 years of age) and 2019 (from 3 years) and therefore the implementation considerations related
to its use in children below 3 years of age are an extension of those currently in place. The main
implementation considerations specifically applicable to this age group are dosing based on the
availability of the 25 mg dispersible formulation and the neuropsychiatric side effects.
50 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Delamanid formulations: In the trial, delamanid was dosed with the dispersible 25 mg tablet
tested in children aged 3–5 years. Bioavailability of delamanid may be altered when the 50 mg
adult tablet is split, crushed or dissolved. There are also concerns that the adult tablet may shatter if
attempts are made to split it, and its contents are exceedingly bitter and unpalatable. The tablets are
susceptible to oxidation and heat; thus, retaining pill fragments for use at any time other than the
time of administration is likely to result in the delivery of lower-than-expected active compound and
unspecified oxidation by-products. The child-friendly formulation of delamanid (25 mg dispersible
tablet, unscored) has been included in the 8th WHO Essential Medicine List for Children (EMLc),
released in October 2021 (88), was approved by the European Medicines Agency in September 2021
and has been available through the Stop TB Partnership GDF since October 2021. Children in the
0–2-year cohort in study 242–12–233 were administered a 5 mg paediatric dispersible formulation
that is not expected to be commercialized. There has been no direct bioequivalence comparison
for the 5 mg paediatric formulation and the 50 mg adult tablet. In a crossover bioequivalence study,
neither Cmax [90%CI GMR 0.701,0.809] nor AUC [90%CI GMR 0.775,0.909] satisfied the criteria for
bioequivalence as specified by regulatory agencies. As such, the 5 mg paediatric and the 50 mg adult
formulations are not interchangeable (96).
Dosing guidance for the use of delamanid in children below 3 years of age is provided in the
operational handbook, based on an expert consultation on dosing that was conducted after the
GDG meeting. The guidance takes into account the availability of the 25 mg dispersible delamanid
formulation.
Administration of delamanid: Bioavailability of delamanid was optimized in the trials by administering
delamanid with a high-fat meal; and therefore administration of delamanid with food is an important
aspect to consider for practical implementation. In neonates, there are higher feeding frequencies,
which aligns well with the goal of administering with higher-fat content.
Regimen building of longer regimens: Guidance on how to construct optimal treatment regimens
for children with MDR/RR-TB (with drugs based on WHO recommended drug classification as well
as optimal treatment duration) who are not eligible for shorter, all-oral regimens is provided in the
operational handbook.
Duration of treatment: Shortening the total treatment duration to less than 18 months may be
considered for children without extensive disease (61). Extensive (or advanced) TB disease refers to the
presence of bilateral cavitary disease or extensive parenchymal damage on CXR. Severe EPTB refers
to the presence of miliary TB or TBM in adolescents and adults over 15 years of age. In children aged
below 15 years, extrapulmonary forms of disease other than lymphadenopathy (peripheral nodes or
isolated mediastinal mass without compression) are considered as severe (adapted from (86)).
Concurrent use of delamanid and bedaquiline, and use of delamanid beyond six months: With
regards to concurrent delamanid and bedaquiline use, evidence assessed by a GDG in November 2019
included new data on concurrent bedaquiline and delamanid use. The new evidence was insufficient
to allow the GDG to make a statement about the effectiveness of concurrent use of both medicines.
However, the group concluded that the safety data assessed in 2019 suggest no additional safety
concerns with regard to the concurrent use of bedaquiline and delamanid. Therefore, bedaquiline and
delamanid may be used in people with MDR/RR-TB who have limited options for other treatment,
such as those with few effective drugs that can be included in their regimen, for example due to an
extensive drug-resistance profile or intolerance to other second-line TB medications. Appropriate
schedules of safety monitoring (at baseline and throughout treatment) should be in place for these
patients, including ECG and electrolyte monitoring, and clinicians should be aware of other medicines
in the regimen that can either prolong the QT interval or cause other potential adverse events. The
available evidence of delamanid use is currently limited to the on-label 6-month duration alongside
other medicines in a longer regimen; prolongation beyond 6 months can be considered on a case-
by-case basis (9).
52 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
5.4. Consolidated recommendations on TB treatment
for children and adolescents
Table 6: WHO recommendations on TB treatment relevant to children and adolescents
36
This recommendation is applicable to adolescents from 15 years of age
54 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Ethambutol may be included in the treatment of MDR/RR-TB patients on longer regimens.
(Conditional recommendation, very low certainty in the estimates of effect)
Pyrazinamide may be included in the treatment of MDR/RR-TB patients on longer regimens.
(Conditional recommendation, very low certainty in the estimates of effect)
Imipenem–cilastatin or meropenem may be included in the treatment of MDR/RR-TB patients
on longer regimens.37
(Conditional recommendation, very low certainty in the estimates of effect)
Amikacin may be included in the treatment of MDR/RR-TB patients aged 18 years or more
on longer regimens when susceptibility has been demonstrated and adequate measures to
monitor for adverse reactions can be ensured. If amikacin is not available, streptomycin may
replace amikacin under the same conditions.
(Conditional recommendation, very low certainty in the estimates of effect)
Ethionamide or prothionamide may be included in the treatment of MDR/RR-TB patients on
longer regimens only if bedaquiline, linezolid, clofazimine or delamanid are not used, or if
better options to compose a regimen are not possible.
(Conditional recommendation against use, very low certainty in the estimates of effect)
P-aminosalicylic acid may be included in the treatment of MDR/RR-TB patients on longer
regimens only if bedaquiline, linezolid, clofazimine or delamanid are not used, or if better
options to compose a regimen are not possible.
(Conditional recommendation against use, very low certainty in the estimates of effect)
Clavulanic acid should not be included in the treatment of MDR/RR-TB patients on
longer regimens.
(Strong recommendation against use, low certainty in the estimates of effect)
The bedaquiline, pretomanid and linezolid (BPaL) regimen for multidrug-resistant
tuberculosis with additional fluoroquinolone resistance
A treatment regimen lasting 6–9 months, composed of bedaquiline, pretomanid and linezolid
(BPaL), may be used under operational research conditions in multidrug-resistant tuberculosis
(MDR-TB) patients with TB that is resistant to fluoroquinolones, who have either had no
previous exposure to bedaquiline and linezolid or have been exposed for no more than 2
weeks.
(Conditional recommendation, very low certainty in the estimates of effect)
Note: this recommendation concerns patients aged 14 years and above.
37
Imipenem-cilastatin and meropenem are administered with clavulanic acid, which is available only in formulations combined with
amoxicillin. Amoxicillin-clavulanic acid is not counted as an additional effective TB agent, and should not be used without imipenem-
cilastatin or meropenem.
56 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
6. Models of TB care for case
detection and provision of TPT
in children and adolescents
This chapter contains two new recommendations and other valid WHO recommendations that
apply to patient support and models of care (section 6.2). The two new recommendations, on the
implementation of decentralized models of care and integrated family-centred models of care to
improve both case detection and the provision of TPT, are described in full detail as this information
is being published for the first time. Other consolidated recommendations in this chapter are related
to patient support, health education and counselling, the provision of treatment support or video
supported treatment and decentralized models of care for MDR/RR-TB services.
The recommendations in this chapter have been consolidated from current WHO guidelines on TB
treatment and the context in which it should be provided, namely the Guidelines for treatment of
drug-susceptible tuberculosis and patient care, 2017 update (97) and the WHO consolidated guidelines
on tuberculosis. Module 4: treatment – drug-resistant tuberculosis treatment, 2020 update (9). For
more information on each recommendation including the remarks, source of evidence, justification,
subgroup-, implementation- and monitoring and evaluation considerations, the source guidelines
or WHO TB KSP should be consulted.
Capacity for paediatric TB is often highly centralized at secondary/tertiary level, where children
may present as seriously ill, after delays in accessing care. At higher levels of care, services are often
managed in a vertical, non-integrated way (10, 98). Health care workers at the PHC level may have
limited capacity and confidence in managing paediatric TB, although this is where most children with
TB or at risk of TB seek care (10). In addition, TB screening is often not systematically part of clinical
algorithms for child health, such as integrated management of childhood illness (IMCI) and integrated
community case management (iCCM). Private sector providers play an increasing role as first point
of care in many countries (99). There are many missed opportunities for contact tracing, as well as
TB prevention, detection and care due to weak integration of child and adolescent TB services with
other programmes and services.
Decentralization and provision of family-centred, integrated care are highlighted as one of 10 key
actions in the 2018 Roadmap (10). The Roadmap highlights that consistently and systematically
addressing gaps and bottlenecks along children’s and adolescents’ pathway through TB exposure,
TB infection and TB disease can lead to reduced transmission of TB, expansion of prevention of TB
infection and earlier TB diagnosis with better outcomes. Achieving this continuum of care requires
collaboration across service areas, practice disciplines and sectors, community engagement, as well
as decentralization and integration of service delivery at the PHC level (10).
The Roadmap suggests actions to integrate child and adolescent TB into family- and community-
centred care, including: (i) strengthening country-level collaboration and coordination across all
health-related programmes engaged in woman, adolescent and child health – especially reproductive
health, maternal, neonatal, child and adolescent health (MNCAH), nutrition, HIV, primary and
community health – with clearly defined roles, responsibilities and joint accountability; (ii) decentralizing
6. Models of TB care for case detection and provision of TPT in children and adolescents 57
and integrating successful models of care for TB screening, prevention and diagnosis with other
existing service delivery platforms for maternal and child health (such as antenatal care (ANC), iCCM,
IMCI) as well as other related services (such as HIV, nutrition, immunization); (iii) ensuring that children
and adolescents with other common comorbidities (such as meningitis, malnutrition, pneumonia,
chronic lung disease and HIV infection) are routinely evaluated for TB; (iv) ensuring community health
strategies integrate child and adolescent TB education, screening, prevention and case finding into
the training and service delivery activities; and (v) increasing awareness of and demand for child and
adolescent TB services in communities and among health workers (10).
This set of PICO questions examined the impact of: (i) decentralization and (ii) family-centred, integrated
approaches of child and adolescent TB services on case detection in children and adolescents who
present with signs and symptoms of TB. They also examine the impact of these approaches on
coverage of TPT among children and adolescents.
58 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
6.1.1. Justification and evidence
PICO questions:
a. In children and adolescents with signs and symptoms of TB, should decentralization of child and
adolescent TB services versus centralized child and adolescent TB services (at referral or tertiary
hospital level) be used?
b. In children and adolescents exposed to TB, should decentralization of child and adolescent TB
prevention and care services versus centralized prevention and care services (at referral or tertiary
hospital level) be used to increase coverage of TPT in eligible children and adolescents?
c. In children and adolescents with signs and symptoms of TB, should family-centred, integrated
services versus standard, non-family-centred, non-integrated services be used?
d. In children and adolescents exposed to TB, should family-centred, integrated services versus
standard, non-family-centred, non-integrated services be used to increase TPT coverage in eligible
children and adolescents?
For definitions of decentralization and family-centred, integrated services, please refer to the section
with definitions from page xi.
Evidence: A systematic review of studies assessing the impact of decentralized, integrated or family-
centred care models on TB diagnosis, treatment or prevention outcomes in children and adolescents
with TB between 0 and 19 years old, comprising both children (0–9 years old) and adolescents (10–
19 years old), was conducted to answer this group of PICO questions. The PubMed, Embase, Web
of Science, Global Index Medicus, Global Health and Cochrane Central databases were searched in
February 2021, as well as the references of 17 related reviews. 3265 abstracts from databases and
129 additional references from related reviews were identified and assessed. 516 full-text articles were
assessed for eligibility, from which 25 comparative studies (7 randomized, 18 observational) were
identified; one unpublished observational study was added making a total of 26 studies. Four studies
(one randomized, three observational) were excluded after review because the care model described
was community-based treatment support, for which a WHO recommendation already exists (100). Of
the remaining included studies, 16 had elements of decentralization, five had elements of integration,
and three had elements of family-centred care; four studies had elements of more than one care
model of interest, but were only included based on their main model, such as either decentralization
or family-centred, integrated care. Most studies focused on the 0–14-year age group.
Studies where the primary intervention was decentralization mostly assessed diagnosis or case
notification outcomes (n=16) (48, 101–115), with fewer assessing TPT outcomes (n=3) (106, 116, 117).
In general, interventions that included both strengthening of diagnostic capacity in primary care
settings as well as strengthening linkages between communities and facilities consistently showed an
increase in case notifications and TPT initiations, while interventions that involved only community-
based activities did not.
Two studies of service integration were identified (118, 119), which showed limited impact on case
notifications of screening in IMCI clinics or co-location of TB and ART services. The two studies of
family-centred care that were identified (120, 121) showed that provision of socioeconomic support
packages to families affected by TB was associated with increased TPT initiation and completion.
The reviewers noted that, while substantial wider literature on integration and family-centred care
is available, evidence for the specific impact on child and adolescent TB outcomes is limited. Some
overlap was noted between integration of TB services into non-specialized settings, such as general
outpatient or primary care services, or decentralization. For the evidence review this was a slightly
artificial separation, while in practice decentralization and integration into PHC may happen together.
6. Models of TB care for case detection and provision of TPT in children and adolescents 59
GDG considerations: Regarding the evidence reviewed on the impact of decentralization on TB case
detection, the GDG observed that two trials (109, 111) and one observational study of home-based
screening (without facility-based strengthening) (114) had fewer diagnoses or notifications among
children aged below 15 years in the intervention group compared to the control group, but that
none of these differences were statistically significant. The GDG discussed that while there may be a
reduction in case notifications at higher levels of care, TB detection may improve if children are seen
by a competent clinician at the first point of access (such as at PHC level). The evidence overall was
recognized as uncertain. The benefit of increased case finding and increased number of children with
TB who are initiated on TB treatment was considered to outweigh the concern for overtreatment.
Therefore, undesirable effects for case detection were considered trivial. The GDG discussed potential
risks of provision and management of TPT at the peripheral level, including undetected drug-related
adverse events such as hepatotoxicity and insufficient capacity to manage these. In addition, there
may be a risk of TB disease being treated with a course of TPT rather than with a complete treatment
regimen. All these undesirable events can potentially happen but were considered rare and not of
major concern. Therefore, undesirable effects for TPT provision were considered trivial as well. Overall,
the GDG agreed that the balance of desirable and undesirable effects probably favours decentralized
TB services for case detection and provision of TPT to children and adolescents. The panel noted that
differences in setting and availability of adequate resources are important considerations.
The GDG discussed that family-centred, integrated care includes interventions at the household level
to identify members of the household requiring evaluation for TB disease, TPT, treatment support,
etc. Some overlap between integration of TB services into non-specialized settings such as general
outpatient or primary care services, and decentralization was noted. For the evidence review, this
was a slightly artificial separation, while in practice decentralization and integration into PHC may
happen together. Overall, despite a lack of evidence on undesirable effects and low quality of the
data, the panel agreed that there is evidence of positive effects of family-centred integrated care. It
was suggested that family-centred, integrated care could be an addition to the standard of care as
well as to specialized services which do not have an integration component. Family-centred care in
the sense of family involvement was highlighted as a core principle of child health care.
The GDG discussed that setting specific factors related to TB burden or the organization of health
services may impact feasibility, acceptability and equity considerations. They also discussed that the
initial health system costs to establish decentralized and family-centred, integrated services may be
relatively high (such as infrastructure, human resources, training, equipment, community engagement),
but that costs are likely to decrease over time, assuming that people with TB are effectively managed
and TPT is provided at the peripheral level, leading to a reduction in TB incidence. Decentralized
and family-centred, integrated services may result in important savings for affected families. Equity
was considered an important cross-cutting issue impacting cost as well. The GDG highlighted that
TPT implementation can be very challenging with high levels of loss-to-follow-up in programmes
implemented at higher levels of the health system, considering that children who are eligible for
TPT are not sick. The panel agreed that decentralization and integration of services can potentially
increase equity and enhance the success of the programme and judged that cost-effectiveness
probably favours decentralized and family-centred, integrated approaches to both case finding and
provision of TPT.
While the GDG stressed the importance of taking into consideration the potential impact of stigma
when decentralizing TB services for children and adolescents to lower levels, the panel judged
that decentralized approaches are probably acceptable to key stakeholders. Overall decentralized
and family-centred, integrated approaches were judged feasible to implement, although feasibility
may vary depending on infrastructure, available funding and the structure of the NTP, among
others. However, adequate investment is critical to enable the acceptability, equity and feasibility of
decentralized approaches.
60 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
6.1.2. Subgroup considerations
Adolescents have a disease presentation that is similar to adults, and therefore may need different
interventions than young children. Additional subgroup considerations for adolescents are included
in the operational handbook, taking into account their specific health-seeking behaviour and the
need for adolescent-friendly services.
TB contacts: Provision of TPT has focused mainly on children under 5 years of age for many years.
In 2018, target groups for the provision of TPT were expanded to include contacts of all ages (122).
Available data from the global TB database (78) show that coverage of TPT in household contacts is
poor, especially in those over 5 years of age.
In children with common illnesses with overlapping signs and symptoms of TB, approaches to integrate
TB services in their care can improve case detection and provision of TPT.
These subgroups include:
• Children with SAM
• Children with severe pneumonia
• Children living with HIV
• Children with other chronic diseases.
6. Models of TB care for case detection and provision of TPT in children and adolescents 61
economic support should be provided to enable them to complete the diagnostic process and the
full course of prescribed treatment.” Multiple descriptions exist that include components of support
and education based on individual needs, building a patient-provider partnership and participatory
decision-making. Family-centred care also includes interventions at the level of the household to
identify members of the household requiring evaluation for TB disease, TPT, treatment support,
etc. As the concept of family-centred, integrated care may be setting specific, one of the first steps
in implementation includes clarifying which definition applies to the setting in which it is to be
implemented. Similarly, the implementation strategy varies by setting and needs to be country- or
region-specific, informed by social, cultural and societal values.
The package of TB services to be provided needs to be defined and developed by the NTP, in close
coordination with other relevant programmes, such as through an existing child and adolescent TB
technical working group. This package needs to be based on identifying and addressing capacity
needs for national programmes interested in the uptake of proposed interventions, and ideally based
on family and community perceptions on the ideal family-centred model of care. It could include
community-based models for active contact investigation, identifying children with TB signs and
symptoms or exposure as part of routine growth monitoring services, or an integrated model for
IMCI integration, starting with the sick child and identifying signs and symptoms pointing to a high
likelihood of TB.
Integration can start within the family, by equipping the family with the knowledge to recognize signs
and symptoms to understand the importance of a history of contact, to know when to seek help at
the health care facility and how to minimize stigma related to TB. High yield entry points provide a
good starting point within the health system. For example, child and adolescent TB services can be
integrated in malnutrition clinics, ANC, the expanded programme on immunization, inpatient sites,
adult TB and chest clinics, HIV and general paediatric clinics. Ideally TB care should be integrated into
general health services, rather than be limited to enhanced coordination between two programmes.
However, defining an optimal patient flow between services and creating strong linkages between child
health entry points and TB clinics remains essential, especially in facilities where services are physically
separated. This is critical to enhance the quality of services, including the follow-up of persons with
TB during the diagnostic evaluation, to also ensure accuracy of recording and reporting. In the early
phase, pilot programmes could be considered, which should be evaluated and adjusted as needed
and then scaled up.
Factors to consider in designing an integrated approach to child and adolescent TB care include
existing infrastructure (such as baseline health infrastructure, needs for expansion or upgrading), the
applicable regulatory framework, financing, choosing between an operational research setting or
programmatic implementation, human resource issues (including staffing requirements and human
resources development such as capacity building/training and consultation skills), monitoring and
evaluation, conducting qualitative research into community needs, perceptions (including views on
stigma) and suggestions.
Differentiated service delivery (DSD): DSD is a person-centred approach developed in the HIV
programme that simplifies and adapts HIV services across the cascade of care in ways that both serve
the needs of people living with and vulnerable to HIV and optimize the available resources in health
systems. The principles of DSD can be applied to prevention, testing, linkage to care, ART initiation
and follow-up, and integration of HIV care and coinfections and comorbidities (123). This approach
embraces the idea that when families are given the choice to interact with the health system, it could
provide a possible mechanism for integration of child and adolescent TB services within primary
health or other programmes. Examples of implementing DSD for children and adolescents with or at
risk of TB are provided in the operational handbook.
62 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
6.1.4. Monitoring and evaluation
Moving to decentralized, family-centred, integrated services requires careful planning and regular
monitoring of implementation against the plan. The capacity needs of NTPs for implementing the
proposed interventions need to be identified and addressed.
Enhanced data collection around child and adolescent TB potentially takes a substantial amount of
additional time and detailed data collection may only be feasible in specific operational research
settings. Programmes generally have registers in place for contact investigation, treatment registration
and outcomes, as well as TPT registers. The use of these (preferably electronic) tools is important
as programmes move to a more decentralized and family-centred, integrated approach, to ensure
comprehensive management and treatment. The use of these tools needs to be evaluated and
enhanced, including through operational research.
It will be important to monitor the number of children diagnosed at different levels of the health
system, including the proportion of children that have bacteriological confirmation, the proportion
who were clinically diagnosed as well as the number of children initiated on and completing TPT.
Disaggregation of data by sex will be important to evaluate the impact on gender equity. Evaluating
the quality of services (covering the quality of all steps in the patient pathway, from screening, to
diagnosis and treatment) as well as client satisfaction are important components as well.
38
Treatment adherence interventions include social support such as patient education and counselling, material support (e.g. food, financial
incentive, and transport fee); psychological support; tracers such as home visit or digital health communication (e.g. SMS, telephone
call); medication monitor; and staff education. The interventions should be selected on the basis of the assessment of individual patient’s
needs, provider’s resources and conditions for implementation.
39
Suitable treatment administration options include various forms of treatment support, such as video-supported treatment and regular
community of home-based treatment support.
6. Models of TB care for case detection and provision of TPT in children and adolescents 63
One or more of the following treatment adherence interventions (complementary and not
mutually exclusive) may be offered to patients on TB treatment or to health care providers:
a. tracers40 or digital medication monitor41 (Conditional recommendation, very low certainty in
the evidence)
b. material support to the patient42 (Conditional recommendation, moderate certainty in
the evidence);
c. psychological support to the patient43 (Conditional recommendation, low certainty in
the evidence)
d. staff education44 (Conditional recommendation, low certainty in the evidence).
The following treatment administration options may be offered to patients on TB treatment:
a. Community- or home-based treatment support is recommended over health facility-based
treatment support or unsupported treatment (conditional recommendation, moderate
certainty in the evidence);
b. Treatment support administered by trained lay providers or health care workers is
recommended over treatment support administered by family members or unsupported
treatment (conditional recommendation, very low certainty in the evidence)
c. Video supported treatment (VST) can replace treatment support when the video
communication technology is available and can be appropriately organized and operated
by health-care providers and patients (conditional recommendation, very low certainty in
the evidence).
Patients with multidrug-resistant TB (MDR-TB) should be treated using mainly ambulatory care
rather than models of care based principally on hospitalization.
(Conditional recommendation, very low certainty evidence)
A decentralized model of care is recommended over a centralized model for patients on
MDR-TB treatment.
(Conditional recommendation, very low certainty in the evidence)
40
Tracers refer to communication with the patient including via SMS, telephone (voice) calls, or home visit.
41
A digital medication monitor is a device that can measure the time between openings of the pill box. The medication monitor may have
audio reminders or send an SMS to remind patient to take medications, along with recording when the pill box is opened.
42
Material support can be food or financial support such as: meals, food baskets, food supplements, food vouchers, transport subsidies,
living allowance, housing incentives, or financial bonus. This support addresses indirect costs incurred by patients or their attendants
in order to access health services, and possibly tries to mitigate consequences of income loss related to the disease.
43
Psychological support can be counselling sessions or peer-group support.
44
Staff education can be adherence education, chart or visual reminder, educational tools and desktop aids for decision-making
and reminder.
64 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
7. Special situations
This chapter includes valid WHO recommendations that apply to children and adolescents in
special situations such as for the management of TB in the context of HIV infection or malnutrition
and optimal feeding of infants of mothers infected with TB. The recommendations have been
consolidated from several current WHO guidelines on TB/HIV coinfection and nutrition, namely
the WHO policy on collaborative TB/HIV activities: guidelines for national programmes and other
stakeholders (124), the Consolidated guidelines on HIV prevention, testing, treatment, service delivery
and monitoring: recommendations for a public health approach, July 2021 (125), the Guideline: updates
on the management of severe acute malnutrition in infants and children, 2013 (60), and the Guideline:
Nutritional care and support for patients with tuberculosis, 2013 (126). For more information on each
recommendation including the remarks, source of evidence, justification, subgroup-, implementation-
and monitoring and evaluation considerations, the source guidelines or WHO TB KSP should be
consulted.
WHO policy on collaborative TB/HIV activities: guidelines for national programmes and
other stakeholders, 2012 (124)
Routine HIV testing should be offered to all patients with presumptive and diagnosed TB.
(Strong recommendation, low certainty of evidence)
Consolidated guidelines on HIV prevention, testing, treatment, service delivery and
monitoring: recommendations for a public health approach, July 2021 (125)
Co-trimoxazole prophylaxis for infants, children and adolescents living with HIV
Co-trimoxazole prophylaxis is recommended for infants, children and adolescents with HIV,
regardless of clinical and immune conditions. Priority should be given to all children younger
than 5 years old regardless of CD4 cell count or clinical stage and children with severe or
advanced HIV clinical disease (WHO clinical stage 3 or 4) and/or those with CD4 cell count
≤350 cells/mm3.
(Strong recommendation, high certainty evidence)
In settings where malaria and/or severe bacterial infections are highly prevalent, co-trimoxazole
prophylaxis should be continued until adulthood, irrespective of whether ART is provided.
(Conditional recommendation, moderate certainty evidence)
In settings with low prevalence for both malaria and bacterial infections, cotrimoxazole
prophylaxis may be discontinued for children 5 years of age and older who are clinically stable
and/or virally suppressed on ART for at least six months and with a CD4 count >350 cells/mm3.
(Strong recommendation, very low certainty evidence).
7. Special situations 65
Co-trimoxazole prophylaxis is recommended for HIV-exposed infants from 4 to 6 weeks of age
and should be continued until HIV infection has been excluded by an age-appropriate HIV test
to establish final diagnosis after complete cessation of breastfeeding.
(Strong recommendation, very low certainty evidence)
Routine co-trimoxazole prophylaxis should be given to all people living with HIV with TB disease
regardless of CD4 cell count.
(Strong recommendation, high certainty evidence)
General recommendations on eligibility for ART
ART should be initiated for all people living with HIV regardless of WHO clinical stage and at
any CD4 cell count.
• Pregnant and breastfeeding women (strong recommendation, moderate certainty evidence)
• Adolescents (conditional recommendation, low certainty evidence)
• Children living with HIV, one year old to less than 10 years old (conditional recommendation,
low certainty evidence)
• Infants diagnosed in the first year of life (strong recommendation, moderate certainty evidence)
Rapid ART initiation should be offered to all people living with HIV following a confirmed HIV
diagnosis and clinical assessment.
(Strong recommendation, high-certainty evidence for adults and adolescents; low certainty
evidence for children).
Rapid initiation is defined as within seven days from the day of HIV diagnosis; people with
advanced HIV disease should be given priority for assessment and initiation.
ART initiation should be offered on the same day to people who are ready to start.
(Strong recommendation, high certainty evidence for adults and adolescents; low certainty
evidence for children)
Timing of ART for children and adolescents with TB
ART should be started as soon as possible within 2 weeks of initiating TB treatment, regardless
of CD4 count, among adolescents and children living with HIV (except when signs and
symptoms of meningitis are present). (Adolescents: strong recommendation, low- to moderate-
certainty evidence; Children and infants: strong recommendation, very low certainty evidence)
ART should be delayed at least 4 weeks (and initiated within 8 weeks) after treatment for TB
meningitis is initiated.
First-line ART regimens
Dolutegravir (DTG) in combination with a nucleoside reverse-transcriptase inhibitor (NRTI)
backbone is recommended as the preferred first-line regimen for people living with HIV
initiating ART
• Adolescents (strong recommendation, moderate certainty evidence)
• Infants and children with approved DTG dosing (conditional recommendation, low
certainty evidence)
A raltegravir (RAL)-based regimen may be recommended as the preferred first-line regimen for
neonates
(Conditional recommendation, very low certainty evidence)
66 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Second-line ART regimens
DTG in combination with an optimized NRTI backbone may be recommended as a preferred
second-line regimen for people living with HIV for whom non-DTG-based regimens are failing.
• Adolescents (conditional recommendation, moderate certainty evidence)
• Children with approved DTG dosing (conditional recommendation, low certainty evidence)
Boosted protease inhibitors in combination with an optimized NRTI backbone are
recommended as a preferred second-line regimen for people living with HIV for whom DTG-
based regimens are failing.
(Strong recommendation, moderate certainty evidence)
Guidelines: updates on the management of severe acute malnutrition in infants and
children, 2013 (60)
Infants with severe acute malnutrition who are admitted for inpatient care should be given
parenteral antibiotics to treat possible sepsis and appropriate treatment for other medical
complications such as tuberculosis, HIV, surgical conditions or disability.
(Strong recommendation, very low certainty evidence)
Guideline: nutritional care and support for patients with tuberculosis, 2013 (126)
Management of severe acute malnutrition
School-age children and adolescents (5 to 19 years), and adults, including pregnant and
lactating women, with TB disease and severe acute malnutrition (very low BMI-for-age)
should be treated in accordance with the WHO recommendations for management of severe
acute malnutrition.
(Strong recommendation, very low certainty evidence)
Children who are less than 5 years of age with TB disease and severe acute malnutrition (mid-
upper arm circumference more than 115 mm or weight-for-height/length more than three
z-scores below the WHO child growth standards median, or with any degree of bilateral
pitting oedema) should be treated in accordance with the WHO recommendations for the
management of severe acute malnutrition in children who are less than 5 years of age.
(Strong recommendation, very low certainty evidence)
Management of moderate undernutrition
School-age children and adolescents (5 to 19 years) and adults, including lactating women,
with TB disease and moderate undernutrition, who fail to regain normal body mass index after
2 months’ TB treatment, as well as those who are losing weight during TB treatment, should
be evaluated for adherence and comorbid conditions. They should also receive nutrition
assessment and counselling and if indicated be provided with locally available nutrient-rich or
fortified supplementary foods as necessary to restore normal nutritional status.
(Conditional recommendation, low certainty evidence)
Children who are less than 5 years of age with TB disease and moderate undernutrition should
be managed as any other children with moderate undernutrition. This includes provision of
locally available nutrient-rich or fortified supplementary foods, in order to restore appropriate
weight-for-height.
(Strong recommendation, very low certainty evidence)
7. Special situations 67
Patients with multidrug-resistant TB and moderate undernutrition should be provided with
locally available nutrient-rich or fortified supplementary foods, as necessary, to restore normal
nutritional status.
(Strong recommendation, very low certainty evidence)
A daily multiple micronutrient supplement at 1× recommended nutrient intake should be
provided in situations where fortified or supplementary foods should have been provided in
accordance with standard management of moderate undernutrition,45 but are unavailable.
(Conditional recommendation, very low certainty evidence)
Nutrition screening as part of contact investigation
In settings where contact tracing is implemented, household contacts of people with TB
disease should have a nutrition screening and assessment as part of contact investigation. If
malnutrition is identified, it should be managed according to WHO recommendations.
(Conditional recommendation, very low certainty evidence)
Remarks:
• There is no evidence that nutritional management of acute malnutrition of patients with TB
disease should be different than for those without TB.
• Concerns about weight loss or failure to gain weight should trigger further clinical assessment
(e.g. resistance to TB drugs, poor adherence, comorbid conditions) and nutrition assessment, in
order to determine the most appropriate interventions.
• Closer nutritional monitoring and earlier initiation of nutrition support (before the first 2 months
of TB treatment are completed) should be considered if the nutritional indicator is approaching
the cut-off value for a diagnosis of severe undernutrition.
45
Pyridoxine supplementation is recommended along with isoniazid treatment for all pregnant (or breastfeeding) women, as well as for
people with conditions such as HIV infection, alcohol dependency, malnutrition, diabetes, chronic liver disease or renal failure. Pyridoxine
provision together with isoniazid treatment was not analysed for the 2013 nutrition guideline.
68 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
8. Research priorities
This chapter includes research gaps or priorities that were identified by the GDG members while
considering the evidence related to each of the PICO questions. Addressing the identified research
gaps has the potential to inform the development of future research questions that can improve
TB prevention and care. This list of research priorities is not exhaustive; but it complements the
existing research agenda outlined in Research priorities for paediatric tuberculosis (127) and other
WHO guidelines.
Diagnostic approaches
The use of Xpert Ultra in gastric aspirate or stool samples to diagnose pulmonary TB in
children (adapted from 2021 rapid diagnostics guidelines (16) and 2018 research priorities
for paediatric tuberculosis (127))
• Evaluation of the benefits and incremental yield of combining multiple specimen types. Limited
data suggest that the combination of non-invasive specimens performs comparably with traditional
gastric specimens or induced sputum specimens.
8. Research priorities 69
• Additional operational and qualitative research to determine the best approach to less invasive
specimen collection in children, including: implementation studies on a method of suction for
nasopharyngeal aspiration that is appropriate for low-skill or low-resource environments; research
on the use of stool as a diagnostic specimen as part of treatment decision algorithms; definition
of laboratory protocols that successfully balance the ease of implementation and diagnostic
performance; and the impact of stool testing on patient-important outcomes.
• Identification, evaluation and validation of host and pathogen associated biomarkers in paediatric
populations as potential novel tests for TB infection, TB disease, risk of disease progression and
response to treatment among children, ideally requiring non-invasive samples and for use at the
point of care.
• Optimization of the current microbiological reference standard by improving and harmonizing
specimen collection; supporting laboratory research to improve specimen processing to optimize
diagnostic yield using current assays; and improving phenotypic and genotypic drug-susceptibility
testing on paediatric clinical specimens, including on stool samples.
• Qualitative research on equity, acceptability and feasibility aspects of diagnostic approaches,
including specimen types and diagnostic tools.
Bedaquiline
• Treatment outcomes in children with MDR/RR-TB of all ages treated with shorter and longer all-
oral, bedaquiline containing regimens.
• Studies aimed at optimizing dosing of bedaquiline in children.
• Specific cost-effectiveness analyses on the use of bedaquiline in children.
• Studies exploring mechanisms of acquisition of resistance to bedaquiline and genetic markers to
identify resistance (this evidence is likely to come from studies on adults with MDR/RR-TB but will
have implications for children and adolescents).
• Studies exploring the optimization of the duration of bedaquiline use in children related to PK
and safety.
• Studies exploring the concomitant use of bedaquiline and delamanid in children related to PK
and safety.
• Qualitative research on acceptability, equity and feasibility issues.
Delamanid
• Data on long-term safety and side-effects of delamanid, especially related to neuropsychiatric
safety signals.
• Studies aimed at optimizing dosing of delamanid in children (some studies are already ongoing,
such as IMPAACT P2005, “A phase I/II open-label, single-arm study to evaluate the PK, safety,
and tolerability of delamanid in combination with optimized multidrug background regimen
for multidrug-resistant tuberculosis (MDR-TB) in HIV-infected and HIV-uninfected children with
MDR-TB)”.
70 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
• Specific cost-effectiveness studies on the use of delamanid in children.
• Studies exploring mechanisms of acquisition of resistance to delamanid and genetic markers to
identify resistance.
• Studies exploring the optimization of the duration of delamanid use in children related to PK
and safety.
Decentralization of TB services for children and adolescents with signs and symptoms
of TB and for children and adolescents exposed to TB
• Cost-effectiveness of decentralization/integration for case detection and provision of TPT.
• Impact of decentralization of services on health equity.
• Acceptability and feasibility of decentralized approaches to child and adolescent TB care for case
detection and for TPT provision.
Family-centred, integrated services for children and adolescents with signs and
symptoms of TB and for children and adolescents exposed to TB
• Detailed description of currently operating family-centred and integrated services; associated costs
and cost-effectiveness.
• Implementation research on the components of these interventions; assessment of real-world
implementation of these programmes.
• Feasibility and acceptability of family-centred, integrated and/or decentralized approaches to child
and adolescent TB care for case detection and TPT provision in different settings, from person with
TB, caregiver and provider perspectives.
• Costs and catastrophic costs.
• Cost-effectiveness evaluations of family-centred, integrated and/or decentralized approaches,
considering currently available resources (some models assume that these interventions are built
upon existing structures that may not be available).
• Outcomes of interest: initiation of TPT; number of additional children and adolescents diagnosed;
delay, retention in care, treatment completion, clinical outcomes (such as treatment success);
qualitative research related stigma, mental health outcome, school interruption, equity.
• Evaluation of outcomes of interest using randomized/non-randomized designs and qualitative design.
• Baseline needs assessment in the community, community perceptions regarding TB care and
prevention for children and adolescents.
• Research on the quality of TB diagnosis in children – addressing both under-diagnosis and
over-diagnosis.
8. Research priorities 71
72 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
9. References
1. Global tuberculosis report 2021. Geneva: World Health Organization; 2021 (https://apps.who.int/iris/
handle/10665/346387, accessed 1 December 2021).
2. Marais B. The natural history of childhood intra-thoracic tuberculosis: a critical review of literature from the
pre-chemotherapy era. Int J Tuberc Lung Dis. 2004;8(4):392-402.
3. Martinez L, Cords O, Horsburgh CR, Andrews JR, Pediatric TB Contact Studies Consortium. The risk of
tuberculosis in children after close exposure: a systematic review and individual-participant meta-analysis.
Lancet. 2020;395(10228):973-84.
4. Snow KJ, Sismanidis C, Denholm J, Sawyer SM, Graham SM. The incidence of tuberculosis among adolescents
and young adults: a global estimate. Eur Respir J. 2018;51:1702352.
5. The Sustainable Development Agenda. [website] (https://www.un.org/sustainabledevelopment/
development-agenda/, accessed 1 December 2021.
6. The END TB Strategy. Global strategy and targets for tuberculosis prevention, care and control after 2015.
Geneva: World Health Organization; 2014 (https://www.who.int/tb/strategy/End_TB_Strategy.pdf, accessed
1 December 2021).
7. Political declaration of the UN General Assembly High-Level Meeting on the Fight Against Tuberculosis, 26
September 2018. New York: United Nations; 2019 (https://www.who.int/docs/default-source/documents/
tuberculosis/political-declaration-un-general-assembly-tb-tuberculosis.pdf?sfvrsn=4f4090dc_1&downloa
d=true, accessed 1 December 2021).
8. Guidance for national tuberculosis programmes on the management of tuberculosis in children. Second
edition. Geneva: World Health Organization; 2014 (https://apps.who.int/iris/handle/10665/112360, accessed
1 December 2021).
9. WHO consolidated guidelines on tuberculosis. Module 4: treatment - drug-resistant tuberculosis treatment.
Geneva: World Health Organization; 2020 (https://apps.who.int/iris/handle/10665/332397, accessed 2
December 2021).
10. Roadmap towards ending TB in children and adolescents. Second edition. Geneva: World Health
Organization; 2018 (https://apps.who.int/iris/handle/10665/275422, accessed 1 December 2021).
11. WHO consolidated guidelines on tuberculosis. Module 2: screening – systematic screening for tuberculosis
disease. Geneva: World Health Organization; 2021 (https://apps.who.int/iris/handle/10665/340255, accessed
1 December 2021).
12. WHO guidelines on tuberculosis infection prevention and control, 2019 update. Geneva: World Health
Organization; 2019 (https://apps.who.int/iris/handle/10665/311259, accessed 1 December 2021).
13. BCG vaccines: WHO position paper - February 2018. Weekly Epidemiological Record. 2018;93(8):73-96.
14. WHO consolidated guidelines on tuberculosis. Module 1: prevention – tuberculosis preventive treatment.
Geneva: World Health Organization; 2020 (https://apps.who.int/iris/handle/10665/331170, accessed 1
December 2021).
15. Dodd PJ, Yuen CM, Sismanidis C, Seddon JA, Jenkins HE. The global burden of tuberculosis mortality in
children: a mathematical modelling study. Lancet Glob Health. 2017;5(9):e898-e906.
9. References 73
16. WHO consolidated guidelines on tuberculosis. Module 3: diagnosis - rapid diagnostics for tuberculosis
detection, 2021 update. Geneva: World Health Organization; 2021 (https://apps.who.int/iris/
handle/10665/342331, accessed 1 December 2021).
17. Commercial serodiagnostic tests for diagnosis of tuberculosis: policy statement. Geneva: World Health
Organization; 2011 (https://apps.who.int/iris/handle/10665/44652, accessed 1 December 2021).
18. Kay AW, Gonzalez Fernandez L, Takwoingi Y, Eisenhut M, Detjen AK, Steingart KR, et al. Xpert MTB/RIF and
Xpert MTB/RIF Ultra assays for active tuberculosis and rifampicin resistance in children. Cochrane Database
Syst Rev. 2020;8:CD013359.
19. Jaganath D, Wambi P, Reza TF, Nakafeero J, Aben EO, Kiconco E, et al. A prospective evaluation of Xpert
MTB/RIF Ultra for childhood pulmonary tuberculosis in Uganda. J Pediatric Infect Dis Soc. 2021;10(5):586-92.
20. Liu XH, Xia L, Song B, Wang H, Qian XQ, Wei JH, et al. Stool-based Xpert MTB/RIF Ultra assay as a tool for
detecting pulmonary tuberculosis in children with abnormal chest imaging: A prospective cohort study. J
Infect. 2021;82(1):84-9.
21. Parigi S, Venturini E, Galli L, Chiappini E. Xpert(®) MTB/RIF Ultra performance in diagnosing paediatric
pulmonary TB in gastric aspirates. Int J Tuberc Lung Dis. 2021;25(1):75-7.
22. Ssengooba W, Iragena JD, Nakiyingi L, Mujumbi S, Wobudeya E, Mboizi R, et al. Accuracy of Xpert Ultra
in diagnosis of pulmonary tuberculosis among children in Uganda: a substudy from the SHINE trial. J Clin
Microbiol. 2020;58(9): e00410-20.
23. Validation of a tuberculosis treatment decision algorithm in HIV-infected children (TB-Speed HIV). Paris:
Institut National de la Santé Et de la Recherche Médicale; 2021 (https://clinicaltrials.gov/show/NCT04121026,
accessed 1 December 2021).
24. Development of a diagnostic prediction score for tuberculosis in hospitalized children with severe acute
malnutrition (TB-Speed SAM). Paris: Institut National de la Santé Et de la Recherche Médicale; 2021 (https://
clinicaltrials.gov/show/NCT04240990, accessed 1 December 2021).
25. Stool Processing Kit (SPK) evaluation for paediatric TB. Switzerland: Foundation for Innovative New
Diagnostics; 2021 (https://clinicaltrials.gov/ct2/show/NCT04899076 accessed 22 February 2022).
26. Kabir S, Rahman SMM, Ahmed S, Islam MS, Banu RS, Shewade HD, et al. Xpert Ultra assay on stool to
diagnose pulmonary tuberculosis in children. Clin Infect Dis. 2021;73(2):226-34.
27. Evaluation of four stool processing methods combined with Xpert MTB/RIF Ultra for diagnosis of intrathoracic
paediatric TB (TB-Speed - stool processing). Paris: Institut National de la Santé Et de la Recherche Médicale;
2021 (https://clinicaltrials.gov/ct2/show/NCT04203628 accessed 1 December 2021).
28. TB-Speed stool processing - instructions for use - sucrose flotation method. Paris: University of Bordeaux;
2020 (https://www.tb-speed.com/wp-content/uploads/2020/09/TB-Speed_Stools_Standard_Operating_
Procedures.pdf, accessed 1 December 2021).
29. Simple One Step (SOS) stool processing method and Xpert MTB/RIF (Ultra) testing for the detection of
Mycobacterium tuberculosis complex and rifampicin resistance. The Hague: KNCV Tuberculosis Foundation;
2021 (https://www.kncvtbc.org/uploaded/2021/03/Stoolbox-SOP1.pdf, accessed 1 December 2021).
30. WHO operational handbook on tuberculosis. Module 3: diagnosis - rapid diagnostics for tuberculosis
detention, 2021 update. Geneva: World Health Organization; 2021 (https://apps.who.int/iris/
handle/10665/342369, accessed 1 December 2021).
31. Graham SM, Cuevas LE, Jean-Philippe P, Browning R, Casenghi M, Detjen AK, et al. Clinical case definitions
for classification of intrathoracic tuberculosis in children: an update. Clin Infect Dis. 2015;61Suppl 3:S179-87.
32. The Union’s desk guide for diagnosis and management of TB in children. Third edition, 2016. Paris:
International Union Against Tuberculosis and Lung Disease; 2016 (https://theunion.org/sites/default/
files/2020-08/2016_Desk-guide_Africa_Web.pdf, accessed 3 December 2021).
74 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
33. Aurilio RB, Luiz RR, Land MGP, Cardoso CAA, Kritski AL, Sant’Anna CC. The clinical and molecular diagnosis
of childhood and adolescent pulmonary tuberculosis in referral centers. Rev Soc Bras Med Trop. 2020;53
(http://www.scielo.br/scielo.php?script=sci_arttext&pid=S0037-86822020000100351&nrm=iso, accessed
1 December 2021).
34. Song R, Click ES, McCarthy KD, Heilig CM, McHembere W, Smith JP, et al. Sensitive and feasible
specimen collection and testing strategies for diagnosing tuberculosis in young children. JAMA Pediatr.
2021;175(5):e206069 (https://www.ncbi.nlm.nih.gov/pubmed/33616611, accessed 1 December 2021).
35. López-Varela E, Augusto OJ, Gondo K, García-Basteiro AL, Fraile O, Ira T, et al. Incidence of tuberculosis
among young children in rural Mozambique. Pediatr Infect Dis J. 2015;34(7):686-92.
36. Myo K, Zaw M, Swe TL, Kyaw YY, Thwin T, Myo TT, et al. Evaluation of Xpert® MTB/RIF assay as a diagnostic
test for pulmonary tuberculosis in children in Myanmar. Int J Tuberc Lung Dis. 2018;22(9):1051-5.
37. Marcy O, Ung V, Goyet S, Borand L, Msellati P, Tejiokem M, et al. Performance of Xpert MTB/RIF and
alternative specimen collection methods for the diagnosis of tuberculosis in HIV-infected children. Clin
Infect Dis. 2016;62(9):1161-8.
38. Hamid M, Brooks MB, Madhani F, Ali H, Naseer MJ, The Childhood Tuberculosis Karachi Group, et al. Risk
factors for unsuccessful tuberculosis treatment outcomes in children. PLoS ONE. 2019;14(9):e0222776
(https://doi.org/10.1371/journal.pone.0222776, accessed 1 December 2021).
39. Mesman AW, Calderon R, Soto M, Coit J, Aliaga J, Mendoza M, et al. Mycobacterium tuberculosis detection
from oral swabs with Xpert MTB/RIF ULTRA: a pilot study. BMC Res Notes. 2019;12(1):349.
40. Mesman AW, Soto M, Coit J, Calderon R, Aliaga J, Pollock NR, et al. Detection of Mycobacterium tuberculosis
in pediatric stool samples using TruTip technology. BMC Infect Dis. 2019;19(1):563.
41. Tafur KT, Coit J, Leon SR, Pinedo C, Chiang SS, Contreras C, et al. Feasibility of the string test for tuberculosis
diagnosis in children between 4 and 14 years old. BMC Infect Dis. 2018;18(1):574.
42. Walters E, Demers AM, van der Zalm MM, Whitelaw A, Palmer M, Bosch C, et al. Stool culture for diagnosis
of pulmonary tuberculosis in children. J Clin Microbiol. 2017;55(12):3355-65.
43. Walters E, Scott L, Nabeta P, Demers AM, Reubenson G, Bosch C, et al. Molecular detection of Mycobacterium
tuberculosis from stools in young children by use of a novel centrifugation-free processing method. J Clin
Microbiol. 2018;56(9): e00781-18.
44. Walters E, van der Zalm MM, Palmer M, Bosch C, Demers A-M, Draper H, et al. Xpert MTB/RIF on stool
is useful for the rapid diagnosis of tuberculosis in young children with severe pulmonary disease. Pediatr
Infect Dis J. 2017;36(9):837-43.
45. Nicol MP, Workman L, Prins M, Bateman L, Ghebrekristos Y, Mbhele S, et al. Accuracy of Xpert MTB/RIF
ultra for the diagnosis of pulmonary tuberculosis in children. Pediatr Infect Dis J. 2018;37(10): e261-e263.
46. Orikiriza P, Nansumba M, Nyehangane D, Bastard M, Mugisha IT, Nansera D, et al. Xpert MTB/RIF diagnosis
of childhood tuberculosis from sputum and stool samples in a high TB-HIV-prevalent setting. Eur J Clin
Microbiol Infect Dis. 2018;37(8):1465-73.
47. Giang do C, Duong TN, Ha DT, Nhan HT, Wolbers M, Nhu NT, et al. Prospective evaluation of GeneXpert
for the diagnosis of HIV- negative pediatric TB cases. BMC Infect Dis. 2015;15:70.
48. Zawedde-Muyanja S, Nakanwagi A, Dongo JP, Sekadde MP, Nyinoburyo R, Ssentongo G, et al. Decentralisation
of child tuberculosis services increases case finding and uptake of preventive therapy in Uganda. Int J Tuberc
Lung Dis. 2018;22(11):1314-21.
49. Manual de recomendações para o controle da tuberculose no Brasil. Brasília: Ministério da Saúde; 2019
(https://bvsms.saude.gov.br/bvs/publicacoes/manual_recomendacoes_controle_tuberculose_brasil_2_
ed.pdf, accessed 30 November 2021).
9. References 75
50. Gunasekera KS, Walters E, van der Zalm MM, Palmer M, Warren JL, Hesseling AC, et al. Development of a
treatment-decision algorithm for HIV-uninfected children evaluated for pulmonary tuberculosis. Clin Infect
Dis. 2021;73(4):e904-e12.
51. Edwards K. The diagnosis of childhood tuberculosis. P N G Med J. 1987;30(2):169-78.
52. Marcy O, Borand L, Ung V, Msellati P, Tejiokem M, Huu KT, et al. A treatment-decision score for HIV-infected
children with suspected tuberculosis. Pediatrics. 2019;144(3):e20182065.
53. Montenegro SH, Gilman RH, Sheen P, Cama R, Caviedes L, Hopper T, et al. Improved detection of
Mycobacterium tuberculosis in Peruvian children by use of a heminested IS6110 polymerase chain reaction
assay. Clin Infect Dis. 2003;36(1):16-23.
54. Marais BJ, Gie RP, Hesseling AC, Schaaf HS, Lombard C, Enarson DA, et al. A refined symptom-based
approach to diagnose pulmonary tuberculosis in children. Pediatrics. 2006;118(5):e1350-9.
55. WHO consolidated guidelines on tuberculosis. Module 4: treatment - drug-susceptible tuberculosis
treatment, 2022 update. Geneva: World Health Organization; 2022.
56. Rapid advice: treatment of tuberculosis in children. Geneva: World Health Organization; 2010 (https://apps.
who.int/iris/handle/10665/44444, accessed 23 February 2022).
57. Turkova A, Wills GA, Wobudeya E, Chabala C, Palmer M, Kinikar A, et al. Shorter treatment for nonsevere
tuberculosis in African and Indian children. N Engl J Med. 2022;386(7).
58. Chabala C, Turkova A, Thomason MJ, Wobudeya E, Hissar S, Mave V, et al. Shorter treatment for minimal
tuberculosis (TB) in children (SHINE): a study protocol for a randomised controlled trial. Trials 2018;19(1):237
(https://doi.org/10.1186/s13063-018-2608-5, accessed 23 February 2022).
59. WHO case definitions of HIV for surveillance and revised clinical staging and immunological classification
of HIV-related disease in adults and children. Geneva: World Health Organization; 2007 (https://apps.who.
int/iris/handle/10665/43699, accessed 6 December 2021).
60. Guideline: updates on the management of severe acute malnutrition in infants and children. Geneva: World
Health Organization; 2013 (https://apps.who.int/iris/handle/10665/95584, accessed 23 February 2022).
61. WHO operational handbook on tuberculosis. Module 4: treatment - drug-resistant tuberculosis treatment.
Geneva: World Health Organization; 2020 (https://apps.who.int/iris/handle/10665/332398, accessed 2
December 2021).
62. Schaaf HS, Marais BJ, Whitelaw A, Hesseling AC, Eley B, Hussey GD, et al. Culture-confirmed childhood
tuberculosis in Cape Town, South Africa: a review of 596 cases. BMC Infect Dis. 2007;7(1):140 (https://doi.
org/10.1186/1471-2334-7-140, accessed 6 December 2021).
63. Wolzak NK, Cooke ML, Orth H, van Toorn R. The changing profile of pediatric meningitis at a referral centre
in Cape Town, South Africa. J Trop Pediatr. 2012;58(6):491-5.
64. Chiang SS, Khan FA, Milstein MB, Tolman AW, Benedetti A, Starke JR, et al. Treatment outcomes of childhood
tuberculous meningitis: a systematic review and meta-analysis. Lancet Infect Dis. 2014;14(10):947-57.
65. Donald PR. The chemotherapy of tuberculous meningitis in children and adults. Tuberculosis (Edinb).
2010;90(6):375-92.
66. Guidelines for the management of tuberculosis in children. Pretoria, South Africa: Department of Health;
2013.
67. van Toorn R, Schaaf HS, Laubscher JA, van Elsland SL, Donald PR, Schoeman JF. Short intensified treatment
in children with drug-susceptible tuberculous meningitis. Pediatr Infect Dis J. 2014;33(3):248-52.
68. van Well GT, Paes BF, Terwee CB, Springer P, Roord JJ, Donald PR, et al. Twenty years of pediatric tuberculous
meningitis: a retrospective cohort study in the western cape of South Africa. Pediatrics. 2009;123(1):e1-8.
76 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
69. Bang ND, Caws M, Truc TT, Duong TN, Dung NH, Ha DTM, et al. Clinical presentations, diagnosis, mortality
and prognostic markers of tuberculous meningitis in Vietnamese children: a prospective descriptive study.
BMC Infect Dis. 2016;16(1):573.
70. Solomons R. Developmental outcome at treatment completion in South African children with tuberculous
meningitis. 2015.
71. Dhawan SR, Gupta A, Singhi P, Sankhyan N, Malhi P, Khandelwal N. Predictors of neurological outcome of
tuberculous meningitis in childhood: a prospective cohort study from a developing country. J Child Neurol.
2016;31(14):1622-7.
72. Gupta R, Kushwaha S, Thakur R, Jalan N, Rawat P, Gupta P, et al. Predictors of adverse outcome in patients
of tuberculous meningitis in a multi-centric study from India. Indian J Tuberc. 2017;64(4):296-301.
73. Thee S, Basu RR, Blázquez-Gamero D, Falcón-Neyra L, Neth O, Noguera-Julian A, et al. Treatment and
outcome in children with tuberculous meningitis – a multi-centre Paediatric Tuberculosis Network European
Trials Group study. Clin Infect Dis. 2021; ciab982 (doi: 10.1093/cid/ciab982, accessed 23 February 2022).
74. Garcia-Prats AJ, Svensson EM, Winckler J, Draper HR, Fairlie L, van der Laan LE, et al. Pharmacokinetics
and safety of high-dose rifampicin in children with TB: the Opti-Rif trial. J Antimicrob Chemother. 2021;
76(12):3237-46.
75. Dodd PJ, Sismanidis C, Seddon JA. Global burden of drug-resistant tuberculosis in children: a mathematical
modelling study. Lancet Infect Dis. 2016;16(10):1193-1201.
76. Jenkins HE, Tolman AW, Yuen CM, Parr JB, Keshavjee S, Pérez-Vélez CM, et al. Incidence of multidrug-resistant
tuberculosis disease in children: systematic review and global estimates. Lancet. 2014;383(9928):1572-9.
77. Jenkins HE, Yuen CM. The burden of multidrug-resistant tuberculosis in children. Int J Tuberc Lung Dis.
2018;22(5):3-6.
78. Global tuberculosis report 2020. Geneva: World Health Organization; 2020 (https://apps.who.int/iris/
handle/10665/336069, accessed 1 December 2021).
79. Harausz EP, Garcia-Prats AJ, Law S, Schaaf HS, Kredo T, Seddon JA, et al. Treatment and outcomes in
children with multidrug-resistant tuberculosis: A systematic review and individual patient data meta-
analysis. PLoS Med. 2018;15(7):e1002591 (https://journals.plos.org/plosmedicine/article?id=10.1371/journal.
pmed.1002591, accessed 23 February 2022).
80. Prescribing information. Sirturo (bedaquiline) product insert. Silver Spring: Food and Drug Administration
(https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/204384s000lbl.pdf, accessed 1 December
2021).
81. The use of bedaquiline in the treatment of multidrug-resistant tuberculosis: interim policy guidance. Geneva:
World Health Organization; 2013 (https://apps.who.int/iris/handle/10665/84879, accessed 1 December
2021).
82. Report of the Guideline Development Group Meeting on the use of bedaquiline in the treatment of
multidrug-resistant tuberculosis, A review of available evidence (2016). Geneva: World Health Organization;
2017 (https://apps.who.int/iris/handle/10665/254712, accessed 1 December 2021).
83. WHO consolidated guidelines on drug-resistant tuberculosis treatment. Geneva: World Health Organization;
2019 (https://apps.who.int/iris/handle/10665/311389, accessed 1 December 2021).
84. WHO consolidated guidelines on tuberculosis. Module 4: treatment - drug-resistant tuberculosis treatment.
Online annexes. Geneva: World Health Organization; 2020 (https://apps.who.int/iris/handle/10665/332678,
accessed 1 December 2021).
85. Svensson EM, du Bois J, Kitshoff R, de Jager VR, Wiesner L, Norman J, et al. Relative bioavailability of
bedaquiline tablets suspended in water: Implications for dosing in children. Br J Clin Pharmacol. 2018;
84(10):2384-92.
9. References 77
86. Wiseman CA, Gie RP, Starke JR, Schaaf HS, Donald PR, Cotton MF et al. A proposed comprehensive
classification of tuberculosis disease severity in children. Pediatr Infect Dis J. 2012;31(4):347-52.
87. Catalogue of mutations in Mycobacterium tuberculosis complex and their association with drug resistance.
Geneva: World Health Organization; 2021 (https://apps.who.int/iris/handle/10665/341981, accessed 1
December 2021).
88. Model List of Essential Medicines for Children – 8th List (2021). Geneva: World Health Organization; 2021
(https://apps.who.int/iris/handle/10665/345534, accessed 1 December 2021).
89. Active tuberculosis drug-safety monitoring and management (aDSM). Framework for implementation.
Geneva: World Health Organization; 2015 (https://apps.who.int/iris/handle/10665/204465, accessed 1
December 2021).
90. Deltyba: EPAR - Product Information. Amsterdam: European Medicines Agency; 2014 (https://www.ema.
europa.eu/en/documents/product-information/deltyba-epar-product-information_en.pdf, accessed 1
December 2021).
91. The use of delamanid in the treatment of multidrug-resistant tuberculosis. Interim policy guidance. Geneva:
World Health Organization; 2014 (https://apps.who.int/iris/handle/10665/137334, accessed 1 December
2021).
92. The use of delamanid in the treatment of multidrug-resistant tuberculosis in children and adolescents: interim
policy guidance. Geneva: World Health Organization; 2016 (https://apps.who.int/iris/handle/10665/250614,
accessed 1 December 2021).
93. WHO treatment guidelines for drug-resistant tuberculosis, 2016 update. October 2016 revision. Geneva:
World Health Organization; 2016 (https://apps.who.int/iris/handle/10665/250125, accessed 1 December
2021).
94. Companion handbook to the WHO guidelines for the programmatic management of drug-resistant
tuberculosis. Geneva: World Health Organization; 2014 (https://apps.who.int/iris/handle/10665/130918,
accessed 1 December 2021).
95. A5343. A trial of the safety, tolerability, and pharmacokinetics of bedaquiline and delamanid, alone and in
combination, among participants taking multidrug treatment for drug-resistant pulmonary tuberculosis.
A limited-center trial of the AIDS Clinical Trials Group (ACTG). Rockville: National Institute of Allergy and
Infectious Diseases; 2018 (https://clinicaltrials.gov/ProvidedDocs/48/NCT02583048/Prot_001.pdf, accessed
1 December 2021).
96. Consolidated guidelines on drug-resistant tuberculosis treatment, 2018 update [online annexes]. Geneva:
World Health Organization; 2018 (https://apps.who.int/iris/handle/10665/332678, accessed 1 December
2021).
97. Guidelines for treatment of drug-susceptible tuberculosis and patient care, 2017 update. Geneva: World
Health Organization; 2017 (https://apps.who.int/iris/handle/10665/255052, accessed 23 February 2022).
98. Roadmap for childhood tuberculosis: towards zero deaths. Geneva: World Health Organization; 2013
(https://apps.who.int/iris/handle/10665/89506, accessed 23 February 2022).
99. Public–private mix for TB prevention and care: a roadmap. Geneva: World Health Organization; 2018
(https://apps.who.int/iris/handle/10665/333885, accessed 1 December 2021).
100. WHO consolidated guidelines on tuberculosis. Module 4: treatment - care and support during tuberculosis
treatment. Geneva: World Health Organization; 2022.
101. Talukder K, Salim MA, Jerin I, Sharmin F, Talukder MQ, Marais BJ, et al. Intervention to increase detection
of childhood tuberculosis in Bangladesh. Int J Tuberc Lung Dis. 2012;16(1):70-5.
102. Khan AJ, Khowaja S, Khan FS, Qazi F, Lotia I, Habib A, et al. Engaging the private sector to increase
tuberculosis case detection: an impact evaluation study. Lancet Infect Dis. 2012;12(8):608-16.
78 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
103. Malik AA, Amanullah F, Codlin AJ, Siddiqui S, Jaswal M, Ahmed JF, et al. Improving childhood tuberculosis
detection and treatment through facility-based screening in rural Pakistan. Int J Tuberc Lung Dis.
2018;22(8):851-7.
104. Maha A, Majumdar SS, Main S, Phillip W, Witari K, Schulz J, et al. The effects of decentralisation of tuberculosis
services in the East New Britain Province, Papua New Guinea. Public Health Action. 2019;9(Suppl 1):S43-S49.
105. Islam Z, Sanin KI, Ahmed T. Improving case detection of tuberculosis among children in Bangladesh: lessons
learned through an implementation research. BMC Public Health. 2017;17(1):131.
106. Catalyzing Pediatric TB Innovation (CaP-TB) project. Unpublished data. 2021.
107. Oshi DC, Chukwu JN, Nwafor CC, Meka AO, Madichie NO, Ogbudebe CL, et al. Does intensified case finding
increase tuberculosis case notification among children in resource-poor settings? A report from Nigeria.
Int J Mycobacteriol. 2016;5(1):44-50.
108. Joshi B, Chinnakali P, Shrestha A, Das M, Kumar AM, Pant R, et al. Impact of intensified case-finding strategies
on childhood TB case registration in Nepal. Public Health Action. 2015;5(2):93-8.
109. Hanrahan CF, Nonyane BAS, Mmolawa L, West NS, Siwelana T, Lebina L, et al. Contact tracing versus
facility-based screening for active TB case finding in rural South Africa: A pragmatic cluster-randomized
trial (Kharitode TB). PLoS Med. 2019;16(4):e1002796 (https://journals.plos.org/plosmedicine/article/
file?id=10.1371/journal.pmed.1002796&type=printable, accessed 23 February 2022).
110. Moyo S, Verver S, Hawkridge A, Geiter L, Hatherill M, Workman L, et al. Tuberculosis case finding for
vaccine trials in young children in high-incidence settings: a randomised trial. Int J Tuberc Lung Dis.
2012;16(2):185-91.
111. Davis JL, Turimumahoro P, Meyer AJ, Ayakaka I, Ochom E, Ggita J, et al. Home-based tuberculosis
contact investigation in Uganda: a household randomised trial. ERJ Open Res. 2019;5(3): 00112 (doi:
10.1183/23120541.00112-2019, accessed 23 February 2022).
112. Fatima R, Qadeer E, Yaqoob A, Haq Mu, Majumdar SS, Shewade HD, et al. Extending ‘contact tracing’ into
the community within a 50-metre radius of an index tuberculosis patient using Xpert MTB/RIF in urban,
Pakistan: did it increase case detection? PLoS ONE. 2016;11(11):e0165813 (https://doi.org/10.1371/journal.
pone.0165813, accessed 1 December 2021).
113. Reddy KK, Ananthakrishnan R, Jacob AG, Das M, Isaakidis P, Kumar AMV. Intensified tuberculosis case finding
amongst vulnerable communities in southern India. Public Health Action. 2015;5(4):246-8.
114. Bayona J, Chavez-Pachas AM, Palacios E, Llaro K, Sapag R, Becerra MC. Contact investigations as a means
of detection and timely treatment of persons with infectious multidrug-resistant tuberculosis. Int J Tuberc
Lung Dis. 2003;7(12 Suppl 3):S501-9.
115. Sachdeva KS, Raizada N, Sreenivas A, van’t Hoog AH, van den Hof S, Dewan PK, et al. Use of Xpert MTB/
RIF in decentralized public health settings and its effect on pulmonary TB and DR-TB case finding in India.
PLoS ONE. 2015;10(5):e0126065 (https://doi.org/10.1371/journal.pone.0126065, accessed 1 December
2021).
116. Yassin MA, Datiko DG, Tulloch O, Markos P, Aschalew M, Shargie EB, et al. Innovative community-based
approaches doubled tuberculosis case notification and improve treatment outcome in Southern Ethiopia.
PLoS ONE. 2013;8(5):e63174 (https://doi.org/10.1371/journal.pone.0063174, accessed 1 December 2021).
117. Zachariah R, Spielmann MP, Harries AD, Gomani P, Graham SM, Bakali E, et al. Passive versus active
tuberculosis case finding and isoniazid preventive therapy among household contacts in a rural district of
Malawi. Int J Tuberc Lung Dis. 2003;7(11):1033-9.
118. Ketema L, Dememew ZG, Assefa D, Gudina T, Kassa A, Letta T, et al. Evaluating the integration of tuberculosis
screening and contact investigation in tuberculosis clinics in Ethiopia: A mixed method study. PLoS ONE.
2020;15(11):e0241977 (https://doi.org/10.1371/journal.pone.0241977, accessed 23 February 2022).
9. References 79
119. Miyano S, Dube C, Kayama N, Ishikawa N, Nozaki I, Syakantu G. Association between tuberculosis
treatment outcomes and the mobile antiretroviral therapy programme in Zambia. Int J Tuberc Lung Dis.
2013;17(4):540-5.
120. Wingfield T, Tovar MA, Huff D, Boccia D, Montoya R, Ramos E, et al. A randomized controlled study of
socioeconomic support to enhance tuberculosis prevention and treatment, Peru. Bull World Health Organ.
2017;95(4):270-80.
121. Rocha C, Montoya R, Zevallos K, Curatola A, Ynga W, Franco J, et al. The Innovative Socio-economic
Interventions Against Tuberculosis (ISIAT) project: an operational assessment. Int J Tuberc Lung Dis. 2011;15
Suppl 2(Suppl 2):50-7.
122. Latent tuberculosis infection: updated and consolidated guidelines for programmatic management.
Geneva: World Health Organization; 2018 (https://apps.who.int/iris/handle/10665/260233, accessed 23
February 2022).
123. Updated recommendations on service delivery for the treatment and care of people living with HIV. Geneva:
World Health Organization; 2021 (https://apps.who.int/iris/handle/10665/341052, accessed 23 February
2022).
124. WHO policy on collaborative TB/HIV activities: guidelines for national programmes and other stakeholders.
Geneva: World Health Organization; 2012 (https://apps.who.int/iris/handle/10665/44789, accessed 1
December 2021).
125. Consolidated guidelines on HIV prevention, testing, treatment, service delivery and monitoring:
recommendations for a public health approach. Geneva: World Health Organization; 2021 (https://apps.
who.int/iris/handle/10665/342899, accessed 1 December 2021).
126. Guideline: nutritional care and support for patients with tuberculosis. Geneva: World Health Organization;
2013 (https://apps.who.int/iris/handle/10665/94836, accessed 6 December 2021).
127. Research priorities for paediatric tuberculosis. New York: Treatment Action Group, Child and Adolescent
TB Working Group; 2018 (https://www.treatmentactiongroup.org/wp-content/uploads/2018/09/Paediatric_
TB_ResearchPriorities_10_8_18_Web.pdf, accessed 1 December 2021).
80 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Annex 1. WHO
recommendations incorporated
in the guidelines on the
management of TB in children
and adolescents
In order of presentation:
WHO consolidated guidelines on tuberculosis. Module 2: screening – systematic screening for
tuberculosis disease. Geneva: World Health Organization; 2021.
Recommendations for investigating contacts of persons with infectious tuberculosis in low- and
middle-income countries. Geneva: World Health Organization; 2012.
WHO guidelines on tuberculosis infection prevention and control, 2019 update. Geneva: World Health
Organization; 2019.
BCG vaccines: WHO position paper – February 2018. Weekly Epidemiological Record. 2018;93(08):73–96.
WHO consolidated guidelines on tuberculosis. Module 1: prevention – tuberculosis preventive
treatment. Geneva: World Health Organization; 2020.
WHO consolidated guidelines on tuberculosis. Module 3: diagnosis – rapid diagnostics for tuberculosis
detection, 2021 update. Geneva: World Health Organization; 2021.
WHO consolidated guidelines on tuberculosis. Module 4: treatment – drug-susceptible tuberculosis
treatment, 2022 update.
WHO consolidated guidelines on tuberculosis. Module 4: treatment – drug-resistant tuberculosis
treatment. Geneva: World Health Organization; 2020.
WHO consolidated guidelines on tuberculosis. Module 4: treatment – care and support during
tuberculosis treatment. Geneva: World Health Organization; 2022.
Guidance for national tuberculosis programmes on the management of tuberculosis in children.
Second edition. Geneva: World Health Organization; 2014.
Updated recommendations on HIV prevention, infant diagnosis, antiretroviral initiation and monitoring:
March 2021. Geneva: World Health Organization; 2021.
Guideline: nutritional care and support for patients with tuberculosis. Geneva: World Health
Organization; 2013.
Guideline: updates on the management of severe acute malnutrition in infants and children. Geneva:
World Health Organization; 2013.
Essential nutrition actions: mainstreaming nutrition through the life-course. Geneva: World Health
Organization; 2019.
Annex 1. WHO recommendations incorporated in the guidelines on the management of TB in children and adolescents 81
82 WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
Annex 2. Supplementary table
Summary of changes to recommendations as
included in the second edition of the Guidance
for national tuberculosis programmes on the
management of tuberculosis in children, 2014
46
Status: Removed (the recommendation has been superseded and is no longer relevant); Copied (the recommendation remains valid and is retained unchanged); Edited (There is no change in the evidence
or in the intention of the recommendation, but the precise wording has been edited); Updated (a new evidence synthesis was conducted with review by the GDG with a full evidence-to-decision procedure);
Developed de novo (a new topic, subgroup or intervention has been covered with a new evidence synthesis and a full evidence-to-decision procedure by the GDG)
Recommendation in Guidance for national
tuberculosis programmes on the management
No. Status46 Updated recommendation and source guideline
of tuberculosis in children (second edition) 2014,
and source guideline
3 Xpert MTB/RIF may be used as a replacement test Updated In adults and children with signs and symptoms of
for usual practice (including conventional microscopy, extrapulmonary TB, Xpert MTB/RIF may be used in lymph
culture, and/or histopathology) for testing of specific node aspirate, lymph node biopsy, pleural fluid, peritoneal
non-respiratory specimens (lymph nodes and fluid, pericardial fluid, synovial fluid or urine specimens as the
other tissues) from children suspected of having initial diagnostic test rather than smear microscopy/culture.
extrapulmonary TB (Conditional recommendation, moderate certainty of evidence
(Conditional recommendation, very low quality for test accuracy for pleural fluid; low certainty for lymph node
of evidence) aspirate, peritoneal fluid, synovial fluid, urine; very low certainty
Source: Automated real-time nucleic acid amplification technology for rapid for pericardial fluid, lymph nodes biopsy)
and simultaneous detection of tuberculosis and rifampicin resistance: Xpert In adults and children with signs and symptoms of
MTB/RIF system for the diagnosis of pulmonary and extrapulmonary TB in extrapulmonary TB, Xpert Ultra may be used in lymph node
adults and children. Policy Update. 2013. aspirate and lymph node biopsy as the initial diagnostic test
rather than smear microscopy/culture.
(Conditional recommendation, low certainty of evidence)
In adults and children with signs and symptoms of
extrapulmonary TB, Xpert MTB/RIF or Xpert Ultra should be
used for rifampicin-resistance detection rather than culture and
phenotypic DST.
(Strong recommendation, high certainty of evidence for test
accuracy for Xpert MTB/RIF; low certainty of evidence for
Xpert Ultra)
In HIV-positive adults and children with signs and symptoms of
disseminated TB, Xpert MTB/RIF may be used in blood, as an
Annex 2. Supplementary table
5 Interferon-gamma release assays (IGRAs) should Updated Either a tuberculin skin test (TST) or interferon-gamma release
not replace the tuberculin skin test (TST) in low- and assay (IGRA) can be used to test for TB infection.
middle-income countries for the diagnosis of TB (Strong recommendation, very low certainty in the estimates
infection in children or for the diagnostic work-up of of effect)
children (irrespective of HIV status) suspected of TB
Source: WHO consolidated guidelines on tuberculosis. Module 1: prevention –
disease in these settings.
Tuberculosis preventive treatment. 2020.
(Strong recommendation, low quality of evidence)
Source: Use of tuberculosis interferon-gamma release assays (IGRAs) in low-
and middle-income countries: policy statement. 2011.
6 Commercial serodiagnostics should not be used Copied Commercial serodiagnostics should not be used in children
in children suspected of active pulmonary or suspected of active pulmonary or extrapulmonary TB,
extrapulmonary TB, irrespective of their HIV status irrespective of their HIV status
(Strong recommendation, very low quality of evidence (Strong recommendation, very low certainty of evidence for the
for the use of commercial serodiagnostics) use of commercial serodiagnostics)
Source: Commercial serodiagnostic tests for diagnosis of tuberculosis: policy Source: Commercial serodiagnostic tests for diagnosis of tuberculosis: policy statement.
statement. 2011. 2011.
Recommendation in Guidance for national
tuberculosis programmes on the management
No. Status46 Updated recommendation and source guideline
of tuberculosis in children (second edition) 2014,
and source guideline
7 Routine HIV testing should be offered to all patients, Copied Routine HIV testing should be offered to all patients, with
including children, with presumptive and diagnosed TB presumptive and diagnosed TB
(Strong recommendation, low quality of evidence) (Strong recommendation, low certainty of evidence)
Source: WHO policy on collaborative TB/HIV activities, guidelines for national Source: WHO policy on collaborative TB/HIV activities, guidelines for national programmes
programmes and other stakeholders. 2012. and other stakeholders. 2012.
47
Defined as countries, subnational administrative units, or selected facilities, where the HIV prevalence among adult pregnant women is ≥1% or among TB patients is ≥5% in the 2014 Guidance for national
tuberculosis programmes on the management of tuberculosis in children (second edition) (8).
48
WHO does not intend to establish thresholds for low, moderate or high levels of prevalence of isoniazid resistance: NTPs will establish definitions for their own countries.
Recommendation in Guidance for national
tuberculosis programmes on the management
No. Status46 Updated recommendation and source guideline
of tuberculosis in children (second edition) 2014,
and source guideline
11 Infants aged 0–3 months with suspected or Copied and edited Infants aged 0–3 months with suspected or confirmed
confirmed pulmonary TB or tuberculous peripheral slightly (reference to pulmonary TB or tuberculous peripheral lymphadenitis should
lymphadenitis should be promptly treated with recommendations 9 be promptly treated with the 6-month treatment regimen
the standard treatment regimens, as described in and 10 is removed (2HRZ(E)/4HR). Treatment may require dose adjustment to
recommendation 9 or 10. Treatment may require and the standard reconcile the effect of age and possible toxicity in young
dose adjustment to reconcile the effect of age and treatment regimen infants. The decision to adjust doses should be taken by a
possible toxicity in young infants. The decision has been replaced clinician experienced in managing paediatric TB.
to adjust doses should be taken by a clinician with the 6-month (Strong recommendation, low certainty of evidence)
experienced in managing paediatric TB. treatment regimen)
Source: Rapid advice: treatment of tuberculosis in children. 2010.
(Strong recommendation, low quality of evidence)
Source: Rapid advice: treatment of tuberculosis in children. 2010.
12 During the continuation phase of treatment, thrice- Updated In all patients with drug-susceptible pulmonary TB, the use of
weekly regimens can be considered for children thrice-weekly dosing is not recommended in both the intensive
known not to be HIV-infected and living in settings and continuation phases of therapy, and daily dosing remains
with well-established directly-observed therapy the recommended dosing frequency.
(DOT). (Conditional recommendation, very low certainty in the
(Conditional recommendation, very low quality of evidence)49
evidence for use of intermittent treatment in children Source: WHO consolidated guidelines on tuberculosis. Module 4: treatment – drug-
in specific settings) susceptible tuberculosis treatment, 2022 update.
Source: Rapid advice: treatment of tuberculosis in children. 2010.
13 Streptomycin should not be used as part of first-line Copied Streptomycin should not be used as part of first-line treatment
treatment regimens for children with pulmonary TB regimens for children with pulmonary TB or tuberculous
Annex 2. Supplementary table
49
Twice-weekly dosing is totally not recommended. See Guidelines for treatment of tuberculosis, fourth edition. Geneva: World Health Organization; 2010 (http://www.who.int/tb/publications/2010/9789241547833/
en/).
89
90
WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
50
The Global Advisory Committee on Vaccine Safety (GACVS) does not use the GRADE methodology for evaluating the quality of evidence; the BCG-related recommendations will therefore remain ungraded.
91
92
WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
16 In children who are known to be HIV-infected, BCG Updated in WHO Children who are HIV-infected when vaccinated with BCG at
vaccine should not be given. position paper birth are at increased risk of developing disseminated BCG
(Recommendation strength and evidence quality have (based on advice disease. However, if HIV-infected individuals including children,
not been graded)4 from the WHO SAGE) are receiving ART, are clinically well and immunologically stable
(CD4% >25% for children aged <5 years or CD4 count ≥200 if
Source: Revised BCG vaccination guidelines for infants at risk for HIV
aged >5 years) they should be vaccinated with BCG.
infection. Weekly Epidemiological Record. 2007;82:193–196.
• In general, populations with high prevalence of HIV infection
also have the greatest burden of TB; in such populations
the benefits of potentially preventing severe TB through
vaccination at birth are outweighed by the risks associated
with the use of BCG vaccine. Therefore, it is recommended
that in such populations:
• Neonates born to women of unknown HIV status should
be vaccinated as the benefits of BCG vaccination outweigh
the risks.
• Neonates of unknown HIV status born to HIV-infected
women should be vaccinated if they have no clinical evidence
suggestive of HIV infection, regardless of whether the mother
is receiving ART.
Recommendation in Guidance for national
tuberculosis programmes on the management
No. Status46 Updated recommendation and source guideline
of tuberculosis in children (second edition) 2014,
and source guideline
Although evidence is limited, for neonates with HIV infection
confirmed by early virological testing, BCG vaccination should
be delayed until ART has been started and the infant confirmed
to be clinically and immunologically stable (CD4% >25%).
Source: BCG vaccines: WHO position paper – February 2018. Weekly Epidemiological
Record. 2018;93(8):73–96.
17 In infants whose HIV status is unknown and who Updated in WHO Children who are HIV-infected when vaccinated with BCG at
are born to HIV-positive mothers and who lack position paper birth are at increased risk of developing disseminated BCG
symptoms suggestive of HIV, BCG vaccine should be (based on advice disease. However, if HIV-infected individuals, including children,
given after considering local factors from the WHO SAGE) are receiving ART, are clinically well and immunologically stable
(Recommendation strength and evidence quality have (CD4% >25% for children aged <5 years or CD4 count ≥200 if
not been graded) 4 aged >5 years) they should be vaccinated with BCG.
Source: Revised BCG vaccination guidelines for infants at risk for HIV • In general, populations with high prevalence of HIV infection
infection. Weekly Epidemiological Record. 2007;82:193–196. also have the greatest burden of TB; in such populations
the benefits of potentially preventing severe TB through
vaccination at birth are outweighed by the risks associated
with the use of BCG vaccine. Therefore, it is recommended
that in such populations:
– Neonates born to women of unknown HIV status should
be vaccinated as the benefits of BCG vaccination outweigh
the risks.
– Neonates of unknown HIV status born to HIV-infected
women should be vaccinated if they have no clinical
Annex 2. Supplementary table
21 Children <5 years of age who are household or Updated Children aged <5 years who are household contacts of people
close contacts of people with TB and who, after an with bacteriologically confirmed pulmonary TB and who
appropriate clinical evaluation, are found not to are found not to have TB disease on an appropriate clinical
have TB disease should be given 6 months of IPT evaluation or according to national guidelines should be
(10 mg/kg per day, range 7–15 mg/kg, maximum given TB preventive treatment even if TB infection testing is
dose 300 mg/day). unavailable.
(Strong recommendation, high quality of evidence) (Strong recommendation, high certainty in the estimates of effect)
Source: Recommendations for investigating contacts of persons with Children aged ≥5 years, adolescents and adults who are
infectious tuberculosis in low- and middle-income countries. 2012. household contacts of people with bacteriologically confirmed
pulmonary TB who are found not to have TB disease by
an appropriate clinical evaluation or according to national
guidelines may be given TB preventive treatment.
(Conditional recommendation, low certainty in the estimates of
effect)
Annex 2. Supplementary table
51
In ages two years and above.
95
96
WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
22 In settings of high HIV prevalence, all household Copied In settings of high HIV prevalence, all household and close
and close contacts of people with TB should be contacts of people with TB should be counselled and tested
counselled and tested for HIV. for HIV.
(Strong recommendation, very low quality of evidence) (Strong recommendation, very low quality of evidence)
Source: Recommendations for investigating contacts of persons with Source: Recommendations for investigating contacts of persons with infectious
infectious tuberculosis in low- and middle-income countries. 2012. tuberculosis in low- and middle-income countries. 2012.
52
In ages 13 and above.
Recommendation in Guidance for national
tuberculosis programmes on the management
No. Status46 Updated recommendation and source guideline
of tuberculosis in children (second edition) 2014,
and source guideline
23 In settings of low HIV prevalence, all household Copied In settings of low HIV prevalence, all household members
members and close contacts of people with TB who and close contacts of people with TB who have symptoms
have symptoms compatible with TB disease may be compatible with TB disease may be offered counselling and
offered counselling and testing for HIV as part of their testing for HIV as part of their clinical evaluation.
clinical evaluation. (Conditional recommendation, very low quality of evidence)
(Conditional recommendation, very low quality Source: Recommendations for investigating contacts of persons with infectious
of evidence) tuberculosis in low- and middle-income countries. 2012.
Source: Recommendations for investigating contacts of persons with
infectious tuberculosis in low- and middle-income countries. 2012.
24 All household contacts of an index case who is a Copied All household contacts of an index case who is a person living
person living with HIV should be counselled and with HIV should be counselled and tested for HIV.
tested for HIV. (Strong recommendation, very low quality of evidence)
(Strong recommendation, very low quality of evidence) Source: Recommendations for investigating contacts of persons with infectious
Source: Recommendations for investigating contacts of persons with tuberculosis in low- and middle-income countries. 2012.
infectious tuberculosis in low- and middle-income countries. 2012.
Annex 2. Supplementary table
97
98
WHO consolidated guidelines on tuberculosis, Module 5: Management of tuberculosis in children and adolescents
27 Children with proven or suspected pulmonary TB Updated In children with MDR/RR-TB aged below 6 years, an all-oral
or tuberculous meningitis caused by multidrug- treatment regimen containing bedaquiline may be used.
resistant bacilli can be treated with a fluoroquinolone (NEW Conditional recommendation, very low certainty of
in the context of a well-functioning MDR-TB control the evidence).
programme and within an appropriate MDR-TB
regimen. The decision to treat should be taken by a This recommendation applies to and complements current
clinician experienced in managing paediatric TB. WHO recommendations on shorter and longer regimens that
contain bedaquiline:
(Strong recommendation, very low quality evidence)
• A shorter all-oral bedaquiline-containing regimen of
Source: Rapid advice: treatment of tuberculosis in children. 2010.
9–12 months duration is recommended in eligible patients
with confirmed multidrug- or rifampicin-resistant tuberculosis
(MDR/RR-TB) who have not been exposed to treatment with
second-line TB medicines used in this regimen for more than
Annex 2. Supplementary table
- New interim recommendation Developed de novo In children with presumptive pulmonary TB attending health
care facilities, integrated treatment decision algorithms may be
used to diagnose pulmonary TB.
(NEW Interim conditional recommendation, very low
certainty evidence)
Source: WHO consolidated guidelines on tuberculosis. Module 5: management of
tuberculosis in children and adolescents, 2022.
- New recommendation Developed de novo In TB high burden settings, decentralized TB services may be
used in children and adolescents with signs and symptoms of
TB and/or those exposed to TB.
(NEW Conditional recommendation, very low
certainty evidence)
Family-centred, integrated services in addition to standard TB
Annex 2. Supplementary table