0% found this document useful (0 votes)
215 views126 pages

Ucm370435 PDF

Long-chain Inulin (lc-inulin) prepared from chicory-derived inulin. Inulin is used in term infant formulas, toddler formulas, and medical foods. Ingredient is exempt from premarket approval requirements of the federal food, drug and cosmetic act.
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
215 views126 pages

Ucm370435 PDF

Long-chain Inulin (lc-inulin) prepared from chicory-derived inulin. Inulin is used in term infant formulas, toddler formulas, and medical foods. Ingredient is exempt from premarket approval requirements of the federal food, drug and cosmetic act.
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 126

GRAS Notice (GRN) No.

477
http://www.fda.gov/Food/IngredientsPackagingLabeling/GRAS/NoticeInventory/default.htm

ORIGINAL SUBMISSION

000001

(b) (6)

000002

June 7, 2013
Via Express Courier
Ms. Paulette Gaynor
GRAS Notification Program
Office of Food Additive Safety (HFS-200)
Center for Food Safety and Applied Nutrition
Food and Drug Administration
5100 Paint Branch Parkway
College Park, MD 20740-3835
Re:

GRAS Notification for Long-chain Inulin (lc-inulin)

Dear Ms. Gaynor:


On behalf of Danone Trading B.V. (the Notifier), ENVIRON International Corporation is pleased to
submit this Notification of the Generally Recognized as Safe (GRAS) Determination for the
ingredient, long-chain inulin (lc-inulin) prepared from chicory-derived inulin for use in term infant
formulas, toddler formulas, and medical foods. This Notification contains 1) the Notifiers GRAS
Exemption Claim; 2) the Expert Panel Report on the GRAS Status of the ingredient; and 3) the
Safety Evaluation report.
Sincerely,
(b) (6)

Gavin Thompson
Principal
GT:gs
28-26968C

ENVIRON International Corp. 1702 East Highland Avenue, Suite 412, Phoenix, AZ 85016
V +1 602.734.7700 F +1 602.734.7701
environcorp.com

000003

GRAS Exemption Claim for


Long-Chain Inulin (lc-inulin)
Danone has determined that the use of long-chain inulin (lc-inulin) prepared from chicoryderived inulin in term infant formulas, toddler formulas, and medical foods is exempt from the
premarket approval requirements of the Federal Food, Drug and Cosmetic Act because Danone
has determined such use to be Generally Recognized As Safe (GRAS). This determination is in
compliance with proposed Sec. 170.36 of Part 21 of the Code of Federal Regulations (21 CFR
170.36) as published in the Federal Register, Vol. 62, No. 74, FR 18937, April 17, 1997.

A. Name and Address of Notifier


Mr. Flemming Morgan
President
Danone Trading B.V.
Schiphol Boulevard 105, Tower E, 1118 BG Schiphol Airport,
The Netherlands

B. Common or Usual Name of GRAS Substance


The substance that is the subject of this GRAS determination is long-chain inulin (lc-inulin).
Other chemical names for this lc-inulin are inulin or high performance inulin.
The lc-inulin product that is the subject of this GRAS determination is prepared from chicoryderived inulin by Orafti (Belgium) and sold under the trade name BeneoTM HP (formerly known
as Raftiline HP).

C. Intended Use
A combination of GOS and lc-inulin, in a 9:1 ratio of GOS:lc-inulin, is intended to be added to
term infant formulas, toddler formulas and medical foods. The intended uses of the GOS+lcinulin mixture and the typical and maximum concentrations of the mixture are detailed in
Table 1.

Table 1: Intended Uses of GOS+Long-Chain Inulin (9:1) in Term Infant Formulas,


Toddler Formulas and Medical Foods
Product Category
Intended Use
Total GOS+lc-Inulin Content(a)
Term infant formulas and
toddler formulas
Non-exempt and exempt
term infant formulas

Healthy term infants beginning at


birth and healthy term infants with
inborn errors of metabolism,
beginning at birth

8 g/L

Toddler formulas

Healthy children from 12 through 35


months of age

12 g/L

000004

GRAS Exemption Claim


for Long-Chain Inulin (lc-inulin)

-2-

Table 1: Intended Uses of GOS+Long-Chain Inulin (9:1) in Term Infant Formulas,


Toddler Formulas and Medical Foods
Product Category
Intended Use
Total GOS+lc-Inulin Content(a)
Maximum
Typical
Medical foods(c)
Nutritionally incomplete oral
medical foods

The products are intended to


supplement the normal diet and
cannot be used as the sole source of
nutrition. Nutritionally incomplete
products may be either general
formulas or disease-specific
formulas.

25 g /1000 kcal(b)

12.5 g/1000 kcal

Nutritionally complete foods


Oral foods

The products are intended to


supplement the normal diet but can
be used as the sole source of
nutrition when given in the
recommended amount. Nutritionally
complete products may be either
general formulas or disease-specific
formulas.

20 g /1000 kcal

10 g /1000 kcal

Tube feedings

Tube feeds are intended for enteral


nutritional support for those patients
with a functional or partially
functional gastro-intestinal tract who
are unable or unwilling to eat
sufficient quantities of conventional
food to meet their nutritional
requirements. The products are
nutritionally complete and intended
as the sole source of nutrition for the
patients. Tube feeds can either be
standard formulas or disease specific
formulas.

20 g /1000 kcal

10 g /1000 kcal

Enteral nutritional products


Sole source enteral feedings or
12 g/L
for children (pediatric
supplemental nutrition for children
nutritionals)
Notes:
a
The ratio of GOS:lc-inulin is 9:1.
b
For certain disease-specific formulas with low energy densities (e.g., products for the dietary management
of metabolic disease), the maximum use of GOS+lc-inulin is 31.25 g per 1000 kcal.
c
The upper levels of GOS+lc-inulin intake may be well tolerated by some but not necessarily all people. A
specific dose of fiber may be achieved by using a fiber-containing medical food in combination of a fiber
free version of the same medical food.
Abbreviations: g = grams; GOS = galacto-oligosaccharide; kcal = kilocalories; L = liter; lc-inulin = long-chain
inulin

000005

(b) (6)

000006

Expert Panel Report on the


Generally Recognized as Safe Status of the
Proposed Uses of Long-Chain Inulin in
Term Infant Formulas, Toddler Formulas,
and Medical Foods
On behalf of Danone Trading B.V. (Danone), ENVIRON International Corporation (ENVIRON) convened
a panel of experts (Expert Panel), qualified by their scientific training and experience to evaluate the
safety of food ingredients, to determine the safety and the suitability and the Generally Recognized As
Safe (GRAS) status of the proposed uses of long-chain inulin (lc-inulin) prepared from chicory-derived
inulin. The Expert Panel Members included Joseph F. Borzelleca, Ph.D. (Professor Emeritus, Virginia
Commonwealth University School of Medicine; David J.A. Jenkins, M.D., Ph.D., D.Sc. (Professor,
University of Toronto; Director, Risk Factor Modification Centre, St. Michaels Hospital); and Judith
K. Jones, M.D., Ph.D. (The Degge Group, Ltd.).
The Expert Panel, independently and collectively, critically evaluated the available information presented
in documents prepared by ENVIRON (the GRAS dossier) and other materials deemed appropriate or
necessary. This information included a history of use and its regulatory status, the chemical identity of
lc-inulin, product specifications (Danone purchases the lc-inulin from BENEO-Orafti S.A. and certain
manufacturing details remain confidential business information and were not provided), intended
conditions of use and use levels, anticipated exposures, and safety assessment.
Following its independent and collective critical evaluation of the available information, the Expert Panel,
convened on 19 December 2012 with representatives of ENVIRON. Following extensive discussion, the
Expert Panel unanimously agreed to the conclusions described herein. A summary of the basis for these
conclusions follows.

Description of Long-Chain Inulin (lc-inulin), Manufacturing Process, Product


Specifications
The substance in this GRAS determination is long-chain inulin (lc-inulin) derived from inulin extracted
from chicory roots (Cichorium intybus). Inulin is a naturally occurring polysaccharide that belongs to a
class of carbohydrates known as fructans. Inulin-type fructans are linear polydisperse carbohydrates
mainly composed of fructose units joined by a series of 2-1 fructosyl-fructose linkages.
BENEO-Orafti S.A. (Beneo) supplies the lc-inulin product to Danone under the trade name Orafti HP
(formerly known as Raftiline HP or Beneo HP). Greater than 99.5% of Orafti HP long-chain inulin is
carbohydrate while water comprises the remaining < 0.5%. In 1623 batches produced since 2005, the
mean concentration of inulin with a DP 5 was 99.5% of total carbohydrate (SD = 0.5%) with fructose,
glucose, and sucrose combined constituting a mean of 0.1% (SD = 0.2%) of total carbohydrates. The
Chemical Abstracts Service Registry Number (CASRN) for inulin is 9005-80-5; lc-inulin does not have a
distinct CASRN.
The food-grade specifications for lc-inulin to be used by Danone are presented in Table 1. All tested
batches met the established specifications, demonstrating that the product meets appropriate
specifications for food-grade material. Regular compositional analyses by Beneo revealed the following
substances to be below the maximum residue levels set in EU regulation EC149/2008: cadmium and
mercury (< 0.01 milligram (mg)/kilogram (kg)); sulfur dioxide (< 5 mg/kg), nitrate (< 50 mg/kg), nitrite
(< 0.5 mg/kg); aflatoxin B1 (< 1microgram (g)/kg), ochratoxin A (< 0.5 g/kg), zearalenone (< 10 g/kg),
ENVIRON International Corp. 1702 East Highland Avenue, Suite 412, Phoenix, AZ 85016
V +1 602.734.7700 F +1 602.734.7701
environcorp.com

000007

GRAS Status Consensus Statement


Danone Trading B.V.
desoxynivalenol (< 0.1 g/kg); and polychlorinated biphenyls, dioxin-like compounds, and pesticides to be
below the levels of detection. Analyses of six batches demonstrated that phthalates are below limits of
quantitation (< 0.2 mg/kg).
Beneo lc-inulin has a guaranteed shelf of three years when stored in its original packaging.

Proposed Uses and Estimated Daily Intakes


Danone intends to use long-chain inulin in (1) exempt and non-exempt infant formulas for term infants
beginning at birth and toddler formulas intended for use by children from12 through 35 months of age;
and (2) medical foods, including nutritionally incomplete oral products, nutritionally complete oral
products, tube feedings, and pediatric nutritionals. The lc-inulin would be added to infant and toddler
formulas and medical foods in combination with galacto-oligosaccharides (GOS), with GOS accounting
for 90% of the total added prebiotics and lc-inulin accounting for the remaining 10%. The intended use
categories, target populations, and lc-inulin use levels are described below and summarized in Table 2.
The maximum target level of lc-inulin in all infant formulas for term infants (non-exempt and exempt) is
0.8 grams (g) lc-inulin per liter (L) formula as consumed, for a total of 8.0 g GOS/lc-inulin per L. Toddler
formulas are those formulas that are considered suitable for children from 12 through 35 months of age.
The maximum target use of lc-inulin in toddler formulas is 1.2 g lc-inulin per L formula as consumed, for a
total of 12 g GOS/lc-inulin per L.
Table 3 provides estimated intakes of lc-inulin based on the proposed use in infant formula. The proposed
use of up to 8 g GOS+lc-inulin per L in term infant formulas and 12 g GOS+lc-inulin per L in toddler
th
formulas, both in a ratio of 9:1, will result in intakes of up to 9.9 g GOS+lc-inulin per day at the 90
percentile of intake; this value represents the estimated intake from formula by infants 0-5 months of age.
th
Older infants and toddlers are estimated to have 90 percentile intakes of GOS+lc-inulin of 8.9 and
7.0 g/d, respectively. Medical foods are expected to provide up to 25 g GOS+lc-inulin per 1000
kilocalories (kcal) from nutritionally incomplete oral medical foods, 20 g/1000 kcal from nutritionally
complete oral foods and tube feedings, and 12 g/L from enteral nutritional products for children.

Safety Assessment
GRAS notifications 44, 118, 236, 286, 334, and 392 have previously addressed safety and tolerance of
2-1 fructans (inulin, oligofructose or FOS substances).
Absorption, Distribution, Metabolism and Excretion
Inulin, oligofructose and FOS are largely resistant to hydrolysis in the human upper intestine because the
majority of glycosidic bonds in inulin and oligofructose are D-fructofuranosyl 2-1 links. No enzyme able
to split this glycosidic linkage is secreted by salivary glands, the pancreas or in the small intestine of
humans.
Acute, Subchronic, Long-term, Carcinogenicity, Reproductive and Developmental and
Mutagenicity Studies
The prior GRAS notifications provided results for 2-1 fructans feeding studies in rats and mice,
teratogenicity study in rats, a chronic bioassay in rats and in vitro genetic toxicity studies. The animal
studies demonstrated no adverse effects from consumption of the fructans and results from in vitro
genetic toxicity studies demonstrated that commercially available FOS lacks any significant genotoxic
potential.

28-26968C

Page 2 of 7

000008

GRAS Status Consensus Statement


Danone Trading B.V.
Human Studies
Infant formulas containing up to 8 g/L of a combination of 90% GOS and 10% lc-inulin are approved for
use in the European Union and the intended use of the GOS+lc-inulin mixture in infant formulas is
therefore the same as the 8 g per L of this mixture approved for formulas in Europe by the Scientific
1
Committee on Food .
In a 2008 report, Food Standards Australia New Zealand (FSANZ) approved the addition of inulin-derived
2
substance and GOS at a maximum of 8 g/L in infant formulas . FSANZ based this decision on the history
of safe use of these substances in the general food supply in Australia and New Zealand and the
available evidence for infant formulas.
Results from 43 studies of healthy, male and female term infants consuming formula supplemented with
up to 0.8 g lc-inulin and 7.2 g GOS per L for periods ranging from two weeks to 12 months provide
evidence that lc-inulin, in combination with GOS, was well tolerated by infants and no adverse effects
were reported. The supplemented formulas supported normal growth. Additionally, results from these
studies suggest that formulas with added lc-inulin in combination with GOS may influence shifts in infant
gut microflora that result in gut microflora more similar to the gut microflora of breast-fed infants.
Results from 13 clinical trials provide additional evidence that the addition of a GOS/lc-inulin mixture in a
9:1 ratio to infant formula for preterm infants was well tolerated for two to 12 week study periods. These
preterm infants were closely monitored throughout the studies, and no adverse effects were reported on
measures of growth, fluid balance, formula intake, or stool characteristics. The absence of reports of
adverse effects in this highly sensitive and closely monitored study population provides strong evidence
for the safety and suitability of GOS and lc-inulin in a 9:1 ratio in infant formulas.
The results of these clinical studies of 2-1 fructan intake in healthy populations and four studies of
chronically ill populations requiring medical foods demonstrates that the proposed typical uses of lc-inulin,
in combination with GOS are well tolerated, safe, and suitable for the target populations.
Prebiotics such as GOS and lc-inulin are minimally digested in the small intestine (less than 10%) and
exert their putative health effects primarily via fermentation in the colon with the production of short-chain
fatty acids which are absorbed along the length of the colon. Gastrointestinal function is not usually
adversely affected in most infants requiring metabolic formulas; therefore, tolerance of exempt metabolic
infant formulas containing added lc-inulin and GOS ingredients would be expected to be equivalent to
tolerance of comparable doses in standard formulas.
The weight of the available evidence supports the safety of Danones proposed uses of lc-inulin as a food
ingredient in term infant formulas, toddler formulas and medical foods. This GRAS determination of the
proposed uses of this ingredient at the target levels described herein is based upon scientific procedures
as described under 21 C.F.R. 170.30(b), and is corroborated by a history of safe use of chicory.

Scientific Committee on Food (SCF). European Commission: Health and Consumer Protection Directorate-General.
2003. Report of the Scientific Committee on Food on the revision of essential requirements of infant formulae and
follow-on formulae: Adopted April 4, 2003. Available at: http://ec.europa.eu/food/fs/sc/scf/out199_en.pdf. Accessed
February 22, 2012.

Food Standards Australia New Zealand (FSANZ). 2008. Final assessment report: proposal P306, addition of
inulin/FOS & GOS to food. July 16. http://www.foodstandards.gov.au/_srcfiles/P306FOS & GOS FAR FINALv2.pdf

28-26968C

Page 3 of 7

000009

GRAS Status Consensus Statement


Danone Trading B.V.
Table 1. Specifications for LC-Inulin
Parameter
Specification Method
Inulin (% CHO)
>99.5
HPLC
Inulin DP 5 (% CHO)
99.0
HPLC
Average DP
23.0
HPLC
Glucose, fructose, sucrose (%
0.5
HPLC
CHO)
Sulfated ash (% at 525C)
<0.2
ICUMSA GS3/4/7/8-11
Conductivity (S/cm at 28 Brix)
<250
ICUMSA GS2/3-17
Lead (mg/kg)
<0.02
ICP-AES
Arsenic (mg/kg)
<0.03
ICP-AES
Total mesophilic bacteria (cfu/g)
1000
ICUMSA GS2/3-41
Yeasts (cfu/g)
20
ICUMSA GS2/3-47
Molds (cfu/g)
20
ICUMSA GS2/3-47
Enterobacteriaceae (cfu/1 g)
Negative
ISO 21528-2
Staphylococcus aureus
Negative
ISO 6888
(cfu/0.1 g)
Salmonella spp. (cfu/250 g)
Negative
AOAC 2000.07
Abbreviations:
AOAC = Association of Official Analytical Chemists
cfu = colony-forming units
CHO = carbohydrates
HPLC = high performance liquid chromatography
ICUMSA = International Commission for Uniform Methods of Sugar Analysis
ICP-AES = inductively coupled plasma-atomic emission spectrometry
ISO = International Organization for Standardization
(The remainder of this page intentionally left blank.)

28-26968C

Page 4 of 7

000010

GRAS Status Consensus Statement


Danone Trading B.V.
Table 2. Intended Uses of GOS/lc-Inulin in Term Infant
and Toddler Formulas, and Medical Foods
Product Category

Intended Use

Maximum or [Typical]
total GOS/lc-inulin
a
Content

Term infant formulas and toddler


formulas

Non-exempt and exempt term


infant formulas

-Formulas for healthy term infants


beginning at birth and healthy term
infants with inborn errors of
metabolism, beginning at birth.

8 g/L

Toddler formulas

-Formulas for healthy children from 12


through 35 months of age.

12 g/L

Nutritionally incomplete oral


medical foods

The products are intended to


supplement the normal diet and
cannot be used as the sole source of
nutrition. Nutritionally incomplete
products may be either general
formulas or disease-specific formulas.

25 g/1000 kcal
[12.5 g/1000 kcal]

Nutritionally complete foods


Oral foods

-The products are intended to


supplement the normal diet but can
be used as the sole source of nutrition
when given in the recommended
amount. Nutritionally complete
products may be either general
formulas or disease-specific formulas.

20 g/1000 kcal
[10 g/1000 kcal]

Tube feedings

-Tube feeds are intended for enteral


nutritional support for those patients
with a functional or partially functional
gastro-intestinal tract who are unable
or unwilling to eat sufficient quantities
of conventional food to meet their
nutritional requirements. The products
are nutritionally complete and
intended as the sole source of
nutrition for the patients. Tube feeds
can either be standard formulas or
disease specific formulas.

20 g/1000 kcal
[10 g/1000 kcal]

Enteral nutritional products for


children (pediatric nutritionals)

-Children who need sole source


enteral feedings or supplemental
nutrition.

Medical foods

12 g/L

The ratio of GOS:lc-inulin is 9:1.


For certain disease-specific formulas with low energy densities (e.g., products for the dietary management of
metabolic disease), the maximum use of GOS/lc-inulin is 31.25 g per 1000 kcal.
c
The upper levels of GOS/lc-inulin intake may be well tolerated by some but not necessarily all people. A specific
dose of fiber may be achieved by using a fiber-containing medical food in combination of a fiber free version of
the same medical food.
Abbreviations: g = grams; GOS = galacto-oligosaccharide; kcal = kilocalories; L = liter; lc-inulin = long-chain inulin
b

28-26968C

Page 5 of 7

000011

GRAS Status Consensus Statement


Danone Trading B.V.

Table 3. Estimated 2-Day Average Intakes of Long-Chain Inulin from Infant Formula

Population

Infants, 0-5 mo
Infants, 6-11 mo
Toddlers, 12-35 mo

101
168
19

Percent
c
users

100
98.5
2.7

2-Day Average lc-inulin Intakes Per User


d
g lc-inulin/d
g lc-inulin/kg-bw/d
[g GOS/lc-inulin/d]
[g GOS/lc-inulin/kg-bw/d]
90th
90th
Mean
Percentile
Mean
Percentile
0.8

1.0

0.13

0.18

[7.5]

[9.9]

[1.3]

[1.8]

0.6

0.9

0.07

0.11

[6.4]

[8.9]

[0.7]

[1.1]

0.3

0.7

0.03

0.06

[3.3]
[7.0]
[0.3]
[0.6]
Breastfeeding infants and children were excluded from the sample population.
b
Number of people consuming infant formula during the study period.
c
Weighted percent.
d
Analysis of intake in terms of g lc-inulin/kg-bw/d was limited to infants with a measured body weight.
Abbreviations: bw = body weight; d = day; g = grams; GOS = galacto-oligosaccharide; kg = kilogram; lcinulin = long-chain inulin; mo = months.
a

(The remainder of this page intentionally left blank.)

28-26968C

Page 6 of 7

000012

GRAS Status Consensus Statement


Danone Trading B.V.

Conclusions
We, the members of the Expert Panel, have independently and collectively, critically evaluated the
available information on long-chain inulin prepared from chicory-derived inulin (presented in the dossier
prepared by ENVIRON and summarized above) and other information deemed appropriate and we
unanimously conclude that the proposed uses in infant formulas and toddler formulas and in medical
foods of long-chain inulin, manufactured consistent with current Good Manufacturing Practice (cGMP)
and meeting the food grade specifications presented in the dossier, are safe and suitable.
We further unanimously conclude that the proposed uses in infant formulas and toddler formulas and in
medical foods of long-chain inulin, manufactured consistent with current Good Manufacturing Practice
(cGMP) and meeting the food grade specifications presented in the dossier, are Generally Recognized As
Safe (GRAS) based on scientific procedures.
It is our opinion that other experts, qualified by scientific training and experience, and evaluating the same
data and information, would concur with these conclusions.

(b) (6)

Joseph F. Borzelleca, Ph.D.


Chair of the Expert Panel
Professor Emeritus
Pharmacology & Toxicology
School of Medicine,
Virginia Commonwealth University
Richmond, Virginia

Signature:

David J.A. Jenkins, M.D., Ph.D., D.Sc.


Professor
University of Toronto
Toronto, Ontario, Canada

Signature:

Date:

(b) (6)

Date:

(b) (6)
Judith K. Jones, M.D., Ph.D.
President and CEO
The Degge Group, Ltd.
Arlington, Virginia

28-26968C

Signature:

Date:

Page 7 of 7

~ ENVIRON
000013

Safety Evaluation of
Long-Chain Inulin
for Use in
Term Infant Formulas,
Toddler Formulas,
and Medical Foods

Prepared for:
Danone Trading B.V.
Schiphol, The Netherlands
Prepared by:
ENVIRON International Corporation
Phoenix, Arizona
Date:
April 30, 2013
Project Number:
28-26968B

000014

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Contents
Page
1

Executive Summary

2
2.1
2.1.1
2.1.2
2.1.3
2.1.4
2.1.5
2.2
2.2.1
2.2.2
2.3
2.3.1
2.3.2

Description of the Substance


Identity
Chemical Name and Identity
Common or Trade Names
CAS Registry Number
Physical and Chemical Properties
Chemical Structure of Long-Chain Inulin
Production Process
Source
Manufacturing Process
Product Specifications
Product Specifications and Batch Data
Product Stability

2
2
2
3
3
3
3
4
4
4
6
6
7

3
3.1
3.2
3.2.1
3.2.2
3.2.3
3.3
3.3.1
3.3.2
3.4
3.4.1
3.4.2

History of Use and Intended Uses


Dietary Exposure to Naturally Occurring Sources of Inulin
Dietary Exposure to Inulin Added to Food
Inulin Ingredients
Long-Chain Inulin Added to Infant Formulas
FOS Added to Enteral Formulas
Intended Uses of Long-Chain Inulin
Term Infant Formulas and Toddler Formulas
Medical Foods
Estimated Intakes of lc-Inulin from the Proposed Uses
Term Infant Formulas and Toddler Formulas
Medical Foods

8
8
8
8
10
10
10
11
12
15
15
16

Intended Effect

18

5
5.1
5.2
5.3
5.4
5.4.1
5.4.2
5.5
5.5.1
5.5.2
5.5.3
5.6
5.6.1
5.6.2
5.7
5.7.1
5.7.2
5.7.3
5.8

Safety Data
Introduction
Prebiotics and Inulin
Resistance of Inulin to Gastric Acidity, Hydrolysis and Absorption
Fermentation of lc-Inulin and Selective Stimulation of Intestinal Bacteria
In Vitro Evidence
In Vivo Evidence
Physiological and Systemic Effects of Inulin
Physiological Effects of Inulin in the Gastrointestinal Tract
Systemic Effects of Inulin
Potential Allergenicity of Inulin
Normal Development and Function of the Intestinal Microflora
Development of Microflora in the Infant
Functionality of Gut Microflora
Human Studies of Inulin and Oligofructose in Infant Formulas
Potential Role of Oligosaccharides in Infant Formula
Long-Chain Inulin and Galacto-Oligosaccharides in Term Infant Formulas and Weaning Foods
Water Balance
Long-Chain Inulin and Galacto-Oligosaccharides in Preterm Infant Formulas

19
19
20
20
22
22
24
26
26
27
28
33
33
34
35
35
36
67
68

Contents

000015

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

5.8.1
5.9
5.9.1
5.9.2

Human Studies of Inulin and Oligofructose in Preterm Infant Formulas


Use of Inulin and Oligofructose in Medical Foods
Potential Role of Fiber in Medical Foods
Tolerance of Inulin and Oligofructose in Medical Foods

68
76
76
76

6
6.1
6.2

Conclusions
Conclusion on the Use of Long-Chain Inulin in Infant Formula
Conclusion on the Use of Long-Chain Inulin in Medical Foods

86
86
86

References

87

List of Tables
Table 2-1.
Table 2-2.
Table 3-1.
Table 3-2.
Table 3-3.
Table 5-1.
Table 5-2.
Table 5-3.
Table 5-4.
Table 5-5.
Table 5-6.

Physical and Chemical Properties of Orafti HP LC-Inulin


Specifications and Batch Data for the Product: Orafti HP LC-Inulin
Enteral Formulas Containing FOS Available in the U.S.
Intended Uses of GOS/lc-Inulin in Term Infant and Toddler Formulas, and Medical Foods
Estimated 2-Day Average Intakes of Long-Chain Inulin from Infant Formula
In vitro Studies of Fermentation of Long-Chain Inulin
Human Studies of Long-Chain Inulin Effects on Microflora
Human Studies of Tolerance of Long-Chain Inulin
Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Studies of 90% GOS/10% Long-Chain Inulin in Preterm Infant Formula
Studies with FOS/Inulin/Oligofructose in Enteral Formulas

3
6
11
14
16
23
25
33
50
73
82

List of Figures
Figure 1. Chemical Structure of Long-Chain Inulin
Figure 2. Production Process of Orafti HP lc-inulin

4
5

List of Appendices
Appendix A.

Batch Data and Methods of Analysis for Orafti HP


Table A 1. Specifications and Annual Analyses of Product: Orafti HP lc-inulin

Appendix B.

GRAS Notifications for the Use of Inulin, Oligofructose and Fructooligosaccharides


as Food Ingredients

Appendix C.

References

Contents

ii

000016

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

1 Executive Summary
Danone Trading B.V. (Danone) intends to add long-chain inulin (lc-inulin) to term infant formulas
(non-exempt and exempt), toddler formulas and medical foods in combination with galactooligosaccharides (GOS) at a ratio of 9:1 (GOS:lc-inulin).
In 2007, ENVIRON prepared a safety assessment and dossier of supporting information for
Nutricias (now Danone) GOS:lc-inulin product, presented it to a panel of food safety experts,
and obtained a generally recognized as safe (GRAS) Consensus Statement for the expert panel
subsequent to the panels deliberations on the GRAS status of Nutricias GOS:lc-inulin
combination.
Danone now wishes to update the lc-inulin safety assessment and dossier in preparation for the
submission of a GRAS notification to the United States Food & Drug Administration (FDA).
To determine that term infant formulas, toddler formulas and medical foods containing lc-inulin
are safe and GRAS at the proposed use levels, Danone engaged ENVIRON International
Corporation (ENVIRON) to prepare an updated safety dossier for lc-inulin for these proposed
uses. ENVIRON collected available characterization and safety data, researched and
assembled the additional publicly available data (in particular literature and information obtained
since the previous 2007 safety dosser was prepared), and conducted a safety review for each of
the principal ingredients. This document presents a safety assessment for the use of lc-inulin
as an ingredient in term infant formulas, toddler formulas and medical foods.
Herein, ENVIRON presents and interprets the currently available chemical characterization and
safety data for the lc-inulin.
ENVIRON concludes that the suitability and safety of the proposed uses is supported by the
appropriate, publicly available, scientifically defensible safety data.
(The remainder of this page intentionally left blank.)

Executive Summary

000017

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

2 Description of the Substance


2.1 Identity
2.1.1 Chemical Name and Identity
The substance that is subject of the evaluation for the GRAS determination is long-chain inulin
(lc-inulin) derived from inulin extracted from chicory roots (Cichorium intybus). Inulin is a
naturally occurring polysaccharide that belongs to a class of carbohydrates known as fructans.
Inulin-type fructans are linear polydisperse carbohydrates mainly composed of fructose units
joined by a series of 2-1 fructosyl-fructose linkages (Roberfroid 2005). The chemical formula
of inulin, from which lc-inulin is derived, is C6H11O6(C6H10O5)nOH and the systemic name for all
fructans is -D-glucopyranoside-(1-2)--D-fructofuranosyl-[(1-2)--D-fructofuranosyl]n (notably,
the -D-glucopyranoside-(1-2) is not always present). The n, which represents the number of
fructose units, ranges from 2 to 60 with some longer chains possible in native (or polydispersed)
inulin.
The individual members of the fructan oligo- and polysaccharides may be distinguished by their
source, method of production, or degree of polymerization (DP; the number of fructose or
glucose residues in the chain). All fructan products contain molecules with a range of DPs.
Inulin and related substances are referred to by various terms, including inulin, oligofructose,
fructo-oligosaccharide (FOS), and short-chain (sc) or long-chain (lc) fructo-oligosaccharide. The
nomenclature is not used consistently in the published literature and these terms are used
interchangeably. The nomenclature from GRN 392 is used in this dossier. The following is a
summary of the nomenclature.
Fructooligosaccharide (FOS): A fructan compound with nearly all DPs of less than 10 that is
obtained by the partial enzymatic hydrolysis of inulin or by enzymatic synthesis of sucrose
(transfructosylation). This definition is consistent with that promulgated by the International
Union of Biochemistry-International Union of Pure and Applied Chemistry Joint Commission on
Biochemical Nomenclature and AOAC International (IUB-IUPAC 1982; Niness 1999).
Short-chain Fructooligosaccharide (scFOS): A fructan compound with nearly all DPs 5,
typically with more than 85% of the molecules with a DP equal to three or four and only
negligible occurrence of DP > 5. Short-chain FOS residues are linked in the (2-1)
configuration and are synthesized from sucrose. Nearly all molecules have a glucose endcap.
Oligofructose: A fructan compound with DP < 10, with more than 25% of the molecules having
DP 5 and less than 75% with DP 4, produced by the enzymatic hydrolysis of inulin.
Inulin: A naturally-occurring fructan with DPs ranging from 2 to 60 or slightly higher. Because a
large portion of the molecules in inulin have DPs of 10 or less, oligofructose is a component of
inulin.
Long-chain inulin (lc-inulin): Inulin processed to remove the shorter chain fractions leaving
essentially fructans with DPs from 10 and higher (e.g., OraftiHP long-chain inulin). Because its
composition is primarily of longer-chain fructans, lc-inulin is a polysaccharide.

Description of the Substance

000018

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Oligofructose-enriched inulin: Mixtures of oligofructose and lc-inulin.


The subject of this evaluation is lc-inulin. Lc-inulin is inulin which has been altered by applying
physical separation techniques to remove short chain molecules and leaving long chain fructans
with a DP of 10 and higher with an average DP of 23.

2.1.2 Common or Trade Names


BENEO-Orafti S.A. (Beneo) supplies the lc-inulin product under the trade name Orafti HP
(formerly known as Raftiline HP or Beneo HP). Beneo commonly refers to the product as
long-chain inulin or High Performance Inulin. Because nomenclature is not formally established
in the published literature, some authors refer to lc-inulin as long-chain fructooligosaccharide,
lcFOS or polyfructose.

2.1.3 CAS Registry Number


The Chemical Abstracts Service Registry Number (CASRN) for inulin is 9005-80-5; lc-inulin
does not have a distinct CASRN.

2.1.4 Physical and Chemical Properties


The general physical and chemical properties of Orafti HP lc-inulin are identified in Table 2-1.
OraftiHP consists essentially of carbohydrate and water (<3%). More than 99.5% of the
carbohydrates are lc-inulin and less than 0.5% mono- and disaccharides. In 1623 batches
produced since 2005, the mean concentration of inulin with a DP 5 was 99.5% of total
carbohydrate (SD = 0.5%) with fructose, glucose, and sucrose combined constituting a mean of
0.1% (SD = 0.2%) of total carbohydrates.
Table 4-1. Physical and Chemical Properties of Orafti HP LC-Inulin
Property
Appearance
Appearance in solution
Taste
Conductivity (28 Brix)
pH (30-50 Brix)
Solubility in water
Carbohydrate content
(of dry matter)
Source: Beneo.

Value
Colorless powder
Colorless
Neutral
<250 S/cm
5.0 7.0
<5 g/L
>99.5%

2.1.5 Chemical Structure of Long-Chain Inulin


The chemical structure of lc-inulin is identified in Figure 1. As shown in the figure, the number
of fructan units, n, may vary, but ranges from approximately 10 to 60 or more. Less than
1% of the chains are shorter than a DP of five. The molecular weight varies depending on the
DP, but ranges from approximately one to over 10 kilodaltons (kDa). Figure 1 depicts a terminal
glucose, however, this is occasionally not present. Figure 1 does not depict branched
molecules, which constitute less than two percent of the total molecules of Orafti HP lc-inulin.

Description of the Substance

000019

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

2.2

Production Process

Orafti HP lc-inulin is produced under International Food Standard and ISO 9001:2008
standards that includes a HAACP system that is regularly audited by certifying bodies.

Figure 1. Chemical Structure of Long-Chain Inulin


Source: Beneo.

2.2.1 Source
Orafti HP lc-inulin is prepared from chicory roots grown for Beneo by individually subcontracted farmers who receive free seeds from Beneo and grow only approved chicory
varieties using approved agronomic practices including the use of only permitted pesticides.
Chicory inulin has a DP range of two to more than 60 and the distribution is variable and
determined by factors such as climate, soil, harvest season, storage, and processing of the raw
inulin to remove sugars and salts.

2.2.2 Manufacturing Process


The manufacturing process of lc-inulin is depicted in Figure 2. The process involves three
phases. Phase I is extraction of raw inulin wherein the chicory roots are washed and sliced,
then extracted in hot water to yield raw juice. The raw juice contains inulin and other soluble
substances (pulp) as well as non-inulin substances (minerals, proteins, some sugars, and cellwall particles). The pulp is mechanically separated from the raw juice and used as animal feed
and the non-inulin substances are precipitated out using food-grade calcium hydroxide. The
precipicated substances are filtered out and the filtrate is evaporated to produce raw inulin.
In Phase II the raw inulin is crystallized by cooling to precipitate out the lower solubility longchain molecules. The solution is filtered and the dissolved molecules (salts, sugars and shorter
chain fructooligosaccharides) pass through the filter leaving the remaining precipitate. This
precipitate is raw HP.
In Phase III the raw HP is redissolved in water and then subjected to ultra-heat treatment to
reduce the microbial load followed by fine filtration. Adsorption with activated carbon may
optionally be used in addition to provide decolorization. The product is dried by evaporation to
reduce the water content to less than 0.5% as specified.

Description of the Substance

000020

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

All the production agents such as precipitation and pressing aids, defoaming agents, antiscaling compounds, flocculants, pH-regulators, biocides, filtration aids, and adsorbents are food
grade or have no food contact. All production aids and additives are approved for use under
regulations listed in parts 173, 182, or 184 of the Code of Federal Regulations except activated
carbon and diatomaceous earth; both are GRAS substances with long histories of safe use in
food processing.

Figure 2. Production Process of Orafti HP lc-inulin


Source: Beneo.

Description of the Substance

000021

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

2.3 Product Specifications


2.3.1 Product Specifications and Batch Data
Beneo has established for Orafti HP lc-inulin the product specifications displayed in Table 2-2
to ensure a consistent food-grade product.
Ten non-sequential batches of product were analyzed with regard to the chemical and
microbiological parameters listed in the specifications. As shown in Table A-1 (Appendix A), all
tested batches met the established specifications, demonstrating that the product meets
appropriate specifications for food-grade material and that a consistent product can be and is
produced.
In addition to the parameters included in the product specifications, Beneo regularly analyzes
raw inulin as well as OraftiHP long-chain inulin for possible contaminants, including the heavy
metals cadmium and mercury; sulfur dioxide, nitrate, and nitrite; polychlorinated biphenyls
(PCB) and dioxin-like compounds (DLC); the mycotoxins aflatoxin B1, ochratoxin A,
zearalenone, and desoxynivalenol; and ~450 pesticides for which maximum residue levels are
set in EU regulation EC149/2008. These analyses have found cadmium and mercury levels to
be < 0.01 mg/kg; sulfur dioxide < 5 mg/kg, nitrate < 50 mg/kg, nitrite < 0.5 mg/kg; aflatoxin B1
< 1g/kg, ochratoxin A < 0.5 g/kg, zearalenone < 10 g/kg, desoxynivalenol < 0.1 g/kg; and
PCBs, DLCs, and pesticides below the levels of detection.
Table 4-2. Specifications for Orafti HP LC-Inulin
Parameter
Inulin (% dry matter)
Inulin DP 5 (% dry matter)
Average DP
Glucose, fructose, sucrose
(% dry matter)

Specification
>99.5
99.0
23.0

HPLC
HPLC
HPLC

Method

0.5

HPLC

Sulfated ash (% dry matter)

<0.2

ICUMSA GS3/4/7/811
ICUMSA GS2/3-17
ICP-AES
ICP-AES
ICUMSA GS2/3-41
ICUMSA GS2/3-47
ICUMSA GS2/3-47
ISO 21528-2

Conductivity (S/cm)
<250
Lead (mg/kg)
<0.02
Arsenic (mg/kg)
<0.03
Total mesophilic bacteria (cfu/g)
1000
Yeasts (cfu/g)
20
Molds (cfu/g)
20
Enterobacteriaceae (cfu/1 g)
Negative
Staphylococcus aureus
Negative
ISO 6888
(cfu/0.1 g)
Salmonella spp. (cfu/250 g)
Negative
AOAC 2000.07
Abbreviations:
AOAC = Association of Official Analytical Chemists
cfu = colony-forming units
HPLC = high performance liquid chromatography
ICUMSA = International Commission for Uniform Methods of Sugar
Analysis
ICP-AES = inductively coupled plasma-atomic emission spectrometry
ISO = International Organization for Standardization

Description of the Substance

000022

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

2.3.2 Product Stability


Long-term shelf stability, thermal and acid stability have been assessed. Three batches of
product were stored in their original packaging for 4 years at 25C and 60% relative humidity.
No changes were observed other than slight water absorption, and the results confirmed the
guaranteed shelf of three years for Orafti HP lc-inulin in its original packaging.
Samples of Orafti HP lc-inulin dissolved in water at pH 5/5.5 and 3.5 were exposed to
conditions mimicking sterilization (115C for 15 minutes) and ultra-heat treatment (145C for
10 seconds). No hydrolysis occurred under either heat condition at the higher pH, but up to
15% hydrolysis occurred at pH 3.5.
Orafti HP lc-inulin was stored for a year under acidic conditions (pH 3.5 and pH 4.0) at
temperatures of 20C or 25C. At the higher pH, the product was completely stable for three
months but hydrolysis was evident at six months and by one year reached 15% at 20C and
25% at 25C. A significantly greater degree of hydrolysis was observed at pH 3.5about
5% after two months and 45% and 60% at one year at temperatures of 20C and 25C,
respectively.
(The remainder of this page intentionally left blank.)

Description of the Substance

000023

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

3 History of Use and Intended Uses


3.1

Dietary Exposure to Naturally Occurring Sources of Inulin

Inulin and oligofructose are naturally occurring components of more than 36,000 plant species
(Niness 1999). A variety of foods containing inulin and oligofructose are commonplace in
contemporary Western diets. These foods include artichokes, asparagus, bananas, garlic,
leeks, onions, and grains including wheat, rye and barley (Van Loo et al. 1995). Approximately
45% of the inulin in chicory root has a DP of 20 or more and approximately 24% has a DP of 10
to 20 (Van Loo et al. 1995). Chicory is indigenous to Europe and has been cultivated since the
16th century. The chicory root is best known for its use as a coffee substitute (Niness 1999).
American diets are estimated to provide an average of 2.6 g inulin and 2.5 g oligofructose per
day, with intakes of each carbohydrate ranging from approximately 1 to 4 g per day (Moshfegh
et al. 1999). These estimates indicate that the inulin consumed by Americans predominantly
has a DP of 20 or less, which is referred to as oligofructose. Major food sources of naturally
occurring inulin and oligofructose in American diets are wheat, which provides about 70% of
these components, and onions, which provide approximately 25% of the estimated intakes
(Moshfegh et al. 1999). Per capita, the daily estimated intake of inulin and oligofructose
combined in the typical European diet ranges from 3.2 to 11.3 g (Van Loo et al. 1995).

3.2 Dietary Exposure to Inulin Added to Food


3.2.1 Inulin Ingredients
Inulin and oligofructose are available in the marketplace for use as ingredients in food. These
ingredients can be used for either nutritional attributes or technological properties such as sugar
and fat replacement, non-digestible and fermentable fibers, reduced caloric value, enhanced
body and mouth feel, improved stability of foams and emulsion, synergy with sweeteners,
improved texture, moisture retention, crispness and expansion, and heat resistance (Franck
2002).
The use of inulin, oligofructose and FOS as an ingredient in a variety of foods has been
determined to be Generally Recognized As Safe (GRAS). The publicly available GRAS
determinations include GRAS Notifications (GRNs) 233, 236, 286, 334 and 392 and the
correspondence from the U.S. Food and Drug Administration (FDA) regarding the notifications
of GRAS status for inulin and FOS are presented in Appendix B.
In 1992, the use of chicory-derived inulin (trade name Raftiline) and oligofructose (trade name
Raftilose) produced by Orafti (now Beneo) was determined to be GRAS in a variety of foods
for the general population by Kolbye Associates. The intended use of both Raftiline and
Raftilose was as a dietary fiber supplement or as a partial replacer of fat or carbohydrates.
Typical use rates of Raftiline were identified as 3 to 10% (by weight) in most applications, and
approximately 40% in some chocolate preparations. The intended use rate of Raftilose was
identified as approximately 2 to 60% (by weight).
In 2002, Imperial Sensus (now Sensus America LLC) notified FDA that its use of inulin derived
from the root of the chicory plant was determined to be GRAS for use in 43 food categories,
including baby food (excluding infant formula), as a bulking agent. The DP of the Imperial

History of Use and Intended Uses

000024

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Sensus chicory-derived inulin is two to greater than 60. Based on these proposed uses,
Imperial Sensus estimated that dietary intake of inulin at the 90th percentile level would be
approximately 6 grams per day for infants less than one year of age, approximately 15 grams
per day for infants one year of age, and approximately 20 grams per day for the general
population (i.e., two years of age and older). Based on the information provided by Imperial
Sensus, as well as other information available to FDA, the agency had no questions regarding
Imperial Sensus' conclusion that inulin is GRAS under the intended conditions of use (FDA
GRN 118 response letter 2003, Appendix B).
GTC Nutrition Company (GTC) notified FDA in May, 2000 that its FOS was determined to be
GRAS for use in selected foods as a bulking agent. The FOS produced by GTC Nutrition
Company is manufactured from sucrose syrup; the action of the fungal enzyme fructofuranosidase on the syrup yields fructose chains (primarily 2-4 units) with a terminal
glucose. The proposed uses of FOS in foods included use in baby foods at levels ranging from
0.1 to 3.6%. Based upon the proposed uses, GTC estimated that dietary exposure to FOS from
its intended use as a bulking agent would range from approximately 3.1 to 12.8 grams per
person per day at the 90th percentile consumption level. Based on the information provided by
GTC Nutrition, as well as other information available to FDA, the agency had no questions at
that time regarding GTC Nutrition's conclusion that FOS is GRAS under the intended conditions
of use (FDA GRN 44 response letter 2000, Appendix B).
In January, 2007, GTC notified FDA that GTCs FOS product (DP 2-4) was determined to be
GRAS for foods in general, excluding meat and poultry products and infant formula, at levels up
to 20 grams per day in the general population and at levels of up to 4.2 grams per day for
infants less than one year of age. Examples of typical FOS use levels were provided for a
variety of foods, including infant foods, with typical FOS use levels ranging from 0.4 to 3.6%,
and toddler foods, with typical FOS use levels ranging from 0.8 to 6.7%. FDA informed GTC
Nutrition that the agency had no questions regarding the companys conclusion that FOS is
GRAS under the intended conditions of use (FDA GRN 44 response letter 2007, Appendix B).
In October of 2010, FDA notified Yakult Pharmaceutical Industry Co., Ltd. that FDA had no
questions regarding Yakults use of GOS in term infant formula at 7.2 grams per liter (FDA GRN
334 response letter 2010, Appendix B). Additionally, Pfizer Nutrition and BENEO-Orafti recently
notified FDA that an expert panel concluded that their oligofructose (DP 2-8) was determined to
be GRAS for up to 3 grams per liter in milk-based term infant formula powder and FDA had no
questions (FDA GRN 392 response letter 2012, Appendix B).
In countries other than the U.S., inulin is often legally classified as a food or food ingredient, and
not as an additive (Coussement 1999). Inulin is not listed as an accepted food additive in the
standard positive lists from the European Union (EU) or from Codex Alimentarius. EU
Regulation 1333/2008 explicitly lists inulin as a substance that is not an additive. Describing
inulin as a dietary fiber is consistent with the Codex Alimentarius definition of dietary fiber.
The Codex Alimentarius dietary fibers are:
carbohydrate polymers with ten or more monomeric units, which are not hydrolyzed by the
endogenous enzymes in the small intestine of humans and belong to the following categories:

History of Use and Intended Uses

000025

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Edible carbohydrate polymers naturally occurring in the food as consumed,

carbohydrate polymers, which have been obtained from food raw material by physical,
enzymatic or chemical means and which have been shown to have a physiological effect
of benefit to health as demonstrated by generally accepted scientific evidence to
competent authorities,

synthetic carbohydrate polymers which have been shown to have a physiological effect
of benefit to health as demonstrated by generally accepted scientific evidence to
competent authorities (Codex 2010).

3.2.2 Long-Chain Inulin Added to Infant Formulas


The European Commission, Scientific Committee on Food (SCF) has stated that non-digestible
carbohydrates may be added to infant formulas for technical reasons or if they provide a source
of fermentable substrates for the gut microflora (SCF 2003). The SCF stated that it had no
major concerns with the inclusion of a combination of 90% galacto-oligosaccharides (GOS) and
10% FOS in infant formulas and follow-on formulas, provided that their total content does not
exceed 8 g per L; this maximum concentration is equivalent to 7.2 g GOS and 0.8 g FOS per L
infant formula. The GOS component of GOS-FOS mixtures added to infant formulas in Europe
consists of molecules with relatively short chain lengths (i.e., less than 10 galactose units), while
the FOS component consists of molecules with long chain lengths and corresponds to lc-inulin.
In a 2008 report, Food Standards Australia New Zealand (FSANZ) approved the addition of
inulin-derived substance and GOS at a maximum of 8 g/L in infant formulas (FSANZ 2008).
FSANZ based this decision on the history of safe use of these substances in the general food
supply in Australia and New Zealand and the available evidence for infant formulas. To reach
their decision FSANZ reviewed a number of studies regarding the use of GOS and lc-inulin in
combination (9:1 ratios). These studies are included in this safety evaluation in section 5.7.2.

3.2.3 FOS Added to Enteral Formulas


A variety of enteral formulas containing FOS (DP 2-4) are marketed in the U.S. These products
are summarized in Table 3-1. As shown in the Table, the FOS content in these tube/sole
source nutrition products ranges from 2.5 to 6.7 g per 1000 kcal, while total fiber content ranges
from 2.5 to 21.1 g per 1000 kcal. These oral supplements provide 1 to 3 g FOS per serving,
and 1 to 4 g of total fiber.

3.3

Intended Uses of Long-Chain Inulin

Danone intends to use long-chain inulin in (1) exempt and non-exempt infant formulas for term
infants beginning at birth and toddler formulas intended for use by children from12 through 35
months of age; and (2) medical foods, including nutritionally incomplete oral products,
nutritionally complete oral products, tube feedings, and pediatric nutritionals. The lc-inulin would
be added to infant and toddler formulas and medical foods in combination with galactooligosaccharides (GOS), with GOS accounting for 90% of the total added prebiotics and lc-inulin
accounting for the remaining 10%. The intended use categories, target populations, and lcinulin use levels are described below and summarized in Table 3-2.

History of Use and Intended Uses

10

000026

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

3.3.1 Term Infant Formulas and Toddler Formulas


Infant formula is defined by the Federal Food, Drug, and Cosmetic Act (FFDCA) as a food
which purports to be or is represented for special dietary use solely as a food for infants by
reason of its simulation of human milk or its suitability as a complete or partial substitute for
human milk (FFDCA 201(z)).
Table 4-1. Enteral Formulas Containing FOS Available in the U.S.

Product

kcal/mL

FOS (g) per 1000 kcal

Total Dietary
Fiber per 1000
kcal

Glucerna Select

Tube/SoleSource

6.7

21.1

Jevity 1.2 Cal

1.2

6.7

15.0

Jevity 1.5 Cal

1.5

6.7

14.7

Nepro w/ carb steady

1.8

5.3

8.7

Optimental

5.0

5.0

Pediasure Enteral Formula with


Fiber and FOS

3.0

5.0

Perative

1.3

5.0

5.0

Pivot 1.5 Cal

1.5

5.0

5.0

Suplena

1.8

5.3

8.7

TwoCal

2.5

2.5

Vital jr

3.0

3.0
Total Dietary
Fiber per Unit
(g)

Unit Size

Calories

FOS (g) per


Unit

Ensure Fiber

8 fl oz.

250

1.0

2.8

Ensure Pudding

4 fl oz.

170

1.0

1.0

Glucerna Shake

8 fl oz.

220

1.0

2.8

Glucerna Weight Loss Shake

11 fl oz.

290

1.4

4.0

ProSure Shake

8 fl oz.

300

3.0

3.0

Product
Oral
Supplement
s

FOS used in all enteral formulas assumed to have DP of 2-4.


Abbrevations: fl oz = fluid ounce; FOS = fructooligosaccharide; g = grams; kcal = kilocalories; mL = milliliter.
Source: Ross Nutrition 2006.

Danone intends to use long-chain inulin in infant formulas for term infants beginning at birth,
including infant formulas specifically designed for infants with metabolic disorders such as
phenylketonuria, tyrosinaemia, maple syrup urine disease, or fatty acid oxidation disorders.
Although the incidence of any particular metabolic disorder is quite rare, approximately 1 in
1,500 individuals has some form of an inborn error of metabolism (Raghuveer et al. 2006).
Formulas specifically formulated for use by infants who have inborn errors of metabolism or low
birth weight, or who otherwise have unusual medical or dietary problems are referred to as
exempt infant formulas (21 CFR 107.3). Metabolic infant formulas and other products for infants
with special nutritional needs are formulated with a selected balance of amino acids that does

History of Use and Intended Uses

11

000027

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

not include one or more amino acids specific to the metabolic disorder. Metabolic infant
formulas are not intended to provide a sole source of nutrition for infants. Rather, infants with
metabolic disorders are fed a combination of standard infant formulas (or human milk) and
exempt infant formulas to meet their nutritional needs. The maximum target use of lc-inulin in
all infant formulas for term infants (non-exempt and exempt) is 0.8 g lc-inulin per L formula as
consumed, for a total of 8.0 g GOS/lc-inulin per L.
Toddler formulas are those formulas that are considered suitable for children from 12 through
35 months of age. The maximum target use of lc-inulin in toddler formulas is 1.2 g lc-inulin per
L formula as consumed, for a total of 12 g GOS/lc-inulin per L.

3.3.2 Medical Foods


Medical foods are used under physician supervision as supplemental or sole sources of nutrition
for individuals who are unable to ingest sufficient nutrition from conventional foods. Danone
intends to add GOS to both nutritionally incomplete and nutritionally complete medical foods,
including pediatric nutritionals.

3.3.2.1 Nutritionally Incomplete Oral Medical Foods


Nutritionally incomplete oral medical foods are products that are intended to supplement the
normal diet. They are not designed to provide all nutrient needs and cannot be used as the sole
source of nutrition. Nutritionally incomplete products may be either general formulas or diseasespecific formulas. General formulas provide high amounts of selected nutrients (often protein)
in a palatable format. The products are used to address malnutrition experienced by patients
suffering from anorexia, cachexia, sarcopenia, burns, hip fracture and other medical conditions,
as well as pre- and post-surgery.
Disease- specific formulas are adapted with respect to macro- and micronutrients to meet the
needs of a specific disease or condition. These products include those for the dietary
management of metabolic disease and also those used to address malnutrition experienced by
renal patients, and patients suffering from pressure ulcers, cystic fibrosis or chronic obstructive
pulmonary disease.
The typical use level of lc-inulin in nutritionally incomplete oral medical foods is approximately
1.25 g lc-inulin per 1000 kcal product. The maximum proposed use level of lc-inulin in these
medical foods is approximately 2.5 g lc-inulin per 1000 kcal product. Typical and maximum use
levels of the GOS/lc-inulin mixture are 12.5 and 25 g GOS/lc-inulin per 1000 kcal, respectively.
For certain disease-specific formulas with low energy densities, for example those used in the
dietary management of metabolic disease, the proposed maximum use level of GOS/lc-inulin
may be up to 25% higher than the use in other medical foods. In these products, the maximum
use of GOS/lc-inulin is 31.25 g per 1000 kcal product, which is equivalent to approximately 28 g
GOS and 3 g lc-inulin per 1000 kcal.

History of Use and Intended Uses

12

000028

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

3.3.2.2 Nutritionally Complete Oral Medical Foods


Nutritionally complete oral medical foods products are intended to supplement the normal diet
but can be used as the sole source of nutrition when given in the recommended amount.
Nutritionally complete products may be either general formulas or disease-specific formulas.
Standard formulas comply with the reference values for macro- and micronutrients for a healthy
population. The products are used to address malnutrition experienced by various patients
such as geriatric patients, patients recovering from illness or patients suffering from neurological
disorders (e.g. Parkinsons, Alzheimers), dysphagia, HIV/AIDS or other diseases affecting food
intake or requiring extra intake of energy and nutrients. Disease-specific formulas are adapted
with respect to macro- and micronutrients to meet the needs of a specific disease or condition.
The products are used to address malnutrition experienced by various patients such as cancer
patients and diabetic patients.
Tube feeds are intended for enteral nutritional support for patients with a functional or partially
functional gastrointestinal tract who are unable or unwilling to eat sufficient quantities of
conventional food to meet their nutritional requirements. The products are nutritionally complete
and intended as the sole source of nutrition for the patients. Tube feeds can either be standard
formulas or disease-specific formulas. Standard formulas comply with the reference values for
macro- and micronutrients for a healthy population. The products are used to supply nutrition to
a variety of patients including pediatric patients, patients with central nervous system disorders,
cerebrovascular disease, multiple sclerosis and respiratory diseases. Disease-specific formulas
are adapted with respect to macro- and micronutrients to meet the needs of a specific disease
or condition. The products are used to supply nutrition to various patients such as cancer
patients and diabetic patients.
Nutritionally complete products are intended to be used when conventional foods cannot be
taken and therefore should supply the same nutrients as conventional foods. The
recommended intake of dietary fiber in the U.S. is 14 g per 1000 kcal, which is equivalent to
21 to 38 g fiber per day for adults (IOM 2002). A diet providing 20-30 g of fiber and 15002500 kcal would provide up to 20 g fiber per 1000 kcal. The typical use level of lc-inulin in
nutritionally complete medical foods is approximately 1.0 g lc-inulin per 1000 kcal product, and
the maximum target use level is approximately 2.0 g lc-inulin per 1000 kcal product. The typical
and maximum use levels of the GOS/lc-inulin mixture in nutritional complete medical foods
therefore are 10 and 20 g GOS/lc-inulin per 1000 kcal, respectively.

3.3.2.3 Pediatric Nutritionals


Danone intends to add long-chain inulin to nutritionals for pediatric populations. These products
are designed for children who need sole source enteral feedings or supplemental nutrition.
Children approximately 1 to 13 years of age are the target population for these nutritional
products. The maximum target use level of lc-inulin in pediatric nutritionals is 1.2 g lc-inulin
per L formula.

History of Use and Intended Uses

13

000029

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 4-2. Intended Uses of GOS/LC-Inulin in


Term Infant and Toddler Formulas, and Medical Foods
Product Category

Intended Use

Maximum or [Typical]
total GOS/lc-inulin
a
Content

Term infant formulas and


toddler formulas
Non-exempt and exempt term
infant formulas

Toddler formulas
Medical foods

-Formulas for healthy term infants


beginning at birth and healthy term
infants with inborn errors of
metabolism, beginning at birth.
-Formulas for healthy children from 12
through 35 months of age.

8 g/L

12 g/L

Nutritionally incomplete oral


medical foods

The products are intended to


supplement the normal diet and
cannot be used as the sole source of
nutrition. Nutritionally incomplete
products may be either general
formulas or disease-specific formulas.

25 g/1000 kcal
[12.5 g/1000 kcal]

Nutritionally complete foods


Oral foods

-The products are intended to


supplement the normal diet but can
be used as the sole source of nutrition
when given in the recommended
amount. Nutritionally complete
products may be either general
formulas or disease-specific formulas.

20 g/1000 kcal
[10 g/1000 kcal]

Tube feedings

-Tube feeds are intended for enteral


nutritional support for those patients
with a functional or partially functional
gastro-intestinal tract who are unable
or unwilling to eat sufficient quantities
of conventional food to meet their
nutritional requirements. The products
are nutritionally complete and
intended as the sole source of
nutrition for the patients. Tube feeds
can either be standard formulas or
disease specific formulas.

20 g/1000 kcal
[10 g/1000 kcal]

Enteral nutritional products for


children (pediatric nutritionals)

-Children who need sole source


enteral feedings or supplemental
nutrition.

12 g/L

The ratio of GOS:lc-inulin is 9:1.


For certain disease-specific formulas with low energy densities (e.g., products for the dietary management of
metabolic disease), the maximum use of GOS/lc-inulin is 31.25 g per 1000 kcal.
c
The upper levels of GOS/lc-inulin intake may be well tolerated by some but not necessarily all people. A specific
dose of fiber may be achieved by using a fiber-containing medical food in combination of a fiber free version of
the same medical food.
Abbreviations: g = grams; GOS = galacto-oligosaccharide; kcal = kilocalories; L = liter; lc-inulin = long-chain inulin
b

History of Use and Intended Uses

14

000030

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

3.4 Estimated Intakes of lc-Inulin from the Proposed Uses


3.4.1 Term Infant Formulas and Toddler Formulas
Danone proposes to use up to 0.8 g lc-inulin/L in exempt and non-exempt infant formulas for
term infants, and up to 1.2 g lc-inulin/L in toddler formulas. Estimates of potential intakes of lcinulin resulting from these intended uses were calculated using food consumption data reported
in the United States Department of Health and Human Services 2003-2004 National Health and
Nutrition Examination Survey (NHANES). This NHANES data set provides nationally
representative nutrition and health data and prevalence estimates for nutrition and health status
measures in the United States (NCHS 2006).
As part of the examination, trained dietary interviewers collected detailed information on all
foods and beverages consumed by respondents in the previous 24 hour time period (midnight to
midnight). A second dietary recall was administered by telephone 3 to 10 days after the first
dietary interview, but not on the same day of the week as the first interview. A total of
9,043 respondents provided complete dietary intakes for the Day 1 recall, and 8,354 of the
individuals provided a complete Day 2 recall. Using the list of food codes and the NHANES
2003-2004 dietary recall data files from individuals with two complete days of dietary recall,
ENVIRON estimated mean and 90th percentile 2-day average intakes of lc-inulin from infant
and toddler formulas.
The food codes for infant formulas reported by NHANES respondents are identified by brand
name and form (e.g., prepared from powder or ready-to-feed). The majority of infant formulas
specified in the dietary recalls are non-exempt formulas for term infants. Intake of exempt
formulas such as protein hydrolysate formulas and formulas for preterm infants was reported for
some survey participants, though no intake of metabolic formulas was reported. All formulas
were included in the estimates of intake.
Infants with metabolic disorders are fed a combination of standard infant formulas (or human
milk) and exempt infant formulas to meet their nutritional needs. We therefore assumed that the
total amount of infant formula consumed by NHANES participants is representative of the total
amount of formula consumed by infants with metabolic disorders, and that the concentration of
lc-inulin is the same in all formulas. We assumed all formulas for infants contained 0.8 g
lc-inulin per L formula (as consumed), and we assumed toddler formulas contained 1.2 g
lc-inulin per L formula. The 2-day average intakes represent the total estimated intakes of
lc-inulin during the two days of recall divided by two (i.e., (IntakeDay 1 + IntakeDay 2)/2).
Intakes were calculated for three subpopulations of infants (0-5 months, 6-11 months, and
12-35 months). The estimates were generated using survey sample weights to adjust for
differences in representation of subpopulations; results therefore are representative of the
U.S. population.
Estimates of lc-inulin intake from infant formula based on the proposed uses of the ingredient
are shown in Table 3-3. The mean estimated intake of lc-inulin by infants 0-5 months old is
0.8 g lc-inulin/day and the mean lc-inulin intake by infants 6-11 months old is 0.6 g lc-inulin/day.
The estimated 90th percentile 2-day average intake of lc-inulin by infants 0-5 months old is 1.0 g
lc-inulin/kg-bw/day, and the estimated 90th percentile intake by infants 6-11 months of age is
0.9 g lc-inulin/kg-bw/day. A small number of infants consume formulas after the first year of life,

History of Use and Intended Uses

15

000031

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

and formula intakes are lower than intakes by infants 0-5 or 6-11 months of age. The estimated
mean and 90th percentile 2-day average intakes of lc-inulin by toddlers, the target consumers of
toddler formulas, are 0.3 and 0.7 g/day, respectively.
Table 4-3. Estimated 2-Day Average Intakes of Long-Chain Inulin from Infant Formula

Population

Infants, 0-5 mo
Infants, 6-11 mo
Toddlers, 12-35 mo

101
168
19

2-Day Average lc-inulin Intakes Per User


d
g lc-inulin/d
g lc-inulin/kg-bw/d
[g GOS/lc-inulin/d]
[g GOS/lc-inulin/kg-bw/d]
90th
90th
Mean
Percentile
Mean
Percentile

Percent
c
users

100.0
98.5
2.7

0.8

1.0

0.13

0.18

[7.5]

[9.9]

[1.3]

[1.8]

0.6

0.9

0.07

0.11

[6.4]

[8.9]

[0.7]

[1.1]

0.3

0.7

0.03

0.06

[3.3]

[7.0]

[0.3]

[0.6]

Breastfeeding infants and children were excluded from the sample population.
Number of people consuming infant formula during the study period.
c
Weighted percent.
d
Analysis of intake in terms of g lc-inulin/kg-bw/d was limited to infants with a measured body weight.
b

Abbreviations: bw = body weight; d = day; g = grams; GOS = galacto-oligosaccharide; kg = kilogram; lc-inulin = longchain inulin; mo = months.

The estimates of potential lc-inulin intake by infants were based on the survey population of
non-breastfeeding infants. The youngest infants (i.e., prior to introduction of weaning foods) in
the sample population therefore are presumably consuming only infant formula. Some infants,
however, consume a combination of human milk and infant formula. The intakes of lc-inulin by
infants consuming a combination of human milk and formula would likely be lower than the
estimates of lc-inulin intake based on the population of exclusively formula fed infants.

3.4.2 Medical Foods


The typical and maximum levels of lc-inulin proposed for use in nutritionally incomplete oral
medical foods are 1.25 and 2.5 g lc-inulin/1000 kcal, respectively. A patient consuming
1000 kcal from nutritionally incomplete medical foods containing the typical proposed use level
of lc-inulin would consume 1.25 g lc-inulin/day, for a total intake of 12.5 g of the GOS/lc-inulin
mixture. Intake based on the maximum proposed use levels of GOS and lc-inulin is estimated
to be 2.5 g lc-inulin, and 25 g of the GOS/lc-inulin mixture. However, these are likely
overestimates of intake because nutritionally incomplete oral medical foods are intended as
supplements rather than as the sole source of nutrition.
Disease-specific medical foods are used to supplement a patients diet with specific nutrients
(e.g., amino acids for the management of a metabolic disease) and typically provide only a
portion of total daily energy needs. For example, a disease-specific formula providing 200 kcal,
or 10% of the daily energy needs for a 2000 kcal diet, would provide 6.25 g GOS/lc-inulin. The

History of Use and Intended Uses

16

000032

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

total intake of GOS/lc-inulin from these disease-specific formulas would not exceed total intakes
of GOS/lc-inulin from other nutritionally incomplete medical foods.
Lc-inulin is proposed for use at a typical level of 1 g/1000 kcal in nutritionally complete oral
medical foods (both orally ingested foods and tube feeds), while the maximum use level is
2 g/1000 kcal. The typical and maximum proposed use levels of the GOS/lc-inulin mixture in
nutritional complete medical foods therefore are 10 and 20 g GOS/lc-inulin per 1000 kcal,
respectively. Assuming a total energy intake in the range of 1500-2500 kcal, the intake of
lc-inulin would be 1.5 to 2.5 g based on typical use levels, which corresponds to a total intake of
15 to 25 g GOS/lc-inulin.
Lc-inulin is proposed for use at a level of 1.2 g/L in enteral nutritional products for children who
need sole source enteral feedings or supplemental nutrition. Most pediatric nutritionals are
designed such that 1000 mL provides at least 100% of recommended dietary intake (RDI) levels
of protein, vitamins, and minerals (IOM 2006) for children 1 to 8 years of age, and that 1500 mL
provides nutrients at levels meeting or exceeding RDIs for children 9 to 13 years of age.
Pediatric nutritional products containing the proposed level of added lc-inulin would provide
1.2 g lc-inulin in 1000 mL, and 1.8 g lc-inulin in 1500 mL of product. If the enteral formula is
consumed as a sole source of nutritional needs, lc-inulin intake would be in the range of 1.2 to
1.8 g/day, and total GOS/lc-inulin intake would be 12 to 18 g/day. If the nutritional products are
consumed as supplements to the childs diet, intakes of GOS (and GOS/lc-inulin) would be
lower.
(The remainder of this page intentionally left blank.)

History of Use and Intended Uses

17

000033

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

4 Intended Effect
In accordance with 21 CFR 170.3(o)(20), the intended effect of adding long-chain inulin to term
infant formulas, toddler formulas and medical foods is as a nutrient necessary for the bodys
nutritional and metabolic processes.
(The remainder of this page intentionally left blank.)

Intended Effect

18

000034

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

5 Safety Data
5.1

Introduction

Inulin, oligofructose, and FOS are characterized by chains of fructose units connected by the
characteristic 2-1 linkage usually connected to a single glucose molecule. The safety of inulin
and related 2-1 fructans has been reviewed and several ingredients have been determined to
be Generally Recognized As Safe for use in foods (Appendix B).
Imperial Sensus notified the U.S. Food and Drug Administration of the GRAS status of chicoryderived inulin in 2002 (GRAS Notice No. GRN 000118). Based on the proposed uses, Imperial
Sensus estimated that dietary intake of inulin at the 90th percentile level would be
approximately 6 grams per day for infants less than one year of age, approximately 15 grams
per day for infants one year of age, and approximately 20 grams per day for the general
population (i.e., two years of age and older). The GRAS uses of chicory-derived inulin were
later revised to include addition of up to 1 g inulin per serving of baby food, excluding infant
formula. The DP in this chicory-derived inulin is from two to greater than 60. Beneo previously
determined that the use of inulin and oligofructose as an ingredient in selected foods is GRAS,
though FDA was not notified of this determination.
FOS (DP 2-4) manufactured from sucrose syrup (GTC Nutrition) was determined to be GRAS
for foods in general, excluding meat and poultry products and infant formula, at levels up to
20 grams per day in the general population and at levels of up to 4.2 grams per day for infants
less than one year of age. FDA was notified in 2000 of the GRAS status of FOS when used in
selected foods (GRAS Notice No. GRN 000044), and in 2007 GTC notified FDA of the GRAS
status of the expanded list of uses. Yakult Pharmaceutical Industry Co., Ltd. notified FDA that
the use of GOS in term infant formula at 7.2 grams per liter was GRAS (GRN 000334) and in
October of 2010, FDA responded that it had no questions regarding Yakults GRAS
determination. Pfizer Nutrition and Beneo recently notified FDA that an expert panel concluded
that their oligofructose (DP 2-8) was GRAS and FDA had no questions (GRN000392; May
2012).
Information on the gastrointestinal fate, animal toxicity, and human tolerance of inulin and
related 2-1 fructans was considered in making the safety evaluations of both chicory-derived
inulin and FOS. As detailed in the GRAS determinations and summarized by FDA
(Appendix B), the 2-1 linkage is largely resistant to digestion in the small intestine and arrives
intact in the colon where it is subject to fermentation. Fermentation by the microflora produces
gases and short chain fatty acids. The short-chain fatty acids are used locally as an energy
source by the resident flora, taken up systemically via the colonocytes and transported to the
liver for caloric utilization by the host, or excreted in the feces.
The GRAS determinations state that results from published animal studies including acute
studies in rats and mice, 6-week feeding studies in rats, a teratogenicity study in rats, and a
chronic bioassay in rats, support the safety determination. Results from in vitro genetic toxicity
studies demonstrated that commercially available FOS lacks any significant genotoxic potential.

Safety Data

19

000035

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Based on a review of studies of human tolerance of inulin and related 2-1 fructans, it was
concluded that adults could regularly consume 40 to 70 grams of inulin per day (preferably in
multiple doses throughout the day) without experiencing any significant adverse effects. The
estimated 90th percentile intake of chicory-derived inulin is approximately 20 g/day for the
general population, which is no more than half the amount shown to be well-tolerated
(Appendix B; FDA 2003).
In the evaluation of the safety of ingestion of long-chain inulin (lc-inulin) under its intended
conditions of use along with GOS, ENVIRON reviewed available information on the digestion,
fermentation and tolerance of this particular subset of 2-1 fructans. The safety review begins
with a description of prebiotics and the general criteria by which inulin has been identified as a
prebiotic. The introduction is followed by a discussion of the digestion of 2-1 fructans and a
review of in vitro and in vivo data regarding the fermentation of lc-inulin. Information on other
gastrointestinal and systemic effects of inulin consumption is then summarized, and data on the
tolerance of lc-inulin are presented. We then review the normal development and function of
the colonic microflora. Studies of infants consuming the proposed combination of 90% GOS
and 10% lc-inulin in infant formulas and weaning foods are reviewed and summarized, as are
studies of individuals consuming medical foods containing a mixture of 90% GOS and 10%
lc-inulin or medical foods containing 2-1 fructans. ENVIRON conducted searches for
information on these topics using Medline and the Toxicology and Medicine categories within
the DIALOG database. The terms used in the searches included inulin, fructooligosaccharide, oligofructose, and HP inulin. Information published from the year 2000 (the
year of the first GRAS Notification for inulin) to 2007 (the time of this safety review) was
included in the search. ENVIRON conducted additional searches using PubMed and Google
Scholar with the same terms to include information published up to mid-2012. Potentially
relevant articles were identified and reviewed; the findings are described below. To maintain
consistent terminology, ENVIRON used lc-inulin to describe lcFOS in the studies where test
substance information was adequately descriptive. If the test substance was inadequately
described to make a determination, then ENVIRON used the same terms as the study.

5.2

Prebiotics and Inulin

Inulin is considered a prebiotic based on its (1) resistance to gastric acidity, hydrolysis by
mammalian enzymes, and gastrointestinal absorption; (2) fermentation by intestinal microflora;
and (3) selective stimulation of the growth and/or activity of those intestinal bacteria that
contribute to health and well-being (Roberfroid 2007). Based on these criteria, inulin has been
determined to be a prebiotic. Galacto-oligosaccharides (GOS) also have been determined to be
a prebiotic.

5.3

Resistance of Inulin to Gastric Acidity, Hydrolysis and Absorption

The majority of glycosidic bonds in inulin and oligofructose are D-fructofuranosyl 2-1 links.
No enzyme able to split this glycosidic linkage is secreted by salivary glands, the pancreas or in
the gastric and small intestinal epithelium brush border in mammals. Inulin, oligofructose and
FOS therefore are largely resistant to hydrolysis in the human upper intestine (Cherbut 2002).
No studies in which the digestibility of lc-inulin was tested were identified; results from studies of
inulin, oligofructose and FOS, however, provide evidence about the digestibility of the polymer.

Safety Data

20

000036

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Results from studies in ileostomy patients provide evidence of the resistance of inulin and
oligofructose to hydrolysis. In a study of 10 patients with conventional ileostomies due to
ulcerative colitis, 88% (95% Confidence Interval, 76-100%) of a 17 g dose of pure inulin (derived
from chicory root with an average DP of 10) was recovered in the ileostomy effluent. Eight of
the original 10 patients were tested again and 89% (95% Confidence Interval, 64-114%) of a
17 g dose of oligofructose (average DP 4) was recovered in the ileostomy effluent (Ellegard et
al. 1997). In another study of ileostomy patients, patients were fed inulin (DP >10 for over 30%
of the chains) extracted from Jersusalem artichoke and recovery was measured in the ileal
effluent (Knudsen and Hessov 1995). Recovery of inulin in the effluent was not different
following ingestion of two separate doses; recovery was 86.4 4.4% and 87 3.3% from
10 and 30 g inulin doses, respectively. The fructose:glucose ratio of the ingested inulin was 4.1.
The fructose:glucose ratio of the recovered inulin in ileal effluent was 4.7 1.1% at an intake of
10 g inulin and 4.5 0.1% at an intake of 30 g inulin. The higher ratio of fructose:glucose
recovered in the effluent as compared to the ratio in the ingested inulin suggests that lowmolecular-weight components of inulin are more sensitive to hydrolysis than the higher
molecular weight components. The investigators speculated that the small loss of inulin during
passage through the small intestine was due to hydrolysis by either acids or enzymes or to
microbial degradation by the microflora colonizing the distal small intestine.
The passage of FOS (DP 2-4) into the colon in healthy adults was investigated using the direct
method of measuring the flow rate in the distal ileum (Molis et al. 1996). After an equilibration
phase, the six healthy subjects consumed 20.1 g FOS per day, given in three postprandial
doses. Results showed that an average of 89 8.3% of the ingested FOS was delivered to the
colon, and analysis of the constituent oligosaccharides in the ileal contents showed that most
unabsorbed FOS was in an intact unhydrolyzed form. Approximately 0.12% (24 12 mg) of the
ingested FOS dose was recovered in 24-hour urine samples, and approximately 1% of the FOS
that disappeared from the small intestine was recovered in urine. It was assumed that excretion
of FOS in the urine resulted from absorption of intact FOS in the small intestine, and that a
fraction of the ingested FOS was hydrolyzed during passage through the gastrointestinal tract
and then absorbed as glucose and fructose. Stool samples contained no FOS, indicating that
FOS that was not hydrolyzed in the upper gastrointestinal tract was completely fermented in the
colon.
Results from breath hydrogen studies in humans following ingestion of FOS or inulin provide
further evidence that the carbohydrates are not absorbed. Stone-Dorshow and Levitt (1987)
measured breath hydrogen excretion in 10 subjects consuming 5 g FOS (Neosugar, Meiji Seika
Co., Japan; DP 2-4) three times daily for a period of 12 days. Following the 12-day period of
FOS ingestion, breath hydrogen excretion after a 10 g dose of FOS was approximately 50%
higher as compared to the baseline measurement; the increase was not statistically significant.
Breath hydrogen excretion after the 10 g FOS dose was similar to excretion after ingestion of
10 g lactulose, which suggests near total malabsorption of FOS. Geboes et al (2003)
conducted breath hydrogen tests using oligofructose (Raftilose), inulin (Raftiline ST), or lc-inulin
(Raftiline HP). Following ingestion of 5 g lc-inulin, an increase in hydrogen excretion of 10 ppm
above baseline was seen in five of six study volunteers in one trial, and in 15 of 17 volunteers in
another trial; lc-inulin was determined to be a suitable substrate for measurement of orocaecal
transit time via the hydrogen breath test. In another study (Schneider et al. 2007), ingestion of

Safety Data

21

000037

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

5 g lc-inulin was found to cause a rise in breath hydrogen excretion in 27 of 29 volunteers, and
again was determined to be an appropriate substrate for measurement of small bowel transit
times.
In summary, results from studies in humans provide evidence that inulin, oligofructose and FOS
are largely undigested in the upper gastrointestinal tract and are not absorbed. Nearly all of the
ingested oligomers are delivered to the colon. Because inulin and its related 2-1 fructans are
largely non-digestible, they can be regarded as dietary fibers. In vivo data on the digestibility of
lc-inulin were not identified, though given that lc-inulin contains the same characteristic
glycosidic linkage as inulin with shorter chain lengths, it is reasonable to assume that lc-inulin
also is largely undigested and functions as a dietary fiber.

5.4

Fermentation of lc-Inulin and Selective Stimulation of Intestinal Bacteria

More than 90% of ingested inulin is estimated to arrive in the colon (Van Loo et al. 1999).
Non-digested carbohydrates that reach the colon intact contribute to the fermentable load.
Fermentation of non-digested carbohydrates in the colon produces short-chain fatty acids such
as acetate, propionate and butyrate; gases including hydrogen, carbon dioxide, sulfur and
methane; and organic acids such as lactate, succinate and pyruvate (Roberfroid et al. 1998).
The increased concentration of short-chain fatty acids and organic acids in turn lowers the
colonic pH.

5.4.1 In Vitro Evidence


In vitro studies provide evidence of the fermentation of inulin, oligofructose or FOS and selective
stimulation of bifidobacteria. These studies have been conducted using pure culture
fermentation or using batch or continuous culture systems of human feces. Roberfroid and
colleagues (1998) reviewed the fermentation of 2-1 fructans and determined that all are well
fermented. Studies of the in vitro fermentation of lc-inulin are summarized in Table 5-1 and
below.
Based on in vitro studies in minimal nutrition media, Biedrzycka and Bielecka (2004) reported
that highly purified inulin with a higher degree of polymerization (22 or 23) was less able to
support the growth of Bifidobacterium longum and B. animalis as compared to FOS,
oligofructose and inulin.
Corradini and colleagues (2004) demonstrated the degradation and fermentation of FOS
(DP 3-10) and lc-inulin (DP 25) using Bifidobacterium and fecal cultures. In two different
cultures, one of B. cuniculi MB280 and one of B. adolescentis ATCC151703, both FOS and lcinulin presented degradation patterns that were closely correlated with bifidobacterial growth of
both strains. In order to evaluate the consumption of the fructans, anaerobic fermentation
vessels were inoculated with fecal samples. Analysis suggested that the fermentation of FOS
and lc-inulin resulted in the complete consumption of these carbohydrates. Mean concentration
of bifidobacteria increased from a baseline level of 1.9 x 105 to 3.7 x 107 and 1.2 x 107 for FOS
and lc-inulin samples, respectively.
Using fecal batch cultures, Langlands and colleagues (2004) found a mixture of oligofructose
and lc-inulin (mixture DP 10) to significantly increase mean ( SEM) surface counts of

Safety Data

22

000038

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

bifidobacteria compared to a control culture (7.3 0.2 log10 CFU/slide vs. 6.6 0.2 log10
CFU/slide), respectively. Mean concentrations of acetate and butyrate were also significantly
higher compared to the control (200 13.9 log10 CFU/slide vs. 90 4.1 log10 CFU/slide for
acetate and 89 5.3 log10 CFU/slide vs. 42 2.0 log10 CFU/slide for butyrate), respectively.
Other bacteria and SCFA concentrations evaluated in the study were not significantly altered as
a result of FOS fermentation.
Perrin and colleagues (2002) compared the fermentation of FOS (92% DP 2-20), native FOS
(60% DP 2-20) and lc-inulin (70% DP > 20) and observed that the lc-inulin had a longer rate of
consumption whereas FOS (92% DP 2-20) was consumed most rapidly.
Rossi and colleagues tested FOS (DP 3-10), lc-inulin (DP 25) and a combination of the two
prebiotics in both pure and fecal batch cultures. The substrates were analyzed in 55 different
Bifidobacterium strains, representing 11 species. Whereas FOS was fermented by most
strains, only eight grew in the lc-inulin substrate. The investigators also observed a straindependent capability to degrade fructans of different lengths.
Overall, results from these in vitro studies of lc-inulin provide evidence that the polymer is
fermented by bifidobacteria and that the rate of fermentation of lc-inulin is slower as compared
to fermentation of shorter chain fructans.
Table 5-1. In vitro Studies of Fermentation of Long-Chain Inulin
Reference

Substrate
FOS (DP 2-4)
Oligofructose (DP 2-8)
Inulin (DP 9)
Inulin-EXL (DP 22)
Inulin HP (DP 23)
Raftilose Synergy:
Raftiline HP (DP 25)
Raftilose P95 (DP 3-10)
Chicory scFOS
(70% DP 2-10)
Chicory lc-inulin
(50% DP 10)
Raftiline HP &
oligofructose
(mixture DP 10)

Study Design
Evaluated growth of
three strains of
Bifidobacterium
longum and ten strains
of B. animalis
Bifidobacterium and
fecal culture
fermentation
Metabolism/breakdown
of substrates by three
strains of
Bifidobacterium

Perrin et al.
2002

scFOS (5% DP >20)


Native chain FOS
(30% DP >20)
lc-inulin (70% DP >70)

Metabolism/breakdown
of substrates by three
strains of
Bifidobacterium

Rossi et al. 2005

FOS (DP 3-10)


Made of: Raftilose
Synergy & Raftilose P95
Raftiline HP (DP 25)

Pure and fecal batch


cultures using 55
Bifidobacterium strains

Biedrzycka and
Bielecka 2004
Corradini et al.
2004

Durieux et al.
2001
Langlands et al.
2004

Batch culture
fermentation

Results
Highly polymerized and highly
purified inulins were not as well
fermented as FOS, oligofructose,
and inulin.
Increase in bifidobacteria
concentration and decrease in pH.
FOS substrates fully consumed by
all test strains.
Substrates increased
bifidobacteria and concentrations
of butyrate and acetate.
Substrates with lower DP
consumed first. Increased DP
resulted in increased residual
FOS and longer rate of
consumption.
Almost all strains grew on FOS
while only eight grew on inulin.
Butyrate predominantly produced
by inulin; acetate and lactate
predominantly produced by FOS.

Abbreviations:
DP = Degree of Polymerization; FOS = fructooligosaccharide; lc-inulin = long-chain inulin

Safety Data

23

000039

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

5.4.2 In Vivo Evidence


Studies in humans provide additional evidence of the fermentation of inulin, oligofructose and
FOS in the colon. Studies of the fermentation of inulin and related fructans have been reviewed
and results indicate that ingestion of the substances is associated with increased numbers of
potentially health promoting colonic bacteria such as bifidobacteria (Roberfroid 2007). The
effects of lc-inulin or combinations of lc-inulin and oligofructose on microbial populations are
summarized below and in Table 5-2. The effects of lc-inulin fermentation on laxation are
summarized later in the document along with tolerance of lc-inulin.
Tuohy and colleagues (2001) evaluated the effects of Raftiline HP (lc-inulin) ingestion on fecal
microflora composition using in situ hybridization (FISH) enumeration techniques. Ten healthy
volunteers (five male, five female, 20 55 years old) consumed 8 g maltodextrin per day for
14 days. Over the following 14 days, the volunteers consumed 8 g lc-inulin/d. Fecal counts
measured after the period of maltodextrin consumption were compared to counts after 7 and
14 days of lc-inulin consumption. Nine of the ten volunteers completed the trial (reason for
dropout not available). Mean fecal populations of Bifidobacterium spp. increased significantly
after seven days of lc-inulin consumption compared to the maltodextrin period (8.9 0.8 vs
8.8 0.9 log10 cells/g, mean standard deviation) and levels remained elevated following
another week of lc-inulin consumption (9.0 0.6 log10 cells/g; not significantly different from
placebo or week one levels). Individuals with lower initial fecal bifidobacterial counts were
observed to have an increased response to lc-inulin ingestion. Mean fecal populations of
Bacteroides spp. and lactobacilli/enterococci did not change during the lc-inulin feeding period,
though total bacteria increased from 10.2 0.2 to 10.4 0.3 log10 cells/g after one week of
lc-inulin intake. A statistically significant increase in fecal Clostridium spp. was measured after
14 days of lc-inulin consumption as compared to levels following maltodextrin consumption
(7.6 0.5 vs 7.2 0.7 log10 cells/g); the investigators determined the increase to be of limited
clinical value given the small magnitude of change. The investigators speculated that lc-inulin is
not completely fermented in the proximal region of the colon, and therefore more of the
nondigestible carbohydrate may be available for fermentation in the distal region.
Fecal bifidobacteria counts in adults consuming 10 g lc-inulin daily for one week were not
different from counts in the placebo group (Bouhnik et al. 2004) as assessed by microbiological
plating techniques. The lc-inulin ingestion had no effect on counts of total anaerobes,
lactobacilli, Bacteroides or enterobacteria.
In the study by Langlands et al. (2004), biopsy samples from the cecum and descending colon
were used to analyze mucosal gut microflora in response to diet. In the study, 14 adults were
administered a total of 15 g lc-inulin and oligofructose (7.5 g of each) daily for two weeks and
other group of 15 adults served as the control group. Total anaerobe counts were unaffected by
the addition of the prebiotic mixture to the diet. Changes in total Bacteroides numbers or
species distribution were also not observed. There was a significant increase in eubacteria and
lactobacilli in both the proximal and distal colon for the prebiotic group compared to the control
at the end of the study. Bifidobacteria concentrations significantly increased compared to the
control, but only in the distal colon.

Safety Data

24

000040

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

In summary, results from these studies indicate that daily intake of 8 g lc-inulin, 7.5 g lc-inulin
plus 7.5 g oligofructose or 9 g of a 9:1 combination of GOS/lc-inulin is associated with increased
populations of bifidobacteria in feces and the mucosa. No change in fecal bifidobacteria counts
was seen after one week of daily intake of 10 g lc-inulin. Ingestion of lc-inulin had little impact
on other microbial populations including bacteroides, lactobacilli/enterococci, coliforms and
clostridia.
Table 5-2. Human Studies of Long-Chain Inulin Effects on Microflora
Reference

Bouhnik et al.
2004

Langlands et al.
2004

Treatment
Population
n=8 per
treatment; mean
30 y

10 g lc-inulin/d
consumed in two
equal portions

Healthy
volunteers

Source: Raftiline
HP, Orafti

n=14; mean 59 y

7.5 g lc-inulin & 7.5


g oligofructose
consumed in three
equal portions

Subjects on
colonoscopy
waiting list

n=17; 23-48 7
Shadid et al 2007

Pregnant women,
third trimester of
pregnancy

n=9; 20-55 y
Tuohy et al. 2001
Healthy
volunteers

Dose

Treatment
Duration

1 wk

2 wk

Source: Raftiline HP
and Raftilose P95;
Orafti
0.9 g lc-inulin and
8.1 g GOS/d
consumed in three
equal portions
8 g lc-inulin/d
consumed in two
equal portions
Source: Raftiline
HP, Orafti

Approx 15
wk

14 d

Effects
-No change in fecal
bifidobacteria in lc-inulin
group.

-Higher levels of
eubacteria in proximal and
distal colon.
-Increase in bifidobacteria
in distal colon.
-No changes in total
anaerobes clostridia,
Bacteroides or coliforms.
-No changes in
proliferation indices.
-Increased percentage of
bifidobacteria in maternal
fecal samples.
-No effect on fecal
lactobacilli.
-Increase in fecal
bifidobacteria and total
bacteria counts after 7 d;
increase in fecal clostridia
after 14 d.
- No effect on fecal
bacteroides or
lactobacilli/enterococci.

Abbreviations:
d = day; g = grams; GOS = galacto-oligosaccharide; lc-inulin = long-chain inulin; y = year.

In contrast to the potentially protective effects on fecal microflora in animals and humans
consuming 2-fructans observed in many studies, a single group of investigators has reported
an increased risk for translocation of salmonella in inoculated rats consuming oligofructose
(Raftilose P95, Orafti) or lc-inulin (Frutafit, Sensus) in combination with a low-calcium diet (Ten
Bruggencate et al. 2005; Ten Bruggencate et al. 2004; Ten Bruggencate 2003; BoveeOudenhoven et al. 2003). Fecal water cytotoxicity, fecal mucin excretion and urinary excretion
of nitrate and nitrite were increased in animals consuming the 2-fructans and low calcium diets
(20 or 30 mmol CaHPO4 2H2O/kg diet). In animals consuming higher levels of calcium
(100 mmol CaHPO4 2H2O/kg diet), however, these effects were not observed

Safety Data

25

000041

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

(Ten Bruggencate et al. 2004). In one study (Ten Bruggencate et al. 2005), intestinal
permeability as measured by urinary excretion of CrEDTA was observed to decrease in animals
consuming a low-calcium diet and FOS.
The same investigators also examined measures of intestinal barrier function in 34 healthy men
consuming 20 g FOS (Ratilose P95, Orafti) in combination with diets containing approximately
300 mg calcium (Ten Bruggencate et al. 2006). In this placebo-controlled crossover study, FOS
had no effects on fecal water toxicity or intestinal permeability. Scholtens and colleagues
(2006a) measured fecal water cytotoxicity, alkaline phosphatase (ALP) activity as a marker of
epithelial cell turnover), and O-linked oligosaccharides (to estimate excretion of mucin-type
oligosaccharides) in 11 healthy adults consuming 25 or 30 g FOS (DP 2-7, Ratilose P95, Orafti)
in combination with their regular diets; ingestion of FOS had no effect on these measures.
Investigators also have reported that ingestion of inulin as part of a tube feed (Sobotka et al.
1997) or lc-inulin in combination with a normal diet (Hond et al. 2000) has no effect on intestinal
permeability. Colome and colleagues (2007) measured intestinal permeability in infants
72.5 30.52 days of age and compared measures by type of formula. The mean lactulose to
mannitol ratio (L/M) in infants consuming formulas supplemented with prebiotics (identified as
FOS and GOS) was not different from the L/M ratio in infants consuming other types of formula
(0.331 0.222 vs 0.299 0.183, respectively).
The effects of oligofructose or inulin on intestinal permeability and bacterial translocation in rats
reported by Ten Bruggencate appear to be related to the low calcium diets (Guarner 2007). The
available evidence in humans does not indicate that consumption of inulin or other fructans as
part of a balanced diet has any adverse effects on intestinal permeability, fecal water
cytotoxicity, fecal ALP activity, the fecal concentration of mucin-type oligosaccharides, or
presents an increased risk for bacterial translocation in humans.

5.5 Physiological and Systemic Effects of Inulin


5.5.1 Physiological Effects of Inulin in the Gastrointestinal Tract
Studies evaluating the ability of inulin and related 2-fructans to impact mineral absorption,
nitrogen balance and carcinogenicity in the intestinal tract have been conducted in both animal
and human models.
Numerous scientific reviews document studies analyzing the impact of 2-1 fructan intake on
mineral absorption (Coudray et al. 2003; Scholz-Ahrens et al. 2007; Weaver 2005). Several
investigators have demonstrated an increase in the gastrointestinal absorption of calcium and
magnesium in rats fed chicory inulin (Delzenne et al. 1995, Ohta et al. 1994, both as cited in
Kaur and Gupta 2002). Chicory long-chain inulin and oligofructose have also been found to
enhance femoral calcium content, bone mineral density, and calcium retention through
enhanced calcium absorption and suppressed bone turnover rates in animals (Weaver 2005).
Such findings suggest the potential for prebiotics to enhance bone mineral content and
structure. Studies in humans have been fewer and less compelling, as factors such as lifestage, diet and dosage of prebiotics make inter-study comparisons difficult and overall findings
inconclusive (Scholz-Ahrens et al. 2007).

Safety Data

26

000042

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Feeding studies have demonstrated variable effects on urea levels and nitrogen balance due to
the dietary intake of inulin. In one study, inulin enhanced fecal nitrogen excretion and reduced
renal excretion of nitrogen in rats (Younes et al. 1995, as cited in Kaur and Gupta 2002). In a
dietary supplementation study of 15% inulin, nitrogen balance was not statistically different from
control animals (Levrat et al. 1993, as cited in Kaur and Gupta 2002). It is difficult to extrapolate
animal findings to humans given the differences in digestive tract structures. One human study
did, however, find consumption of nondigestible carbohydrates to result in a higher fecal
excretion of nitrogen (Mortensen 1992, as cited in Kaur and Gupta 2002). Consequently,
dietary supplementation with fructans may produce moderate changes in nitrogen status. This
effect may be considered beneficial because of potential adverse health consequences of
excessive resorption of toxic ammonia from the large intestine.
Research on experimental animal models has revealed anticarcinogenic properties of inulintype fructans. In a systematic review of twelve studies, 21 of 24 treatment groups measuring
aberrant crypt foci (ACF) observed a significant reduction; 5 of 5 treatment groups measuring
tumors also observed significant reductions (Pool-Zobel 2005). In vitro studies utilizing human
cells have suggested possible mechanisms for these findings in animal studies including:
inhibited cell growth, modulated differentiation and reduced metastasis activities (Pool-Zobel
2005).

5.5.2 Systemic Effects of Inulin


Potential systemic effects of inulin and related fructans on glucose metabolism, blood lipid levels
and immunomodulation have been investigated.
The potential for inulin and other fructans to alter glucose utilization has been studied in animal
models as well as humans. In rat studies, both normal and streptozotocin-induced diabetic
animals consuming dietary oligofructose at dosages of 10% of the diet for 30 days and 20% for
two months, respectively, reduced postprandial glycemia (Oku et al. 1984, as cited in Kaur and
Gupta 2002). In healthy human volunteers, ingestion of 20 g/d fructans for four weeks did not
alter fasting plasma glucose or insulin, while diabetic subjects consuming 8 g/d fructan for
14 days demonstrated a decrease in fasting blood glucose levels (Luo et al. 1996, as cited in
Kaur and Gupta 2002). Overall, effects may depend on physiological and disease state.
Ingestion of 2-fructans has also been associated with effects on lipid metabolism. In a review
by Beylot (2005), studies demonstrating that feeding inulin to rats has significantly reduced
plasma levels of cholesterol and triacylglycerols were identified. The hypotriglyceridemic effect
observed is believed to be attributable to a reduction in liver lipogenesis; the decrease in plasma
cholesterol is understood to be due to the inhibition of cholesterol synthesis by propionic acid
and modifications in bile acid metabolism. Clinical studies present conflicting results on inulins
impact on human blood lipid levels. Lower doses used in human studies to avoid
gastrointestinal side effects may be responsible for the difference between animal and human
studies (Kaur and Gupta 2002). Individual human metabolic status and diet may also contribute
to observed discrepancies. Generally, there appears to be a trend in human studies for more
distinct effects on plasma cholesterol in subjects with hyperlipidemia (Beylot 2005).

Safety Data

27

000043

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Inulin may have immunomodulatory capabilities through its interaction with the gut-associated
lymphoid tissue (GALT) (Watzl et al. 2005). Although limitations in study design prevent the
formulation of conclusive evidence, animal studies have linked inulin to changes in phagocytic,
macrophage and natural killer cell activity (Watzl et al. 2005).

5.5.3 Potential Allergenicity of Inulin


5.5.3.1 Case Reports
Carbohydrates including polysaccharides such as inulin are not known to provoke food
allergies. There appear to be only two case reports of anaphylactic reactions to ingested inulin
and oligofructose described in the published literature, and two case reports of anaphylactic
reactions following the intravenous administration of sinistrin, a naturally occurring
polyfructosane which is isolated from red squill (the bulb of Urginea maritima).
In one case report (Gay-Crosier et al. 2000), a 39-year-old man had four episodes of
anaphylaxis after ingestion of salsify, artichoke leaves, and commercial products (margarine
and candy) containing added lc-inulin (Raftiline HP) or oligofructose (Raftilose P95). Local
wheal and flare reactions also were observed after the man touched artichokes. The patient
had a very strong reaction to lc-inulin (diameter of wheal, 12 by 20 mm) in a skin-prick test with
the substance, strong reactions to salsify, artichoke, oligofructose, and the products containing
added lc-inulin, and strong reactions to intradermally injected undiluted inulin (Inutest 25%; an
inulin product used for renal function tests) and oligofructose. In contrast, three control subjects
had no reactions to skin-prick tests with lc-inulin extract or to intradermal tests with (Inutest
25%) or oligofructose. The patient also experienced generalized urticaria, rhinoconjuctivitis, and
a 20% drop in the peak expiratory flow rate within ten minutes of ingesting 10 g lc-inulin in a
double-blind, placebo-controlled food challenge while the patient had no reaction following
ingestion of the placebo. However, an open oral challenge with 40 g oligofructose was
negative.
In another case report (Franck et al. 2005), a 50-year-old woman, with a past history of allergy
to artichokes, experienced generalized urticaria after eating a biscuit containing approximately
0.38 g lc-inulin (Raftiline HP). Approximately 1 years later, she had a severe prelethal
anaphylactic shock episode with loss of consciousness after eating yogurt containing
approximately 2.5 g lc-inulin (Raftiline HP). She had previously experienced several attacks of
urticaria after eating artichokes; however, daily intake of dairy products was well tolerated. Skin
prick tests were positive for raw and cooked artichoke (22 mm), salsify (6.5 mm), yogurt
containing 2% Raftiline (10 mm), though the prick test to Raftiline was negative. A leukocyte
histamine release test was negative to artichoke heart and weakly positive to inulin. In a
double-blind, placebo controlled food challenge test, the woman reacted to ingestion of 45 mL
yogurt, which contained approximately 0.9 g inulin, with erythema, urticaria, laryngeal pruritus,
and tachycardia without a drop in blood pressure. Results from the dot blot analysis showed
evidence of binding of specific IgEs to samples of yogurt with inulin, milk or BSA heated in the
presence of inulin, though there was no IgE binding to the control membrane, BSA, Raftiline, or
the nonheated milk and Raftiline mixture. The results suggest that specific IgEs were directed
to inulin-protein products formed by heating.

Safety Data

28

000044

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

A case report by Pirson et al. (2009) supports the protein hypothesis. The authors reported the
case of a 32-year old man who developed acute asthma and rhinoconjuncitivitis as a result of
occupational exposure to the dust from dry chicory roots. Skin prick test results were positive
for fresh and dry chicory and birch pollen, but negative for inulin. The authors identified
immunoglobulin (Ig) E sensitization as the allergic mechanism. Using immunoblotting and
specific IgE antibodies, the authors determined that the causative agent of the
rhinoconjunctivitis and asthma was a protein (a 17-kD Bet v 1 homologue) with a secondary oral
allergy syndrome after exposure to Bet v 1-related foods.
In a case report by Fux et al. (2004), a healthy, 25-year old male Chinese volunteer in a Phase I
clinical trial to assess glomerular filtration rate (GFR) with sinistrin (Inutest Fresenius Pharma
Austria, Graz) developed dypsnea, along with prickling and itching on the neck and face four
minutes after intravenous administration of 2 g sinistrin in 45 mL NaCL 0.9% solution with 100
mg sodium lactate. The subject had a positive skin prick test, indicating that the reaction was to
intravenous sinistrin. In a case report by Chandra and Barron (2002), a 45-year-old male
patient with Wegeners granulomatosis developed anaphylaxis and cardiorespiratory arrest after
administration of bolus intravenous sinistrin (2.5 g in 10 mL of solution). He was successfully
resuscitated and a skin prick test for sinistrin was positive with rapid wheal and flare. These
events appear to be extremely rare, as intravenous administration of sinistrin is the gold
standard method used to evaluate GFR, and there have only been two cases of adverse
allergic responses reported in the scientific literature.
Bacchetta et al. (2008a) reported the case of an 11-year-old boy who had been diagnosed with
IgA nephropathy (IgAN) two years prior and was administered an inulin infusion to measure his
GFR. Though the patient had a normal blood pressure and serum creatinine, he experienced
an anaphylactic reaction and a concomitant relapse of IgAN after administration of inulin (5 mL
Inutest 25% in 55 mL sodium chloride). After intravenous treatment with a steroid and an
antihistamine (dexchlorpheniramine) the symptoms disappeared. A skin prick test with inulin
was negative. After a second infusion (beginning with a 1/10 dose, then full dose) was
performed under dexchlorpheniramine, the patient experienced hypersensitivity symptoms and
oliguria, but not respiratory symptoms or arterial hypotension. Two days after the infusion the
patient suffered acute renal failure and relapse of IgAN. The authors speculated that the inulin
activated Toll-like transmembrane receptors and the innate immune system and, ultimately, the
infection response initiated the IgAN relapse.
In a follow-up study, Bacchetta et al. (2008b) evaluated the frequency of hypersensitivity to
inulin to determine if it is a rare response or underreported. The authors used data derived from
three nephrology units over two separate periods of approximately three years each. Of
57 cases of adverse reactions out of a total of 12,414 patients receiving GFR-related inulin
administrations, the authors obtained complete clinical data for 42 patients who experienced
hypersensitivity reaction. The authors observed no life-threatening complications. The authors
stated that because the use of inulin of food had increased during the study duration, they
should have observed an increased incidence of hypersensitivity. But the authors note that the
incidence between the two time periods was similar. Notably, the authors found that after some
patients experienced reactions to their first GFR inulin exposure, they did not experience
reactions to subsequent GFR inulin exposures as would be expected for an allergenic

Safety Data

29

000045

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

mechanism. The authors speculated that inulin may have activated Toll-like receptors and the
innate immune system. The authors concluded that hypersensitivity to inulin is rare, mostly
benign, and of an unknown mechanism.

5.5.3.2 Review of Allergenicity Findings


In response to the case reported by Gay-Crosier et al. (2000), Orafti reported that further testing
was conducted in an attempt to better understand the reported reaction of the male patient.
Orafti reported that in the same patient, no allergic reactions to foods containing oligofructose
were observed. In addition, evaluations of Raftiline products by SDS-PAGE or Coomassie Plus
Protein Assay did not detect any proteins, which are known to be associated with inducing
allergic responses, as opposed to polysaccharides.
Since 2003, Danone has maintained a registry of adverse events attributed to the use of their
products in European markets. Over a nearly three-year period beginning in November 2005
(the oldest available records), approximately 200 consumer complaints were registered under
the keyword allergic reactions. Danone modified its registry program though information from
2007 to 2012 is consistent with the earlier information. The majority of the complaints are in
regard to infant formulas or exempt infant formulas (powder products), a few are for liquid
products (toddler formulas), and none are concerned with medical foods. The complaints
regarding infant formula (non-exempt) primarily refer to disturbances in gastrointestinal function
while one reports heavier breathing and some report an allergic reaction or an allergic skin
reaction without additional details. The complaints regarding exempt infant formula include
reports of skin rash and wheezing, often in infants who had recently started using hydrolyzed
formula containing GOS/lc-inulin. Hydrolyzed formulas are for infants who are, or suspected to
be, intolerant to cows milk protein. For infant formulas based on hydrolysates claiming a
reduction of risk to allergy to milk proteins, European legislation requires the product label to
state that the product is not for infants allergic to the intact proteins from which it is
manufactured unless generally accepted clinical tests demonstrate tolerance in more than 90%
of infants (confidence interval 95%) hypersensitive to proteins from which the hydrolysate is
manufactured 1. In other words, up to 10% of infants starting on infant formula based on protein
hydrolysates may experience intolerance problems to the milk protein component of the
product. These infants are generally recommended to move to a semi-elemental product or, in
rare cases where problems persist, an amino acid based formula. Danone uses GOS/lc-inulin
in hydrolyzed formulas (both protein hydrolysates and semi-elemental products) but not in
amino acid based formulas since these formulas are free from ingredients of dairy origin.
Although it is not possible to detect whey protein in GOS, the manufacturing process of GOS
does not exclude the possibility that trace amounts of native whey protein may remain in the
ingredient. In hydrolyzed formulas, GOS is therefore subjected to the same enzymatic
hydrolysis step as the native cows milk protein used in the product.
It is important to note that the registry of adverse events are self-reported incidents with little
detail about the actual products ingested or other potential sources of allergens that the
individual may have been exposed to. In addition, no adverse effects suggestive of an allergic
1

Commission Directive 2006/141/EC; Official Journal of the European Union L 401/1, 30.12.2006; See Annex IV;
http://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:L:2006:401:0001:0001:EN:PDF

Safety Data

30

000046

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

reaction were observed in a double-blind, placebo controlled clinical study of 249 infants
suffering from atopic dermatitis (AD) (Moro et al., 2006). In fact, there were fewer reports of AD
infants in the group that received formula containing 0.8 g GOS/FOS per 100 ml (10/102 (9.8%)
than the control group that received maltodextrin-containing formula (24/104, 23%).
In summary, inulin-containing foods have been in the food supply for more than a decade.
During this time, two case reports of allergic reactions to inulin in foods in adults with artichoke
allergies have appeared in the publicly available literature. The available evidence indicates
that it is reasonable to conclude that the risk of allergic reactions to inulin in foods, including
infant formula and medical foods, is extremely small.

5.5.3.3 Human Tolerance of lc-Inulin and Effects on Laxation


Previous reviews of clinical studies of inulin and oligofructose ingestion have indicated that
humans can comfortably consume 20 g of the substances or more daily (Carabin and Flamm
1999; ENVIRON 2002). Potential adverse effects resulting from excessive intake are typically
gastrointestinal disturbances including bloating, flatulence, and abdominal distention. The
effects are generally mild in nature and reduced when the oligosaccharide is consumed in
divided doses throughout the day. Continued exposure to the oligosaccharide also tends to
result in reduced gastrointestinal symptoms as a consequence of adaptation. Longer chain
fructans are more slowly fermented than shorter chains (e.g., scFOS) and therefore tend to be
better tolerated. Fermentation of inulin and oligofructose in the colon also has been shown to
increase stool bulk, fecal water content, and frequency of defecation. Studies in which the
tolerance of lc-inulin and effects on laxation were assessed are summarized below and in Table
5-3.
Hond et al. (2000) studied the effects of the administration of lc-inulin (Raftiline HP) on healthy,
constipated adults in a double-blind, placebo controlled cross-over study. Six participants, one
male and five females, with a mean age of 28.5 years were enrolled in the study. None of the
participants had undergone abdominal surgery or had taken medication that could influence
intestinal motility or flora in the three months prior to study initiation. All participants went
through a washout period of one week at the initiation of the study. During week 2, subjects
were randomly assigned to consume lc-inulin (n=3) or sucrose, the placebo, (n=3). Week
3 consisted of another washout period. In week 4, participants switched treatments. Each
participant was administered 5 g lc-inulin or placebo, three times per day, for a total daily intake
of 15 g/d. By the end of the study, each participant had consumed lc-inulin for a period of one
week. Participants were instructed to consume a standard diet with the exclusion of any pre- or
probiotics. During the study weeks (lc-inulin and placebo administration), subjects maintained a
diary on fecal habits and gastrointestinal complaints. The mean number of stools significantly
increased after lc-inulin supplementation compared to baseline measurements (from 4.0 per
week at baseline to 6.5 per week after lc-inulin supplementation). For each individual, an
increase of at least one stool per week was observed. No significant increases in mean stool
weight, gastric emptying rate, orocecal transit time or total transit time were observed. During
the lc-inulin period, average complaints were made 0.8 and 1.2 times per week for flatulence
and abdominal cramps, respectively. No reports of flatulence or abdominal cramps were
reported for participants during the placebo period. This difference did not reach statistical
significance and researchers noted a large inter-individual variation for these gastrointestinal

Safety Data

31

000047

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

parameters. Intestinal permeability, expressed as the 6-hour urinary percent dose excretion of
[51Cr]EDTA also did not change after ingestion of lc-inulin.
Bouhnik and colleagues (2004) monitored the digestive symptoms in healthy volunteers
consuming one of seven types of nondigestible carbohydrates or a placebo (n = 8 per group).
The nondigestible carbohydrates provided to study participants included lc-inulin (Raftiline HP)
and FOS (53% DP 3). Participants consumed 10 g of each test substance daily in two divided
doses (5 g after lunch and dinner) for one week. Significant increases in excess flatus, bloating,
borborygmi and abdominal pain were reported by participants in each group, including the
placebo group, though there were no differences in gastrointestinal symptoms among all study
groups (Bouhnik et al. 2004). No significant change was observed in the number of daily stools
and diarrhea was not reported in any of the groups.
In a study by Langlands and colleagues (2004), 14 adult subjects consumed a mixture of 7.5 g
lc-inulin (Raftiline HP) and 7.5 g oligofructose (Raftilose P95) daily for two weeks; the mean
DP of the mixture was 10. Compliance was reportedly high, 97 percent in the treatment group,
and the majority of gastrointestinal complaints including flatulence and bloating were mild to
moderate in nature.
Results from these studies indicate that a total intake of 10 to 15 g of lc-inulin (or a combination
of lc-inulin and oligofructose) is well tolerated by adults. The noted side effects included
flatulence, bloating or abdominal pain. Daily intake of 15 g lc-inulin was associated with
increased stool frequency.
(The remainder of this page intentionally left blank.)

Safety Data

32

000048

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-3. Human Studies of Tolerance of Long-Chain Inulin

Reference

Bouhnik et
al. 2004

Treatment
Population

n=8; mean 30 y
Healthy volunteers

n=6; mean 28.5 y


Hond et al.
2000

Healthy,
constipated adults

n=14; mean 59 y
Langlands
et al. 2004

Subjects on
colonoscopy
waiting list

n=17; 23-48 y
Shadid et
al. 2007

Pregnant women,
third trimester of
pregnancy

Treatment
Duration

Dose

10 g lc-inulin/d
consumed in two equal
portions

1 wk

Source: Raftiline HP
(Orafti)
15 g lc-inulin/d
consumed in three
equal portions

1 wk

Source: Raftiline HP;


Orafti
7.5 g lc-inulin & 7.5 g
oligofructose
consumed in three
equal portions

2 wk

Source: Raftiline HP
and Raftilose P95;
Orafti
0.9 g lc-inulin and 8.1
g GOS/d
consumed in three
equal portions

Approx 15
wk

Effects
-No differences in digestive
symptoms among treatments;
increased symptoms reported
for all treatments.
-No effect on number of
stools; diarrhea not reported.
-Dropouts not reported in
study.
-Increase in fecal frequency.
-No effect on gastric emptying
rate, orocecal transit time or
total transit time.
-No difference in GI side
effects.
-No dropouts reported.
-Increase in reports of
flatulence, bloating and
laxative-effect; 20 of the 27
complaints mild to moderate;
7 reported as severe.
-Average compliance of 97%.
-No dropouts reported.
-No effects on bowel habits,
stool frequency or stool
consistency.
-No effect on stool or vaginal
pH.

Abbreviations:
d = day; g = grams; GOS = galacto-oligosaccharide; lc-inulin = long-chain inulin; wk = weeks; y = years.

5.6 Normal Development and Function of the Intestinal Microflora


5.6.1 Development of Microflora in the Infant
At birth, the intestinal tract of a human infant is sterile. The development of infant microflora
occurs in several stages: initial acquisition of microflora during birth and the first week of life,
development of microflora during feeding, and changes during weaning. Various factors
influence this development, including method of delivery (vaginal or Cesarean section), birth
environment (home or hospital), hygienic measures in place at the time of birth, developmental
stage at birth (preterm/term), type of infant feeding (breast-fed versus formula-fed), and the use
of antimicrobials (Heavey and Rowland 1999, as cited in Rodricks et al. 2007; Orrhage and
Nord 1999; Penders et al. 2006). In addition, following birth, the mother delivers additional
microbial strains to the infant during suckling, kissing, and caressing (Mackie et al. 1999).

Safety Data

33

000049

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Because of these various factors, the normal microflora of infants is variable. During vaginal
birth, an infant is colonized by bacteria from its mother (vaginal and feces microflora) and, to a
lesser extent, from the environment (Boehm et al. 2005; Dai and Walker 1999, as cited in
Rodricks et al. 2007; Hammerman et al. 2004, as cited in Rodricks et al. 2007; Mackie et al.
1999). Infants delivered via Cesarean-section (C-section) often have microflora dominated by
environmental/hospital isolates. Consequently, the gastrointestinal microflora of vaginallydelivered infants is quite different from infants delivered via C-section (Penders et al. 2006).
Infants born through C-section have lower numbers of bifidobacteria and Bacteroides, whereas
they are more often colonized with Clostridium difficile compared to vaginally-born infants
(Penders et al. 2006).
Over the first weeks of life, facultative anaerobic bacteria begin to predominate as the oxygen in
the gut is utilized and depleted (Heavey and Rowland 1999, as cited in Rodricks et al. 2007;
Orrhage and Nord 1999; Conway 1997; Wold and Adlerberth 2000, as cited in Rodricks et al.
2007; Fanaro et al. 2003; Penders et al. 2006). Anaerobic bacteria including various
bifidobacteria species and Bacteroides spp. are found as early as day 2 in the breast-fed
infants microflora, though peak counts are reached closer to day 7 (consistent with the
depletion of oxygen). Other anaerobic bacteria that appear during this change include
Clostridium spp., Eubacterium spp., and Lactobacillus spp. (Stark and Lee 1982; Lundequist et
al. 1985; Balmer and Wharton 1989, as cited in Rodricks et al. 2007; Harmsen et al. 2000;
Penders et al. 2006). Streptococci, Enterobacteria (including Escherichia coli), staphylococci,
and Enterococci have been identified as colonizing the infant gut one to two days after birth
(Conway 1997; Lundequist et al. 1985; Balmer and Wharton 1989, as cited in Rodricks et al.
2007; Stark and Lee 1982; Balmer et al. 1989).
An important determinant of gut microflora in infants is feeding; differences have been found
between the microflora of breast-fed infants and bottle-fed infants. The earliest studies of the
infant microflora showed that breast-fed infants had a microflora dominated by bifidobacteria,
which easily out-compete other genera and the presence of which are thought to depend on the
occurrence of certain glycoproteins in human breast milk (Cummings et al. 2004, as cited in
Rodricks et al. 2007). In contrast, formula-fed infants have a more complex flora which
resembles the adult gut in that bacteroides, clostridia, bifidobacteria, lactobacilli, gram positive
cocci, coliforms, and other groups are all represented in fairly equal proportions (Cummings et
al. 2004, as cited in Rodricks et al. 2007). Formula-fed infants have also been found to have
higher fecal levels of potentially harmful bacterial metabolic by-products (Edwards and Parrett
2002).

5.6.2 Functionality of Gut Microflora


The colonic microflora is generally considered fully developed and adult-like by two years of
age, although recent research has suggested that gut microflora continues to develop beyond
this age and changes throughout the life of an individual (Hopkins et al. 2002). The
gastrointestinal tract exists in symbiosis with a large number and variety of bacteria that
contribute to the health of the individual. Gut microorganisms contribute to diverse mammalian
processes. Along its entirety, the microflora of the gut acts as an effective barrier against
opportunistic and pathogenic microorganisms. Other advantageous effects include modulation
of the immune system, development of intestinal microvilli, production of short chain fatty acids

Safety Data

34

000050

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

(SCFA) upon which the colonic mucosa is dependent for energy, fermentation of non-digestible
dietary fiber and anaerobic metabolism of peptides and proteins resulting in the recovery of
metabolic energy for the host and removal of carcinogens and toxins (Rodricks et al. 2007).
Many factors including age and disease-state of an individual can alter gut microflora. For
example, elderly people tend to have fewer bifidobacteria and higher populations of
enterobacteria compared to younger adults (Hopkins et al. 2002). Moreover, elderly people with
Clostridium difficile associated diarrhea have been observed to have lower species diversity and
higher numbers of facultative anaerobes and low levels of bifidobacteria and bacteroides
compared to healthy elderly people (Hopkins et al. 2001). A number of physiological changes
occur in the body with advancing age. These include decreased acid secretion by the gastric
mucosa and greater permeability of mucosal membranes in the gut. Thus, it is likely that certain
bacterial strains take advantage of new ecological niches, thereby inducing a shift in the
composition of the gut microflora (Hopkins et al. 2002).
Shifts in microflora composition can have a direct impact on immune function. The intestine is a
major component of the immune system with the gut-associated lymphoid tissue (GALT)
comprising the largest mass of lymphoid tissue in the human body (Rodricks et al. 2007). The
lymphoid tissues produce immunoglobulin A (IgA). The secreted IgA is resistant to proteolysis
in the lumen of the intestine and does not activate complement or inflammatory responses.
Because IgA secreted by the GALT is a component of the common mucosal immune system,
immune responses mediated by GALT-derived IgA can affect immune responses at other
mucosal surfaces including the respiratory tract, salivary glands, and mammary glands (Isolauri
et al. 2001).

5.7 Human Studies of Inulin and Oligofructose in Infant Formulas


5.7.1 Potential Role of Oligosaccharides in Infant Formula
The oligosaccharides in human milk are complex and represent great diversity in their structure
(Boehm and Stahl 2007). To date, researchers have identified approximately 200 molecular
species of oligosaccharides in pooled samples of human milk (German et al. 2008). Unbound
oligosaccharides are present in mature human milk at levels ranging from approximately 5 to
10 g/L (Bode 2006), and therefore are a major solid constituent of human milk (Newburg 2000).
The oligosaccharides in human milk are believed to provide both local and systemic benefits
including prebiotic effects in the colon; antiadhesive and antimicrobial effects in the
gastrointestinal tract and urinary tract; modifications of the glycome of intestinal epithelial cells;
and immunomodulatory effects (Bode 2006). Human milk oligosaccharides are resistant to
digestion in the upper gastrointestinal tract and provide a fermentable substrate in the colon
(Engfer et al. 2000). German et al. (2008) note that because infants lack the enzymatic
capability to digest them, human milk oligosaccharides provide a carbon source for
bifidobacteria in the lower intestine. Results from in vitro studies provide evidence that select
species and strains of bifidobacteria are able to ferment human milk oligosaccharides and
therefore the oligosaccharides may function as prebiotics (Ward et al. 2006; 2007). In
comparison to human milk oligosaccharides, mature bovine milk contains very low levels of
oligosaccharides (Gopal and Gill 2000). Consequently, infants consuming bovine milk-derived
formulas lack a dietary source of oligosaccharides from formula or human milk and therefore
may be deprived of some of the potential benefits associated with their ingestion. Sherman et

Safety Data

35

000051

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

al. (2009) state that the initial bacterial colonization of the lower intestine is necessary for the
development of intestinal immune defenses and that prebiotics such as inulin and oligofructose
in infant formula may fulfill this same role as human milk oligosaccharides. A review by
Veereman et al. (2007) found that the bifidogenic capabilities of GOS/inulin mixtures is
established. Vandenplas et al. (2011) and Veereman et al. (2007) acknowledged that immune
mediated effects such as less fever, fewer intestinal and respiratory infections, and less atopic
dermatitis have been demonstrated but are less well established than the bifidogenic effects.
Oligosaccharides derived from plants, such as inulin and oligofructose, provide a potential
alternative source of non-digestible carbohydrates for infants who are not exposed to human
milk. No studies were identified in the published literature in which lc-inulin or inulin alone was
added to infant formula. Numerous studies have been conducted to assess the effects of a
combination of galacto-oligosaccharides (GOS) and lc-inulin on measures of growth, tolerance
and microbiological endpoints in populations of term and preterm infants. In all of the identified
studies, GOS and lc-inulin were provided in a ratio of 9:1. This ratio of GOS to lc-inulin is
designed to mimic the size distribution of neutral oligosaccharides in human milk (Boehm et al.
2003).

5.7.2 Long-Chain Inulin and Galacto-Oligosaccharides in Term Infant Formulas


and Weaning Foods
The effects of infant formulas containing added non-digestible carbohydrates comprised of 90%
GOS and 10% lc-inulin (lcFOS) on various measures of suitability in populations of term infants
were assessed in 43 clinical trials. The GOS/lc-inulin-supplemented formulas were
administered for time periods ranging from two weeks to up to twelve months, at dosages of
4 g/L (0.4 g lc-inulin and 3.6 g GOS per L) to 8 g/L (0.8 g lc-inulin and 7.2 g GOS per L). The
GOS/lc-inulin ingredient also was added to weaning foods. These studies are summarized in
Table 5-4 and briefly described below.
Knol and colleagues (2005a) assessed the effects of infant formula containing 0.8 g lc-inulin per
L and 7.2 g GOS per L on fecal microflora over a period of 6 weeks. Infants were 7-8 weeks of
age at study enrollment. Twenty-four infants were randomized to the GOS/lc-inulinsupplemented formula group, and 23 infants were randomized to a standard control formula
group. A total of 15 and 19 infants in the GOS/lc-inulin and control groups completed the study,
respectively. There was no difference in dropouts between the two groups. Symptoms of
flatulence and posseting were reported to be mild in all study groups during the study. The
percent of fecal bifidobacteria was higher in the GOS/lc-inulin -supplemented group as
compared to the control group. Infants in the GOS/lc-inulin -group were found to have lower
fecal pH, and increased proportion of fecal acetate and a decreased proportion of propionate.
Haarman and Knol (2005, 2006) subsequently identified and quantified the species of
bifidobacteria and lactobacillus present in fecal samples of 10 infants randomly selected from
infants consuming the GOS-supplemented formula, the standard formula and a breast-fed
reference group. The investigators found B. infantis, B. breve and B. longum to be the
predominant bifidobacteria in both the GOS/lc-inulin and breast-fed groups while L. acidophilus,
L. paracasei and L. casei were the predominant lactobacilli in the two respective groups. The
standard formula group tended to result in an adult-like microbiota, with lower levels of B. breve
and L. paracasei and relatively higher levels of B. catenulatum, B. adolescentis and

Safety Data

36

000052

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

L. debueckii. At the end of the 6-week study, the investigators also noted that bifidobacteria and
lactobacilli accounted for 80% of fecal microflora in the breast-fed and GOS/lc-inulin groups
while lactobacilli and bifidobacteria accounted for 50% of fecal microflora in infants fed the
standard formula.
Moro et al. (2002) studied the effects of consumption of a formula supplemented with GOS and
lc-inulin on infant microflora. Ninety term infants between 6.3 to 7.2 days old were randomly
assigned to one of three formula groups: standard formula (n=33), formula supplemented with
0.4 g lc-inulin per L and 3.6 g GOS per L (n=30), or formula with 0.8 g lc-inulin per L and 7.2 g
GOS per L (n=33). Infants consumed the assigned formula for four weeks. A dose-dependent
increase in fecal bifidobacteria was observed in the GOS/ lc-inulin groups, with baseline levels
of 8.5 CFU/g and 7.7 CFU /g, respectively, and levels of 9.3 CFU /g and 9.7 CFU /g,
respectively by the end of the study period. Fecal lactobacilli increased in the GOS/ lc-inulin
groups as compared to the control group, but there was not a difference between the two GOS
groups. A dose-dependent decrease in stool consistency was found; while stool frequency
increased and fecal pH decreased only in infants consuming formula supplemented with 0.8 g
lc-inulin per L and 7.2 g GOS per L. Weight gain and length increment were similar among
groups, and the diets did not influence the incidence of crying, regurgitation or vomiting, and no
infant had diarrhea during the study period. Fecal samples from infants in the high-dose group
(0.8 g lc-inulin per L and 7.2 g GOS per L) and the control groups were examined for the
presence of GOS and lc-inulin (Moro et al. 2005). The oligosaccharides were detected in stool
samples from all infants consuming the GOS/ lc-inulin -supplemented formula and none of the
infants in the control group. The amounts of GOS and lc-inulin detected in the feces were not
quantified. The investigators noted that human milk oligosaccharides also can be detected in
feces of infants consuming human milk.
Schmelzle and colleagues (2003) studied the effects of a formula containing partially hydrolyzed
protein, a high level of -palmitic acid and nondigestible oligosaccharides (GOS/lc-inulin)
versus a standard formula in a population of healthy, term infants. Infants consumed the
assigned formula from or before age 2 weeks until the age of 12 weeks. There were no
differences between groups in the number of dropouts or reasons for dropping out. Infants
consuming the new formula with added GOS/lc-inulin were found to have increased fecal
bifidobacteria compared to baseline levels, a higher percentage of fecal bifidobacteria as
compared to infants in the control group, and a higher proportion of softer stools. Formula
intake and energy intake per kg bodyweight were lower in infants consuming the supplemented
formula versus the control formula, though there were no differences at 12 weeks between
groups in weight gain.
In another study involving a formula containing hydrolyzed protein, -palmitate and GOS/lcinulin, 14 infants consumed the test formula from birth to 2 months of age (Rigo et al. 2001).
Infants consuming breast milk or standard formulas in previous studies were used as reference
controls. Growth and anthropomorphic development of the infants followed a standard healthy
infant growth curved throughout the study. Infants consuming the new formula had a higher
percentage of endogenous bifidobacteria compared to baseline levels when measured at 25,
45, and 68 days of age. One dropout was reported in the study due to gastro-esophageal
reflux.

Safety Data

37

000053

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Bakker-Zierikzee et al. (2005) studied the effects of a GOS/lc-inulin-supplemented infant


formula versus a probiotic formula containing Bifidobacterium animalis on the microflora of
healthy infants over the first 16 weeks of life. Within three days post-partum, 19 infants were
enrolled in each of the three formula groups (0.6 g lc-inulin per L and 5.4 g GOS per L, 6 x 1010
CFU B animalis per L, or control). Sixty-three breast-fed infants were followed as a reference
group. Infants consuming the GOS/lc-inulin supplemented formula were found to have a lower
fecal pH, higher percentages of fecal acetate and lower percentages of propionate, butyrate and
isobutyrate, isovalerate and valerate compared with infants consuming the control formula. No
differences among groups were found in the percentage of fecal bifidobacteria. During the
intervention, fecal sIgA levels were higher in infants consuming the GOS/lc-inulin supplemented
formula as compared to the control formula (Bakker-Zierikzee et al. 2006). All formulas were
reportedly well accepted and tolerated.
Savino et al. (2003, 2006) conducted two studies on older infants experiencing minor
gastrointestinal problems including colic. The first study was an observational study in which
604 infants with an average age of 1.35 months consumed a partially hydrolyzed formula
containing GOS/lc-inulin (0.8 g lc-inulin per L and 7.2 g GOS per L), palmitic acid in the
position and low levels of lactose for a period of two weeks (Savino et al. 2003). At the end of
the feeding period, infants experienced fewer episodes of colic, fewer regurgitation problems,
and less constipation. Reductions in the incidence of crying in infants with colic also were
observed in a randomized, controlled trial of infants consuming either a formula with partially
hydrolyzed whey proteins, GOS/lc-inulin (0.8 g lc-inulin per L and 7.2 g GOS per L) and a high
-palmitic acid level or a standard formula and simethicone (Savino et al. 2006).
Rinne et al. (2005) conducted a trial that evaluated microflora of infants at 6 months of age with
a family history of atopic eczema, allergic rhinitis or asthma. Inclusion criteria required infants to
have been on their respective formulas since at least 2 months of age. Of the 32 infants in the
study, eight consumed the GOS/lc-inulin formula and the remaining infants consumed a
standard formula, breast-milk or a probiotic-supplemented formula. Infants consuming the
GOS/lc-inulin-supplemented formula had increased fecal bifidobacteria after four months, and a
lower occurrence of atopic eczema compared to the standard formula group at 10 months.
Weight and length of infants were comparable among all formula groups.
Moro and colleagues (2006) also tested GOS/lc-inulin formula on infants at high risk of
developing atopy. Inclusion criteria required infants to have started formula feeding within the
first two weeks of life. Study visits were scheduled for each infant at 3 and 6 months of age. Of
the 206 infants completing the study, 102 were in the GOS/lc-inulin group with the remaining in
a maltodextrin placebo group. In a subset of 98 of the infants, 50 coming from the GOS/lc-inulin
group, fecal samples were analyzed. Infants consuming the GOS/lc-inulin formula were found
to have increased fecal bifidobacteria levels at 3 and 6 months of age as well as an increase in
stool frequency and softer stool consistency. Infants in the treatment group also had a
significantly lower incidence of dermatitis. Reasons for dropouts were similar between groups,
with reports of regurgitation and crying significantly lower in the GOS/lc-inulin group.
Alliet and colleagues (2007) studied the impact of a GOS/lc-inulin formula on serum cholesterol
and triacylglycerol levels in 8-week old infants. The treatment group included 75 infants

Safety Data

38

000054

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

followed until 26 weeks of age, for a total treatment period of 18 weeks. Since infants were not
restricted from breastfeeding, a sub-group of infants that consumed formula for 97.5% of their
total number of feedings (the number of infants in this sub-group was 22) was also followed.
Infants in the treatment group consumed 5.4 g/L GOS and 0.6 g/L lc-inulin. They were
compared to a control formula group (that also had a sub-group of high-compliance infants) and
an exclusively breast-fed group. For analytical purposes, both treatment formula and standard
formula groups were analyzed both as an entire group and with just the high-compliance infants
with the breast-fed group. Lipid levels were measured at 8 and 26 weeks of age and compared
between groups. The investigators observed no difference in median HDL or triacylglycerol
levels among groups at either of the time points measured in each. Median cholesterol and LDL
levels were significantly lower for both the entire and sub-group of infants in both formula groups
compared to the breast-fed infants. The median cholesterol levels for the entire GOS/lc-inulin
group and the entire control group at 26 weeks of age were and 130.00 and 121.50 mg/dL,
respectively while breast-fed infants had a median cholesterol level of 146.00 mg/dL at the
same time point. Median LDL levels for the entire treatment and control groups were 70.00 and
66.00 mg/dL compared to 86.00 for the breast-fed group. Of the 215 infants recruited,
187 completed the study.
One infant study was identified that involved the administration of GOS/lc-inulin as a solid
weaning food (Scholtens et al. 2006b). This randomized, double-blind, placebo-controlled trial
involved 35 fully formula-fed infants with an average age of 16 weeks. Two treatments were
administered: standard baby food with maltodextrin as the placebo and baby food with a
GOS/lc-inulin formulation. Nineteen infants were in the GOS/lc-inulin-supplemented group,
11 of whom were included in the per-protocol analysis (which specified compliance of 60%
consumption during the final two weeks of the study). Over the course of the 6-week study
period, infants consumed a total of 0.45 g lc-inulin and 4.05 g GOS per day in three equivalent
doses. Infants consuming the GOS/lc-inulin-supplemented food had a significant increase in
fecal bifidobacteria (43% at week 0 vs. 57% at week 6) above baseline levels, indicating the
ability of supplemental GOS/lc-inulin to alter fecal microflora in the weaning period.
Costalos and colleagues (2007) studied the impact on infant growth, tolerance and fecal
microflora of a GOS/lc-inulin formula in 70 healthy term infants 14 days of age. Infants
consumed 0.4 g lc-inulin per L and 3.6 g GOS per L for 12 weeks. Anthropometric
measurements were conducted at study entry and repeated at age 6 and 12 weeks. Stool
samples were taken at 6 weeks from 32 randomly selected infants. Mothers kept a diary and
questionnaire in which intake of formula and stool characteristics were recorded. Results were
compared to a control group of 70 infants. In total, 140 of the 160 infants originally enrolled
completed the study. Ten infants from the GOS/lc-inulin-group and ten from the control group
dropped out of the study. No clinically relevant adverse events related to the study formula were
reported. Nine infants (5 in the GOS/lc-inulin group and 4 in the control group) dropped out due
to change in formula and seven (4 in the GOS/lc-inulin and 3 in the control group) dropped out
due to minor gastrointestinal complaints. Four infants dropped out of the study for unknown
reasons. Measurements of weight gain, length gain and head circumference gain did not differ
between groups at 6 or 12 weeks of age. The GOS/lc-inulin infants experienced an increase in
stool frequency and consistency. No difference between groups with regard to infant
satisfaction, tolerance or occurrence of possetting was reported. No changes in the median

Safety Data

39

000055

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

proportion of fecal bifidobacteria, clostridia or E. coli as a percentage of the total


microorganisms reached statistical significance.
In a 6-week study involving 46 infants, the effects of consumption of milk containing 5.4 g per L
GOS in combination with lc-inulin and AOS (acidic oligosaccharides) were compared to milk
containing exclusively AOS and a control formula (Fanaro et al. 2005). There was a significant
increase in fecal bifidobacteria in the GOS/lc-inulin-supplemented formula group (n = 15) as
compared to both the AOS and control groups. Softer stools were observed compared to the
control group. The investigators reported no difference in growth, crying, vomiting or
regurgitation patterns among the groups.
In a study partially financed by Numico (now part of Danone), Bisceglia et al. (2009) evaluated
the ability of a prebiotic supplemented formula to modulate jaundice in otherwise healthy term
newborns (similar gestational age, birth weights, Apgar scores, method of delivery and initial
bilirubin). The study was a prospective, randomized, double-blind, placebo-controlled, clinical
trial. The formula was supplemented with either 0.8g/dL of a scGOS/lc-inulin mixture at a
9:1 ratio (n = 39; 18 m/21 f) or maltodextrin as the placebo (n = 37; 15 m/17 f) for a period of
28 days. Formula was provided when breast milk was not available; therefore, the precise
doses for each newborn are unknown. Newborns with a previously jaundiced sibling were
excluded. The study endpoints were bilirubin levels (transcutaneous measurement; TCB),
increases in weight, length, and head circumference, stool frequency and adverse effects.
The authors reported no adverse effects in either group and no statistically significant
differences in weight, length, or head circumference between groups. The prebioticsupplemented formula group had a significantly higher stool frequency compared to the placebo
group (3.4 0.7 daily evacuations versus 1.7 0.9; p < 0.001; repeated measures ANOVA).
Bilirubin was measured at 2, 24, 48, and 72 hours and at 5, 7, 10, and 28 days of life.
Beginning at 72 hours and at every point thereafter the prebiotic-supplemented formula group
had lower bilirubin levels (p < 0.05; repeated measures ANOVA; time x group interaction post
hoc test: Dunn test for multiple comparisons, p < 0.05). Bisceglia et al. (2009) concluded that,
consistent with Indrio et al. (2009a; 2009b), the pre-biotic supplementation may have increased
gastric activity and reduced emptying time thereby increasing stool frequency. The authors also
noted that the prebiotics may assist in the prevention or treatment of hyperbilirubinemia;
however, none of the subjects were in the pathological range and thus efficacy of prebiotics for
this condition is unsupported. The authors do state that the prebiotic supplementation appear to
carry no risks though endpoints were limited.
In a randomized, double-blind, controlled study involving 393 healthy 12-month-old toddlers,
Firmansyah et al. (2011) fed the toddlers either a mixture of probiotics (Bifidobacterium longum
BL999, Lactobacillus rhamnosus LPR), prebiotics (30% inulin and 70% fructo-oligosaccharide)
and long-chain polyunsaturated fatty acids (LCPUFA; arachidonic acid and docosahexaenoic
acid) or a control milk. The toddlers were fed approximately 400 mL/day for 12 months. The
authors recorded the toddlers weight gain, body length, head circumference, body mass index,
gastrointestinal tolerance, stool bacterial counts, adverse events, anti-vaccine immunoglobulin
G and neurodevelopment at 12, 14, and 16 months of age. 290 toddlers completed the study.
There were no significant differences between groups in anti-vaccine immunoglobulin G, mental

Safety Data

40

000056

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

and motor development measures or cognitive and adaptive behavior scores. There were no
significant differences between groups stool characteristics, length, head circumference, body
mass index, gastrointestinal tolerance. The toddlers in the pro-/prebiotic and LCPUFA group
did have a significantly higher weight gain. The higher weight gain was the only significant
difference between the two groups. There were no study related adverse events and the
frequency of adverse events did not vary significantly between the two groups.
Kim et al. (2007) conducted a cross-over study with 14 infants with an average age of
12.6 6.4 weeks. The infants were randomly assigned into two groups of seven and fed either
a milk-based formula containing 1.5 g of oligosaccharides (raffinose, GOS, & inulin) or a control
treatment of the same formula without the oligosaccharide supplement for three weeks. After
three weeks the groups switched formulas with no washout period between treatments. The
authors reported that nurses recorded the amount of feces, the frequency of defecation daily
and the feces consistency (on a scale from 1 (watery) to 4 (hard pellets)). The author reported
no significant difference in the total number of anaerobic bacteria or Bacteroides spp between
treatment groups. The authors did find a significant increase (p < 0.05) in the Bifidobacterium
spp (9.85 0.741 colony forming units (cfu) per gram (g)) and the Lactobacillus spp. (9.09
0.377) in the oligosaccharide treatment group (9.22 0.741 cfu/g and 8.61 0.741,
respectively). The only significant difference in the stool parameters was in the amount of feces
with the inulin treatment group averaging 167 80 g versus 75 30 grams from the control
group (p < 0.01). The authors did not observe any adverse effects for either group during the
study.
Lugonja et al. (2010) performed a 28-day clinical study on 21 infants that they assigned to one
of two groups, the first group (7 boys, 3 girls) received breast milk and the other group (6 boys,
5 girls) received an infant formula with 4.0 g/liter (L) chicory-derived inulin and FOS (source not
declared). The authors collected the infants weight and length, number of feeds, stool
frequency and consistency, and tolerance to formula intake every day of the study. The stools
were collected three times per day for bacteriological and biochemical analysis. Lugonja et al.
(2010) observed no differences in the fecal numbers of lactobacilli, total aerobes, anaerobes or
yeasts and fungi. The authors did observe a statistically significant increase in the numbers of
bifidobacteria in the stools of the prebiotic supplemented infants. The authors noted that the
incidence of side effects (flatus and regurgitations) and stool frequency and consistency did not
differ between the groups. Lugonja et al. (2010) concluded that the prebiotic supplemented
formula has similar bifidogenic effects as mature breast milk.
Magne et al. (2008) performed a double-blind, placebo-controlled, randomized intervention
study involving 82 healthy, term, partially breast-fed infants one week to three months of age
(72 infants completed the study). The study was sponsored by Numico Research (now Danone
Research). The authors excluded infants from the study under the following criteria: exclusive
breast feeding, exclusive formula feeding from birth, history of family atopic diseases, prior
incidence of diarrhea, illnesses that required systemic antibiotic treatment, consumption of foods
or products modulating intestinal microbiota, malnutrition, and twins or infants living in the same
house. The infants received one of three whey-based formulas provided in 400 g cans: a wheybase control formula (n = 23); a formula with scGOS and lc-inulin (n = 25); or a formula with
scGOS, lc-inulin and partially unsaturated and methylated acidic oligosaccharides (pAOS)

Safety Data

41

000057

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

(n = 24). Treatments were administered for two month partial formula-feeding period followed
by a two month formula only feeding period.
After the treatments, the authors analyzed the following: Total bacterial count; bacteria group
ratios (identification by in situ fluorescence hybridization); safety; stool characteristics; child
growth. The authors found that only the bacteria group populations differed significantly. The
authors found no significant difference between groups after breast feeding or after partial
formula feeding, but after formula-only feeding there was an increase in Bifidobacterium
(p = 0.0001) and a decrease in Bacteroides (p = 0.02) and C. coccoides (p = 0.01) in both the
scGOS/lc-inulin and scGOS/lc-inulin/pAOS treatment groups and the scGOS/lc-inulin/pAOS had
a significant increase (p = 0.01) over the scGOS/lc-inulin group. No mothers spontaneously
stopped the formula and there were no significant adverse effects. The authors concluded that
the scGOS/lc-inulin and scGOS/lc-inulin/pAOS supplemented formulas were clinically safe and
effective on fecal microbiota.
Panigrahi et al. (2008) performed a double-blind, randomized, placebo-controlled study
involving 31 newborns (1-3 days post-birth; all delivered by cesarean section) to evaluate the
long-term colonization potential Lactobacillus plantarum in neonatal guts. Treatments consisted
of receiving either oral preparation of L. plantarum and fructo-oligosaccharides (n = 19) or a
dextrose saline placebo (n = 12) for 7 days. All infants were exclusively breast fed for
approximately six months after delivery and the authors note that vaginal delivery and formula
feeding were not addressed by this study. The authors reported no significant adverse effects
in either group.
Puccio et al. (2007) performed a prospective, randomized, double-blinded, reference-controlled,
parallel-group, clinical trial study sponsored by Nestl Nutrition, Nestec Ltd. (no affiliation with
Danone). The researchers assessed the safety of an experimental probiotic formula containing
B. longum culture (2 x 107 cfus) and a GOS:FOS (9:1 ratio; 4 g/L) prebiotic mixture compared to
a reference (control) formula. The authors did not evaluate a prebiotic-only formula. By
14 days after their birth, 138 infants ceased to be breast fed and for seven months thereafter the
infants received either the control formula (n = 55; 25 m/30 f) or the experimental formula
(n = 42; 20 m/22 f). The primary endpoint was weight gain during the first four months of life
with length, head circumference and gastrointestinal (GI) tolerability as secondary endpoints.
GI symptoms were recorded by parents and included stool frequency (events per day), color,
consistency, and odor; flatulence; frequency of vomiting or spitting up; infant comfort (frequency
of crying (episodes >10 min), restlessness, irritability); and the occurrence of parent-diagnosed
colic. Formula intake was recorded by parents as 3 day means and reported by the authors as
mL/day.
Of the 138 infants recruited, 97 completed the study (control = 55; experimental = 42). Puccio
et al. (2007) reported data for both the intention-to-treat (ITT) population and the per-protocol
(PP) population. The ITT results do not differ significantly from the PP results, therefore, only
the PP results are described herein. Puccio et al. (2007) found that the infants fed the
experimental formula had a statistically significant increase in stool frequency (2.2 0.7 versus
1.8 0.9 occurrences/d; t-test, p = 0.018), but this was not correlated with a decrease in growth.
The authors also found a lower incidence in constipation and the higher stool frequency was

Safety Data

42

000058

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

comparable to breast-fed infants and did not indicate or correlate with diarrhea or decrease in
growth. The authors established predefined equivalence boundaries of 3.9 g/d for weight gain
and the mean weight gain in the two groups fell within the boundaries and are, therefore,
equivalent (+1.09 g/d; 90% confidence interval -0.98 to +3.15). The mean changes in length
and head circumference were also not statistically significant (p > 0.1 for both parameters).
Infants receiving the experimental formula had a reduced incidence of constipation (chi-square
test, p = 0.03). Puccio et al. (2007) evaluated the high drop-out rate and found no statistical
difference between the control and experimental group. The authors noted that adverse events
in the ITT population ranged from serious (including one death in the experimental group and a
congenital disorder in one of the control group subjects) to non-serious with no significant
difference between groups in either total incidence or severity of event. The respiratory tract
(bronchiolitis and cough) was the most frequently affected organ system in both groups. The
authors reported that no adverse events were related to the study formula. Puccio et al. (2007)
concluded that the experimental formula was safe and well tolerated and may have beneficial
effects on full-term infants.
To evaluate the effect of prebiotics on basal immune parameters, Raes et al. (2010) performed
a double-blind, placebo-controlled study with 215 healthy term infants for the first 26 weeks of
their life. The infants received either breast milk, a non-supplemented infant formula (provided
by Danone Research) or the formula supplemented with 0.6 g/100 mL scGOC:lc-inulin
(9:1 ratio) ad libitum throughout the study period. (The breast fed group cannot be randomized,
but was incorporated for explorative reasons according to the authors.) The infants did not vary
significantly based on gestational age, birth weight, birth length, head circumference, Apgar
score, gender, or parental allergies. Of the 215 initial subjects, 187 infants completed the study.
The evaluated the following parameters at weeks 8 and 26: white blood cell counts, lymphocyte
(subsets and absolute), C-reactive protein, and total immunoglobulin A, G, M, and E. The only
statistically significant difference between the breast fed and formula fed groups that Raes et al.
(2010) found was the higher levels of white blood cell counts at week 26 in the breast fed group.
The authors found no significant differences between formula fed groups. Raes et al. (2010)
noted that the presented data do not indicate any negative side effect of the studied prebiotics;
however, the authors did not present information on adverse events or reasons for the
28 dropouts.
Raes et al. (2010) concluded the prebiotic mixture, when compared to a non-supplemented
formula and exclusive breastfeeding in the first six months of life, did not alter the basal level of
the measured immune system parameters in healthy infants. Disclosures: This study was
funded by Danone Research and one of the authors, P. A. M. J. Scholtens, was affiliated with
Danone Research and the formula was provided by Danone Research. The lc-inulin was
Raftiline HP and provided by Orafti (now Beneo, the manufacturer of Danones lc-inulin).
Rao et al. (2009) identified 24 randomized control studies of prebiotic supplementation efficacy
and safety in term infants and performed a systematic review of eleven of these studies. The
lc-inulin studies included in Rao et al. (2009) are individually abstracted herein (Alliet et al.,
2007; Bakker-Zierikzee et al., 2005; Ben et al., 2004; Costalos et al., 2008; Fanaro et al., 2005;
Moro et al., 2002; Moro et al., 2006). The studies compared the effects of formula milk with and
without supplementation by prebiotics on infants of age 28 days or less and continuing for at

Safety Data

43

000059

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

least two weeks. The authors defined prebiotics as GOS, FOS or both. The authors focused
the following endpoints: stool colony counts (bifidobacteria, lactobacilli, and pathogens), pH,
consistency, frequency, anthropometry, and symptoms of intolerance (Rao et al. 2009). The
meta-analysis by the authors of pooled data found that the infants in the prebiotic group had a
higher weight gain (weighted mean difference (WMD) of 1.07 g, p = 0.02) and lower stool pH
(WMD of -0.65, p < 0.00) though the statistical heterogeneity was significant (p < 0.001).
Rao et al. (2009) did not perform a meta-analysis for the bacteria counts because the methods
for measuring and reporting counts in the studies were too different; however, of the 11 trials
(n = 1459), nine evaluated the effect of prebiotics on bifidobacteria colony counts. The authors
found that six reported significant increases in bifidobacteria counts after prebiotic
supplementation; three studies did not find a statistically significant difference. Additionally, the
authors noted that of three studies that reported stool frequency, all reported higher frequency in
the prebiotic supplemented groups. These studies noted the frequencies were similar to those
of breastfed infants and not an adverse outcome. According to the authors, seven of eight of
the studies that reported on tolerance reported that the prebiotic-supplemented formula groups
did not differ from the control group. Rao et al. (2009) reported that a previous study had found
an increase in eczema, irritability, and diarrhea in the prebiotic group; however, Rao et al.
(2009) noted that Moro et al. (2006) found a decrease in atopic dermatitis and wheezing at six
and 24 month follow-ups. Rao et al. (2009) concluded that there is inadequate evidence
regarding the use of prebiotics for prevention of atopic diseases. The lack of consistent
analytical methods complicated the meta-analysis of efficacy data though the prebiotic
supplements were well tolerated across studies.
In a double-blind, randomized, placebo-controlled study, Salvini et al. (2011) evaluated the
effects of a commercial formula supplemented with 8 g/L scGOS/lc-inulin (9:1) mixture (a
Danone product) on 20 term infants that were not breast fed and were born to hepatitis C virusinfected mothers (note: all enrolled infants tested negative when measured for qualitative
plasma HCV viral load). Danone Research supported the study and two of the authors were
employed by Danone at the time. The authors objective was to determine if the prebiotic
supplementation could result in long-term colonization of bifidobacteria when compared to a
placebo group receiving a formula with the same amount of maltodextrin. The authors
measured each subjects blood leukocytes, fecal microbes and anthropometric variables at
birth, three, six, and 12 months and hepatitis B vaccine-specific IgG at 12 months. Two
subjects of the initial 22 did not complete the study: One in the placebo group due to
noncompliance and one in the prebiotic group because follow-up was discontinued for unknown
reasons. All infants were born via vaginal delivery and neither group differed in sex, gestational
age, weight, length, or head circumference. No clinically relevant infections or antibiotic
treatments occurred during the month prior to each evaluation. Salvini et al. (2011) found that
though bifidobacteria and lactobacilli increased during the first three months and then stabilized
in both groups, the counts of bifidobacteria and lactobacilli were higher in the prebiotic group
(p = 0.016 and p = 0.0042, respectively). The authors also observed that the prebiotic group
had lower fecal pH than the control group at 3 months (5.24 0.32 and 6.25 0.51,
respectively; p = 0.0006) but not at six months (5.88 0.33 and 6.2 0.58; p = 0.3). Serum IgE
and IgG levels were not affected by treatment. The authors did not record any adverse effects
with either group and noted that both formulas were well tolerated. The study is limited by the
following: the size of the study (n = 20) is very small; the authors used culturing methods of

Safety Data

44

000060

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

microbial quantification rather than molecular techniques such as fluorescent in situ


hybridization.
Arslanoglu et al. (2007) investigated the effects of a prebiotic-supplemented formula (8 g/L of a
scGOS/lc-inulin mixture, 9:1 ratio, provided by Numico) on infections in term infants with a
parental history of atopy (n = 206). The double-blind, randomized, placebo-controlled study was
funded by Numico and a Numico researcher co-authored the paper. The infants received either
a hypoallergenic formula with extensively hydrolyzed cows milk whey protein with maltodextrin
as a placebo or the scGOS/lc-inulin supplemented formula for six months. The authors
evaluated parent-reported infections (type, number, and antibiotic use). The prebiotic treatment
group had fewer infectious episodes (p = 0.01) and a lower cumulative incidence of any
recurring infection (3.9% vs. 13.5%; p < 0.05) and recurring respiratory infection (2.9% vs. 9.6%;
p < 0.05). Both treatment populations had the same number of dropouts citing flatulence, hard
stools, or no reason (a total of five in each group).
Arslanoglu et al. (2008) performed a two-year double-blind follow-up study on the treatment
population described in Arslanoglu et al. (2007) to investigate the cumulative incidence of
allergic manifestations as well as number of infectious episodes and growth in the infants first
two years of life. The authors observed lower incidences of recurrent wheezing (7.6 vs. 20.6%),
atopic dermatitis (13.6 vs. 27.9%), and allergic urticaria (1.5% vs. 10.3%) in the prebiotic group
(p < 0.05). The prebiotic group also had fewer episodes of infections (p < 0.01), fever
(p < 0.00001), and fewer antibiotic prescriptions (p < 0.05), and no significant difference in
growth (Arslanoglu et al. 2008).
In a prospective, randomized, placebo-controlled, open (unblinded) study, Bruzzese et al.
(2009) evaluated the effects of prebiotic supplementation on the incidence of intestinal and
respiratory tract infections and anthropometric measures in the first 12 months of life. The
authors noted that the study was partially funded by Numico Research and that none of the
authors received personal gain from the study. The prebiotic formula consisted of 0.4 g/100mL
GOS/FOS (9:1 ratio). The authors randomized 342 infants of which 201 completed the study.
Dropouts were due to lack of follow-up (56), protocol violations (80) or discontinued intervention
(5; due to intolerance of cows milk protein). Bruzzese et al. (2009) found that the incidence of
gastroenteritis was lower in the prebiotic group than the control group (0.12 0.04 vs. 0.29
0.05 episodes/infant/12 months; p = 0.015) as was the number of infants with multiple antibiotic
courses per year (prebiotic: 24/60 vs. 43/65; p = 0.004). The authors also observed a
temporary increase in body weight (lasting the first six months) in the infants fed the prebioticsupplemented formula. The authors reported no major side effects and that the infants
receiving prebiotic supplementation generally had softer stools, though the prebiotic formula
was not withdrawn in any case.
Healthy, term, formula-fed and breast-fed infants (n = 1130) without a family history of allergic
disease were followed by Grber et al. (2010) in a double-blind, placebo-controlled, randomized
prospective intervention study that evaluated the occurrence of atopic dermatitis up to the first
year of life. The prebiotic-supplemented formula consisted of 6.8 g/L of scGOS/lc-inulin
(9:1 ratio) and 1.2 g/L pAOS in a non-hydrolyzed cows milk-based formula. The final study
population consisted of the breast-fed group (n = 243), the prebiotic-supplemented group

Safety Data

45

000061

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

(n = 292), and the control group (n = 300). The authors reported that the prebiotics were safe
and that the reasons for dropout included occurrence of disease, intolerance of formula,
protocol violations and changes to formula and were similar across groups. Grber et al. (2010)
found that the incidence of atopic dermatitis was lower in the prebiotic group (5.7% 1.2%) than
the control group (9.7% 1.5%; p = 0.04) but not significantly different than the breast-fed group
(7.3% 1.6%). The authors did not observe a significant difference in the severity of atopic
dermatitis between groups. In addition to providing research support, honoraria, or employment
to several of the study authors, Danone Research supplied the study formulas and facilitated
data collection and analysis.
Piemontese et al. (2011), Stam et al. (2011) and van Stuijvenberg et al. (2011) performed
additional studies and analysis on the same treatment population as Grber et al. (2010).
Piemontese et al. (2011) found that infants receiving a prebiotic-supplemented formula did not
vary significantly from a non-supplemented formula control group with respect to mean weight,
length, and head circumference, skin fold thicknesses, arm circumference gains and stool
frequency. The group of breast-fed infants showed significantly larger skin fold thicknesses at
eight weeks of age but smaller thicknesses at 52 weeks. During the study, the authors recorded
640 adverse events in 431 infants consisting mostly of illnesses; but there was no significant
difference in the number of adverse events between the groups. In 2011, van Stuijvenberg
et al. evaluated the number of fever episodes reported by the infants parents. The researchers
found that the number of fever episodes did not vary between the groups (prebiotic group
median number of episodes = 1.19; control formula group median = 1.16; breast-fed group
median = 1.24). Stam et al. (2011) evaluated the effect of the prebiotic supplementation on
immunoglobulin responses to Haemophilus influenza type b (Hib) and tetanus immunizations in
a subset of the study population (n = 164). The authors observed no differences in Hib antibody
levels (> 1.0 ug/mL) between the prebiotic group (91.9%) and the control group (91.2%) and no
differences in the tetanus immunizations (median values of immunoglobulin G > 0.1 IU/mL
cut-off in both groups). Stam et al. (2011) concluded that the prebiotic formula did not affect
responses to vaccinations.
Scholtens et al. (2008) evaluated the effect of a formula with and without prebiotic
supplementation or breast-feeding on the composition of intestinal microbiota and the fecal
secretory immunoglobulin A (sIgA) response in healthy infants (n = 215; 187 completed the
study) during the first 26 weeks of life. In their randomized, double-blind, placebo-controlled
study, the authors found that the prebiotic (6 g/L scGOS/lc-inulin; 9:1 ratio; provided by Numico)
formula-fed infants had a higher percentage of bifidobacteria (60.4% vs. 52.6%; p = 0.04), a
lower percentage of Clostridium spp. (0.0% vs. 3.27%; p = 0.006) and a higher concentration of
sIgA (719 g/g vs. 263 g/g; p < 0.001) than the unsupplemented formula-fed infants. The
intestinal microbiota were determined by quantitative fluorescent in situ hybridization. The
authors recorded 96 adverse events: 40 (of 85 infants or 47%) in the prebiotic group, 40 (of
89 infants or 45%) in the control formula group and 16 (of 38 infants or 42%) in the breast fed
group. The authors do not describe the adverse events or the reasons for dropout.
Schouten et al. (2011) and van Hoffen et al. (2009) reported results of studies evaluating the
effects of scGOS/lc-inulin supplementation in infant formula on atopic dermatitis (AD) and
immune response, respectively. The studies are based on methods and a study population

Safety Data

46

000062

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

established by Moro et al. (2006) that was partially funded by Numico Research and includes
several of the same authors (G. Boehm of Danone Research, S. Arslanoglu, and G. Moro).
Moro et al. (2006) provided their study population either a hypoallergenic whey formula with
8 g/L scGOS/lc-inulin (9:1 ratio) or a maltodextrin placebo for the first six months of life.
Based on the Moro et al. (2006) finding that AD occurred less frequently in infants receiving the
scGOS/lc-inulin formula, van Hoffen et al. (2009) evaluated a subset of the same population
(n = 84) for effects of the scGOS/lc-inulin formula on the immune response in the infants.
Specifically, the authors measured plasma total immunoglobulins (Ig) E, G1, G2, G3, G4, cows
milk protein (CMP)-specific IgG1 and diphtheria, tetanus, and polio (DTP)-specific Ig by
enzyme-linked immunosorbent assay (ELISA) at three months post-vaccination for DTP
(six months of age). The authors observed a significant reduction in plasma IgE, IgG1, IgG2,
IgG3 (p < 0.01), and CMP-specific IgG1 (p < 0.05), but no difference in IgG4 and DTP-specific
Ig. van Hoffen et al. (2009) concluded that the prebiotic supplementation effectively modulated
the response to CMP without negatively affecting the vaccination.
In a study based on a subset of the Moro et al. (2006) study population (n = 74), Schouten et al.
(2011) measured plasma kappa and lambda immunoglobulin free light-chain (Ig-fLC)
concentrations in infants at risk of developing allergic disease. The authors evaluated the
Ig-fLC concentrations because, they note, these have been found in higher concentrations in
patients suffering from AD, cows milk allergy, allergic rhinitis, or asthma. Consistent with their
assumptions, the authors found that the plasma of infants suffering from AD had higher total
kappa and lamba Ig-fLC than infants without AD. Additionally, the infants receiving the
scGOS/lc-inulin supplemented formula had significantly lower Ig-fLC levels (p < 0.001) which
are consistent with the reduced incidence of AD in the infant population receiving the
scGOS/lc-inulin formula found in Moro et al. (2006).
In a study funded by Danone Research, Vaisman and colleagues (2010) evaluated the effects
of 6 g/day supplementation of prebiotics (80% GOS/lc-inulin, 9:1 ratio, 20% pAOS) on cytokine
levels (IL-1, IL-1RA, IL-6, IL-8, IL-10, sIL-2R and TNF-) in infants and toddlers aged 9 24
months with acute diarrhea (n = 104). The double-blind, placebo-controlled, prospective,
randomized study evaluated the number of stools and stool consistency based on parents
answers to a questionnaire and cytokine levels based on blood samples. The authors
evaluated cytokine levels to determine if prebiotics modulate inflammatory processes of acute
diarrhea. According to Vaisman et al. (2010), intervention with prebiotics did not significantly
affect stool characteristics or cytokine levels except for TNF-, though the authors note that this
did not appear to be of clinical relevance.
Vivatvakin et al. (2010) investigated the impacts of prebiotic supplementation in formula on
gastrointestinal comfort in infants in a study funded by Nestec Ltd. In this prospective, doubleblind, randomized, placebo-controlled study the authors evaluated 144 healthy full-term infants
fed either a whey-based formula containing LCPUFAs and 4 g/L of a GOS/FOS mixture
(9:1 ratio), a control formula, or breast milk from 30 days to 4 months of age. The researchers
measured gastric emptying time (by ultrasound), stool characteristics (frequency, consistency,
color, smell, and bacterial analysis), intestinal transit time, digestive tolerance (episodes of colic,
occurrence of crying, spitting up, and vomiting) and anthropometric measurements (weight,

Safety Data

47

000063

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

length, head circumference). When compared to the control group, Vivatvakin et al. (2010)
found that the prebiotic formula group had fewer hard stools (7.5 vs. 0.7%, respectively;
p < 0.001) and more soft stools (82.3% vs. 90.8%; p < 0.05) while the bacterial composition,
gastric emptying time, and anthropometric measurements did not differ significantly between
groups. The authors reported that adverse events occurred in all groups at the same
percentage with no significant difference and for similar issues: upper respiratory tract
infections, diarrhea, vomiting, and constipation.
In a series of three papers based on a double-blind, placebo-controlled study involving
90 infants aged < 7 months and formula fed, exclusively, van der Aa et al. (2010; 2011; 2012)
investigated the effects of a 8 g/L scGOS/lc-inulin mixture (9:1 ratio) in a hydrolyzed wheybased formula that included Bifidobacterium breve M-16V on infants with atopic dermatitis (AD;
van der Aa et al. 2010), AD and asthma-like symptoms (van der Aa et al. 2011), and systemic
immunomodulatory effects in infants with AD (van der Aa et al. 2012). Danone Research
funded the studies which included authors associated with Danone. In van der Aa et al. (2010)
the infants received either the synbiotic formula (with B. breve M-16V and the scGOS/lc-inulin at
8 g/L) or a placebo formula that excluded the synbiotics and B. breve for 12 weeks. The
authors evaluated the primary outcome of change in severity of AD using the SCORAD index to
account for the extent of the eczema, the intensity of lesions and subjective symptoms.
Secondary outcome measures were change in topical corticosteroid use; total serum IgE,
specific IgE (against food and inhalant allergens); change in microbiota composition, SCFAs,
lactate, and pH; stool frequency and consistency, and occurrence of GI symptoms, diaper
dermatitis and adverse events. The authors found that the synbiotic mixture of B. breve M-16V
and the scGOS/lc-inulin did not have a beneficial effect on the severity of infant AD, but did
change the composition (increased bifidobacteria, reduced pathogenic bacteria) and metabolic
activity of intestinal microbiota (low pH, high lactate and low SCFA. The authors noted that the
synbiotic mixture was well tolerated and did not cause any adverse events.
A follow up performed after one year with 75 children of the original study population (70.1%
male, average age of 17.3 months) found that the synbiotic group had a significantly lower
prevalence of frequent wheezing and wheezing and/or noisy breathing apart from colds than
the placebo group and also had fewer children using asthma medication (van der Aa et al.
2011). Total serum IgE levels did not differ significantly between the synbiotic and placebo
groups. The third study based on this treatment population evaluated ex vivo cytokine
production by peripheral blood mononuclear cells as well as circulating regulatory T cell
percentage and found no detectable effects due to the synbiotic supplemented formula (van der
Aa et al. 2012).
Veereman-Wauters et al. (2011) performed a 28-d prospective, randomized, double-blind
clinical trial with 110 full-term healthy infants. The authors randomized the infants into four
formula groups: control formula (n = 21), formula plus 4 g/L of a 50:50 mixture of lc-inulin and
oligofructose (SYN1; n = 21), formula plus 8 g/L SYN1 (n = 20), and formula plus 0.8 g/L
GOS/lc-inulin (9:1 ratio; n = 19). Beneo supported this study. The lc-inulin used in this study
was Orafti HP, the same product that is the subject of this dossier. A fifth group consisted of
breast-fed infants as a control (n = 29). Veereman-Wauters et al. (2011) stated that the study
objective was to assess physiological parameters (safety and tolerance) and bifidogenic effects

Safety Data

48

000064

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

of different doses of prebiotic supplemented formulas as compared to standard formula and


breast fed infants. The authors evaluated the results of parental diaries that recorded formula
intake, stool frequency and consistency, crying behavior, occurrence of regurgitation or
vomiting. The authors also evaluated anthropometric data (length, weight, and head
circumference) and the fecal bacteria in stool samples collected on days 3, 14 and 28 (flora
analysis).
Veereman-Wauters et al. (2011) found no significant differences between groups for
anthropometric parameters, stool frequency, crying behavior, regurgitation or vomiting. The
authors found that prebiotic supplement at 8 g/L of formula contributed to softer stools with a
consistency resembling that of the breast fed infants. The authors also found infants receiving
the 8 g/L prebiotic supplemented formulas also had comparable levels of bifidobacteria as the
breast fed group. Veereman-Wauters et al. (2011) reported that no serious adverse events
occurred during the study. Non-serious issues included hunger, regurgitation, and symptoms of
gastroesophageal reflux. The number of dropouts was evenly distributed across groups (6 8
per group) including controls leading the authors to conclude that the intervention was not a
contributing factor.
In summary, results from 43 studies of healthy, term infants consuming formula supplemented
with up to 0.8 g lc-inulin and 7.2 g GOS per L for periods ranging from two weeks to up to twelve
months provide evidence that lc-inulin, in combination with GOS, is well tolerated by infants and
produces no adverse effects. The supplemented formulas supported normal growth.
Additionally, results from these studies suggest that formulas with added lc-inulin in combination
with GOS may influence shifts in infant gut microflora that result in gut microflora more similar to
the gut microflora of breast-fed infants.

(The remainder of this page intentionally left blank.)

Safety Data

49

000065

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Study Design &
Objective

Reference

Alliet et al.
2007

To study the effects


of a 9:1 GOS/lc-inulin
mixture on
cholesterol and
triacylglycerol levels
in infants.

Treatment
Population a

n = 75;
age = 8 wks

Arslanoglu
et al. 2007

Arslanoglu
et al. 2008

Randomized, doubleblind, placebocontrolled.


To investigate the
sumulative incidence
of allergic
manifestations,
infectious episodes,
and growth.

Formula with 5.4 g


GOS/L & 0.6 g lcinulin/L

Treatment
Duration

18 weeks

Source: Numico (now


Danone)

Randomized, doubleblind, controlled.


To investigate the
effects of a prebioticsupplemented
formula on infections
in infants with a
history of atopy.

Test Substance and


Dose

n = 206;
age = 2 wks

8 g/L of scGOS/lcinulin; 9:1 ratio

6 months

Source: Numico

Randomized, doubleblind, placebocontrolled.

Safety Data

n = 134;
age = up to 2 yrs

8 g/L of scGOS/lcinulin; 9:1 ratio for


first 6 months of life
Source: Numico

50

6 months

Results of GOS/lc-inulin Ingestion


Growth: Only baseline anthropometric data given.
Tolerance: Not measured.
Dropouts: 86 infants entered GOS/lc-inulin treatment
group and 75 completed the study; no explanation of
dropouts provided.
Other Endpoints:
-No difference in levels of total cholesterol and LDL
cholesterol between GOS/lc-inulin and standard
formula-fed infants; total cholesterol and LDL cholesterol
for both formula groups lower than breast-fed infants.
-No difference in HDL cholesterol or triacylglycerols
among groups.
Growth: No difference in growth.
Tolerance: Not stated.
Dropouts: same as in placebo group (5); flatulence,
hard stools, relocation, or no reason.
Other Endpoints:
-Fewer infection episodes.
-Lower cumulative incidence of recurring infection.
-Lower cumulative incidence of recurring respiratory
infection.
-Increased bifidobacteria.
Growth: No difference in growth.
Tolerance: Not stated.
Dropouts: See Arslanoglu et al. 2007.
Other Endpoints:
-Lower incidences of recurrent wheezing, atopic
dermatitis, and allergic urticaria.
-Fewer episodes of infections and fever.
-Fewer antibiotic prescriptions.

000066

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Study Design &
Objective

Reference

BakkerZierikzee et
al. 2005,
2006

Bisceglia et
al. 2009

Study the effects of


GOS/lc-inulin on fecal
microflora and SIgA
levels.

Treatment
Population a

n = 19;
age = 3 days
postpartum

Randomized, doubleblind.

To evaluate ability of
prebiotics to
modulate jaundice in
otherwise healthy
term newborns.
Prospective,
randomized, doubleblind, placebocontrolled.

Test Substance and


Dose

Treatment
Duration

Formula with 5.4 g


GOS/L & 0.6 g
lc-inulin/L
16 weeks
Sources: Friesland
Foods Domo (GOS);
Orafti (FOS)

Formula
supplemented with
0.8g/dL of a 9:1
scGOS/lc-inulin or
maltodextrin placebo.

n = 76

Formula was
provided when breast
milk was not
available; therefore,
the precise doses for
each newborn are
unknown

28 days

Results of GOS/lc-inulin Ingestion


Growth:
-Data not measured/reported.
Tolerance:
-No effect on fecal consistency or frequency.
Dropouts:
-5 infants dropped out of the GOS/lc-inulin group; 4
dropped out of the control group; and 4 dropped out of
the Bb-12 group. Reasons for dropping out included
colic, suspicion of cows milk allergy, constipation, and
practical problems. Reasons by group were not
specified.
Other Endpoints:
-No change in % fecal bifidobacteria.
-Decreased fecal pH.
-No change in total fecal SCFA; increased % fecal
acetate, decreased % fecal propionate, butyrate,
isobutyrate, isovalerate, valerate.
-No change in fecal lactate.
-Increase in median fecal SIgA concentration at week
16.

Growth: Gain in weight, length and head circumference


did not differ.
Tolerance: The authors reported no adverse effects in
either group.
Dropouts: 10 due to denied consent; 8 for unknown
reasons.
Other Endpoints:
-Beginning at 72 hours and at every point thereafter the
prebiotic-supplemented formula group had lower
bilirubin levels.
-Higher stool frequency.

Source: Numico

Safety Data

51

000067

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Reference

Bruzzese
et al. 2009

Study Design &


Objective
To evaluate the
effects of prebiotics
on the incidence of
intestinal and
respiratory tract
infections and
anthropometric
measures.

Treatment
Population a

n = 201;
age = 15 120 d

Test Substance and


Dose

4 g/L GOS/lc-inulin
9:1 ratio
Source: Not reported.

Treatment
Duration

First 12
months of
life.

Growth: Temporary increase in body weight of


prebiotic-fed infants.
Tolerance: Softer stools in with prebiotics.
Dropouts: 56 due to lack of follow-up, 80 due to
protocol violations, 5 due to intolerance of cows milk
protein (342 started, 201 completed).
Other Endpoints:
-Lower incidence of gastroenteritis.
-Fewer infants had multiple antibiotic courses per year.

12 weeks

Growth:
-Gain in weight, length and head circumference did not
differ.
Tolerance:
-Increase in stool frequency and number of stool per
day.
-No difference in formula satisfaction, tolerance or
occurrence of possetting.
Dropouts:
-10 infants dropped out of both groups; no adverse
events related to study formula reported.
Other Endpoints:
-At 6 wk, median bifidobacteria, clostridia and E. coli
fecal proportions as a percentage of the total
microorganism count did not differ.

Prospective,
randomized, placebocontrolled, open.

Costalos et
al. 2007

To study the effects


of GOS/lc-inulin on
infant growth,
acceptability and
fecal microflora.
Randomized, doubleblind, placebocontrolled

Safety Data

n = 70;
age = mean 4.7
days

Formula with 3.6 g


GOS/L & 0.4 g lcinulin/L

Source: Numico

52

Results of GOS/lc-inulin Ingestion

000068

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Study Design &
Objective

Reference

Fanaro, et
al. 2005

Firmansya
h et al.
2011

To study tolerance
and the effects of
formula with AOS or
GOS+FOS/AOS on
intestinal flora and
stool characteristics
in term infants.

To determine if
Bifidobacterium
longum, Lactobacillus
rhamonosus,
prebiotics, and longchain
polyunsaturated fatty
acids (LCPUFA)
increased growth
versus a control milk.

Treatment
Population a

n = 46

n = 393

Test Substance and


Dose

3 treatments:
Standard formula,
formula with 5.4 g
GOS/L & 0.6 g lcinulin/L + AOS,
formula with AOS

Mixture of probiotics
(Bifidobacterium
longum, Lactobacillus
rhamnosus),
prebiotics (30% inulin
and 70% FOS),
LCPUFA or a control
milk.

Treatment
Duration

6 weeks

12 months

400 mL/day.
Randomized, doubleblind, placebocontrolled.

Grber et
al. 2010

To assess the effect


of prebiotics on
occurrence of atopic
dermatitis in infants.
Randomized, doubleblind, placebocontrolled.

Safety Data

Source: Not reported.

n = 1130;
age < 8 wks

6.8 g/L of scGOS/lcinulin (9:1 ratio) and


1.2 g/L pAOS in a
non-hydrolyzed cows
milk-based formula
Source: Nutricia

53

1 year

Results of GOS/lc-inulin Ingestion


Growth:
- No difference in growth patterns.
Tolerance:
-Softer stool consistency compared to control group.
-No difference in crying, vomiting or regurgitation
patterns.
Dropouts:
-None reported.
Other Endpoints:
-Increased fecal bifidobacteria compared to control and
AOS groups.
-Decreased pH compared to control and AOS groups.
Growth: pro-/prebiotics w/LCPUFA had higher weight
gain; length, head circumference did not differ.
Tolerance: No study related adverse events.
Dropouts: 38 dropped out of experimental milk group,
41 dropped out of control milk group.
Other Endpoints:
- There were no significant differences between groups
in anti-vaccine immunoglobulin G, mental and motor
development measures, cognitive and adaptive behavior
scores, stool characteristics, body mass index,
gastrointestinal tolerance.
Growth: No difference (see Piemontese et al. 2011).
Tolerance: No difference between groups.
Dropouts: 129 total; similar across groups; due to
withdrawal, occurrence of disease, protocol violations,
intolerance of formula, change of formula.
Other Endpoints:
-Lower incidence of atopic dermatitis than the placeboformula group, but not than the breast-fed group.
-No difference in severity of atopic dermatitis.

000069

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Study Design &
Objective

Reference

Kim et al.
2007

Cross-over study to
investigate the effect
of native inulin in
formula-fed babies.

Safety Data

Treatment
Population a

n = 14
age = 12.6 6.4
wks

Test Substance and


Dose
0.25g/kg/day, 3
weeks inulin; followed
by 3 weeks without,
or vice versa.
Average intake of
inulin was 1.5( 0.3
g/d) milk-based
formula of 1.5 g OS
(raffinose, GOS &
inulin) or control of
same formula without
OS supplement for 3
weeks. After 3
weeks the group
switched formulas.

54

Treatment
Duration

Results of GOS/lc-inulin Ingestion

3 weeks

Growth: No differences in growth.


Tolerance: No study-related adverse effects.
Dropouts: None reported.
Other Endpoints:
-No significant difference in total # of anaerobic bacteria
or Bacteroides spp between treatment groups.
-Increase in Bifidobacterium spp cfu/g and Lactobacillus
spp in oligosaccharide treatment group.
-Consumption of inulin increased the content of
Bifidobacterium and Lactobacillus in the feces of
formula-fed babies, without affecting the number of
Bacteroides or the total anaerobic count.

000070

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Study Design &
Objective

Reference

Treatment
Population a

Test Substance and


Dose

Treatment
Duration

Results of GOS/lc-inulin Ingestion


Growth: Data not measured/reported.
Tolerance: Increase in median stool frequency. Mild
flatulence in all groups.
Dropouts: Eight dropouts; 3 from test group, 3 from
control group and 2 from reference group.

Knol et al.
2005a;
Haarman
and Knol
2005, 2006

Determine effects of
GOS/lc-inulin on
microflora in infants
exclusively formulafed since birth.

Randomized, doubleblind, placebocontrolled


intervention.

Safety Data

n = 15-19;
age = mean 7.7
wks

Formula with 7.2 g


GOS/L & 0.8 g
lc-inulin/L
6 weeks
Source: Aptamil,
Milupa

55

Reasons included rotavirus-infection (n=1), medical


problems of regurgitation (n=1) and formula-related
complaints (n=4; 2 from test and 2 from control).
Unknown reasons for drop outs in reference group.
Other Endpoints:
-Increased % fecal bifidobacteria.
-B. infantis, B. breve and B. longum remained
predominant species in GOS and HM groups.
-Decreased % B. adolescentis from baseline, decreased
%. B catenulatum from control.
-Increased % fecal lactobacilli.
-Increased L. acidophilus, L. paracasei and L. casei and
decreased L delbrueckii.
-At study completion fecal bifidobacteria and lactobacilli
accounted for 80% of fecal bacteria.
-Fecal bacteria more closely resembled HM-fed infants.
-Increased fecal acetate and decreased butyrate and
propionate.
-Decreased fecal pH.
-Increased fecal lactate and % D-lactate.

000071

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Study Design &
Objective

Reference

Lugonja et
al. 2010

Magne et
al. 2008

To study the
bifidogenic effect of
an infant formula
supplemented with
inulin and FOS
clinically and in vitro.

To test the safety and


effect of formula with
GOS, lc-inulin & AOS
on fecal microbiota in
infants at the age of
partial formula
feeding.
Double-blind,
placebo-controlled,
randomized
intervention.

Safety Data

Treatment
Population a

n = 21

Test Substance and


Dose

Formula
supplemented with
chicory-derived inulin
and FOS at 4.0 g/L.

Treatment
Duration

28 days

Growth: Gain in weight and length did not differ.


Tolerance: No significant adverse effects.
Dropouts: None reported.
Other Endpoints:
-Bifidobacteria levels in the stools from the formula-fed
infants were significantly higher than in the HM-fed
infants.
-The number of lactobacilli increased in both the
formula-fed and HM-fed infants.
-Increase in bifidobacteria and lactobacilli populations,
decrease in the total aerobes, anaerobes and yeasts
and fungi levels

2 months
partial
formulafeeding
period,
followed by
two month
formula
only
feeding
period

Growth: Weight gain did not differ.


Tolerance: No significant adverse effects.
Dropouts: 10 (did not discontinue breast feeding).
Other Endpoints:
-Bacteria group populations differed significantly after
formula-only feeding.
-There was an increase in bifidobacteria, decrease in
Bacteroides and C. coccoides in both the scGOS/lcinulin and scGOS/lc-inulin/AOS treatment groups, the
scGOS/lc-inulin/AOS had a significant increase over the
scGOS/lc-inulin group.

Source: Not reported.

n = 82

Formula with scGOS


and lc-inulin or
formula with scGOS,
lc-inulin and partially
unsaturated and
methylated AOS
Source: Numico

56

Results of GOS/lc-inulin Ingestion

000072

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Study Design &
Objective

Reference

Evaluate effects of
GOS/lc-inulin on
bifidobacteria.

Moro et al.
2002; 2005

Treatment
Population a

Test Substance and


Dose

Treatment
Duration

Formula with 3.6 g


GOS/L & 0.4 g lcinulin/L
n = 27-33; 6.3-7.2 d
postpartum

Randomized,
placebo-controlled

or

4 wk

7.2 g GOS/L & 0.8 g


lc-inulin/L
Source: Numico

Moro et al.
2006;
Arslanoglu
et al. 2007

Study the effect of


GOS/lc-inulin on the
incidence of atopic
dermatitis and
infections in high risk
infants.
Prospective, doubleblind, randomized,
placebo controlled

Safety Data

n = 102

Formula with 7.2 g


GOS/L & 0.8 g lcinulin/L
Source: Numico

57

< 6 mo

Results of GOS/lc-inulin Ingestion


Growth:
-No effect on growth.
Tolerance:
-Dose-dependent decrease in stool hardness, and
increase in stool frequency in high-dose group.
-No effect on incidence of crying, regurgitation, or
vomiting.
-No diarrhea occurred during study.
Dropouts:
-None reported.
Other Endpoints:
-Dose-dependent increase in fecal bifidobacteria, and
increase in fecal lactobacilli (not dose-dependent).
-Dose-dependent decrease in fecal pH.
-GOS and FOS detected in feces.
Growth: Data not measured/reported.
Tolerance:
-Increase in stool frequency and softer stool consistency
in treatment group.
-Lower reports of regurgitation and crying in treatment
group; incidence of vomiting same between groups.
Dropouts:
-53; 26 control (21 started breastfeeding, 5 other
reasons); 27 treatment (22 started breastfeeding, 5
other reasons).
Reasons for dropout not significant between groups.
Other Endpoints:
-Increase in bifidobacteria count in treatment group.
-Decrease in incidence of dermatitis in treatment group
compared to control group.
-Severity of dermatitis unaffected by diet.
-Fewer episodes of all types of infections, and lower
incidence of recurring infections.

000073

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Reference

Panigrahi
et al. 2008

Study Design &


Objective
To evaluate the longterm colonization
potential
Lactobacillus
plantarum in neonatal
guts.

Treatment
Population a

n = 31;
1-3 days post birth;
all cesarean
section.

Double-blind,
placebo-controlled,
randomized.

Piemontes
e et al.
2011

Puccio et
al. 2007

To assess the
tolerance and safety
of the prebiotics in
infants.

Prospective,
randomized, doubleblinded, referencecontrolled, parallelgroup clinical trial.

Safety Data

Receive either an
oral preparation of L.
plantarum and FOS
or dextrose saline
placebo for 7 days.

Treatment
Duration

6 months

Source: Not reported.

n = 1130;
age 8 wks

Double-blind,
placebo-controlled,
randomized.
To assess the safety
of an experimental
probiotic formula
containing B. longum
culture and
GOS:FOS.

Test Substance and


Dose

6.8 g/L of scGOS/lcinulin (9:1 ratio) and


1.2 g/L pAOS in a
non-hydrolyzed cows
milk-based formula

n = 138

Source: Nestl
Nutrition

58

Growth: No difference in % increase in weight.


Tolerance: No significant adverse effects reported.
Dropouts: 2 withdrawals; 1 protocol violation.
Other Endpoints:
-The number of bacteria species was significantly
higher.

1 year

Growth: No difference in growth between groups.


Tolerance: No difference in adverse events between
formula groups.
Dropouts: 129 total; similar across groups; due to
withdrawal, occurrence of disease, intolerance of
formula, change of formula.
Other Endpoints:
-No difference in mean weight, length, head
circumference, skin fold thickness, arm circumference
gains, & stool frequency between groups.

7 months

Growth: No difference in weight gain, length and head


circumference.
Tolerance: No study formula-related adverse events.
Dropouts: 41 (no statistical difference between control
and treatment groups).
Other Endpoints:
-Reduced incidence of constipation.
-Increased stool frequency.

Source: Nutricia

Experimental
probiotic formula
containing B. longum
culture (2 x 107 cfus)
and GOS/FOS (4 g/L)

Results of GOS/lc-inulin Ingestion

000074

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Study Design &
Objective

Reference

Raes et al.
2010

To evaluate the effect


of scGOS/lc-inulin on
basal immune
parameters.

Treatment
Population a

n = 215

Randomized, doubleblind, placebocontrolled.

Rao et al.
2009

Systematic review of
randomized control
studies.

Rinne et al.
2005

Assess the
quantitative and
qualitative differences
of microflora in
infants following
GOS/lc-inulin intake
of formula.

Safety Data

Test Substance and


Dose
Formula with 0.6
g/100 mL scGOC/lcinulin;
unsupplemented
formula or HM ad
libitum throughout the
study period.

Treatment
Duration

26 weeks

Results of GOS/lc-inulin Ingestion


Growth: Not reported.
Tolerance: No negative side effects from the prebiotics.
Dropouts: 28; no explanation reported.
Other Endpoints:
-No difference in supplemented groups in C-reactive
protein, total IgA, IgG, IgM & IgE.
-Breast fed group had higher white blood cell counts.

Source: Danone

Varied by study.
Not applicable.

n = 8; >8 wk
Infants had at least
one close relative
afflicted with atopic
eczema, allergic
rhinitis or asthma.

Sources: Varied by
study.

Not
applicable.

Formula with GOS/lcinulin


Dose not reported
Source:
Omneo, Nutricia,
Cuijk

59

4+ mo

Growth: Not evaluated.


Tolerance: All studies reviewed reported that the
substances were well tolerated.
Dropouts: Not applicable.
Other Endpoints:
-6 of 9 studies reported increased bifidobacteria counts.
-of 3 studies, all 3 reported higher stool frequency
(similar to breast fed, not an adverse outcome).
-Inadequate evidence regarding the prevention of atopic
diseases.
Growth:
-No differences in length or weight gain.
Tolerance:
-Clinical characteristics comparable among groups at 6
and 12 mo of age.
Dropouts:
-None reported.
Other Endpoints:
-Increase in fecal bifidobacteria after 4 mo; no effects on
fecal Clostridia, Lactobacilli/enterococci or Bacteroides.
-Lower occurrence of atopic eczema in breastfed and
pre/pro-biotic groups at 10 months.

000075

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Study Design &
Objective

Reference

Rigo et al.
2001

Salvini et
al. 2011

Study the effects of a


hydrolyzed protein, palmitic acid formula
with GOS/lc-inulin on
growth, miceral
accretion and
intestinal flora
development.

To determine if the
prebiotic
supplementation
could result in longterm colonization of
bifidobacteria.
Double-blind,
randomized, placebocontrolled.

Safety Data

Treatment
Population a

n = 14; at birth

Test Substance and


Dose

Formula with 3.6 g


GOS/L & 0.4 g lcinulin/L

Treatment
Duration

2 mo

Source: Numico

n = 20;
infants who were
not breast fed and
born to hepatitis C
virus- infected
mothers.

8 g/L scGOS/lc-inulin
supplemented
formula.
Source: Danone

60

6 months

Results of GOS/lc-inulin Ingestion


Growth:
-Growth parameters for study infants followed standard
infant growth curve.
Tolerance:
-Investigators reported feeding tolerance as being
excellent.
-Adequate volume formula intake observed throughout
study, although milk intakes were lower than those
previously observed in infants consuming a standard
formula.
Dropouts:
-One drop-out due to gastro-esphagial reflux.
Other Endpoints:
-Increase in the percentage of endogenous
bifidobacteria observed through 68 days of age (when
final measurement was taken).
Growth: Not evaluated.
Tolerance: Well tolerated; no adverse events.
Dropouts: 2; 1 control (non-compliance); 1 treatment
(unknown reason).
Other Endpoints:
-No change in fecal pH, serum IgE and IgG levels.
-Higher counts of bifidobacteria and lactobacilli.

000076

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Study Design &
Objective

Reference

Savino et
al. 2003

Study effect of a
partially hydrolyzed
formula with GOS/lcinulin, palmitic acid &
low levels of lactose
on formula-fed infants
with G.I. problems.

Treatment
Population a

n=604; mean 1.35


mo

Test Substance and


Dose

Formula with 7.2 g


GOS/L & 0.8 g lcinulin/L

Treatment
Duration

2 weeks

Source: Not indicated

Observational study.

Savino et
al. 2006

Study effects of a
formula containing
GOS/lc-inulin in
infants with colic.
Prospective
randomized
controlled study

Safety Data

n = 96; < 16 wk

Formula with 7.2 g


GOS/L & 0.8 g lcinulin/L and 41%
palmitic acid
Source: GOS Friesland Foods
Domo; Orafti

61

2 weeks

Results of GOS/lc-inulin Ingestion


Growth:
-Data not reported/measured.
Tolerance:
-Fewer regurgitation problems
-Increase in daily number of stools
-Fewer daily colic episodes
Dropouts:
-None reported.
Other Endpoints:
-91% positive subjective rating from parents.
-95% positive subjective rating from pediatricians.
Growth:
-Data not measured/reported.
Tolerance:
-Reduction in crying episodes after 7 and 14 days.
Dropouts:
-None reported; exclusion from analysis occurred with
infants from both groups due to not meeting inclusion
criteria and loss to follow-up.
Other Endpoints:
-Not relevant.

000077

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Study Design &
Objective

Reference

Schmelzle
et al. 2003

Study the nutritional


efficacy and
bifidogenic
characteristics of a
formula containing
partially hydrolyzed
whey protein, high palmitic acid content,
starch and
GOS+FOS vs. a
standard formula.

Treatment
Population a

n = 49; < 2 wk
postpartum

Test Substance and


Dose

Two treatments:
Standard formula,
formula with 7.2 g
GOS/L & 0.8 g lcinulin/L

Treatment
Duration

10 weeks

Source:
Omneo/Comformil,
Numico

Randomized, doubleblind study

Scholtens
et al.
2006b

Study effects of solid


foods with GOS+FOS
on microflora in term
infants.
Randomized, double
blind, placebocontrolled
intervention trial

Safety Data

Intent-to-treat:
n = 19; 16 wk
Per protocol
analysis:
n = 11; 16 wk (data
presented in paper)

baby food with 4.05 g


GOS/d & 0.45 g lcinulin/d; divided into 3
equal servings
6 weeks
Sources:
GOS - Borculo
Domo;
FOS - Orafti

62

Results of GOS/lc-inulin Ingestion


Growth:
-At 6 wk, girls had larger weight gains and at 12 wk girls
had larger head circumference gains.
-Boys had larger total skinfold thickness gains.
-Weight and length gains were similar at 12 wk and
groups were in-line with published growth curves.
Tolerance:
-Softer stools.
-Formulas were well tolerated.
Dropouts:
-None reported.
Other Endpoints:
-Decreased energy intake from formula.
-Increased fecal bifidobacteria compared to baseline,
and higher % bifidobacteria vs control group at 6 wk.
-No effects on serum total protein, albumin or urea; no
clinically significant differences in prealbumin and amino
acid values.
Growth:
-Data not measured/reported.
Tolerance:
-Non-significant decrease in stool consistency and
increase in frequency compared to control for baseline
measurements of 12 infants from per-protocol group.
Dropouts:
-11 infants withdrawn from final evaluation due to noncompliance; 1 infant withdrawn due to age > study
protocol.
Other Endpoints:
-Increase in % fecal bifidobacteria; no effect on fecal
bifidobacteria count (no effect in intent-to-treat analysis).
-No effect on change in fecal pH, total SCFA, or %
acetate, propionate, or sum of valerate, isovalerate and
isobutyrate; decrease in % butyrate.

000078

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Reference

Scholtens
et al. 2008

Schouten
et al. 2011

Study Design &


Objective
To evaluate the effect
of prebiotics on
intestinal microbiota
composition and fecal
sIgA.
Randomized, doubleblind, placebocontrolled.
To measure the
plasma kappa and
lambda Ig-fLC
concentrations in
infants at risk of
developing allergic
disease.

Treatment
Population a

n = 215

Stam et al.
2011

Double-blind,
placebo-controlled,
randomized.

Safety Data

6 g/L scGOS/lc-inulin
(9:1 ratio)

Treatment
Duration

Results of GOS/lc-inulin Ingestion

First 26
wks of life.

Growth: Not reported.


Tolerance: similar percentage of adverse events
between groups (42 47%); descriptions of adverse
events not reported.
Dropouts: 28; no reasons provided.
Other Endpoints:
-Prebiotic group had higher percentage of bifidobacteria,
higher concentration of sIgA.

6 months

Growth: Data not measured/reported.


Tolerance: See Moro et al. (2006).
Dropouts: See Moro et al. (2006).
Other Endpoints:
-Plasma of infants suffering from atopic dermatitis had
higher total kappa and lamba Ig-fLC than infants without
atopic dermatitis.
-Infants receiving the scGOS/lc-inulin supplemented
formula had significantly lower Ig-fLC levels (p < 0.001).

1 year

Growth: No difference in growth (see Piemontese et al.


2011).
Tolerance: See Piemontese et al. 2011.
Dropouts: See Piemontese et al. 2011.
Other Endpoints:
-No difference in Haemophilus influenza type b antibody
levels.
-No difference in tetanus immunization response (IgG
levels).

Source: Numico

n = 74
(see Moro et al.
2006)

Hypoallergenic whey
formula with 8 g/L
scGOS/lc-inulin
(9:1 ratio)
Source: Numico

Prospective, doubleblind, randomized,


placebo controlled.
To evaluate the effect
of the prebiotics on Ig
responses to
vaccinations.

Test Substance and


Dose

n = 164;
(subset of Grber
et al. 2010 and
Piemontese et al.
2011)

6.8 g/L of scGOS/lcinulin (9:1 ratio) and


1.2 g/L pAOS in a
non-hydrolyzed cows
milk-based formula
Source: Nutricia

63

000079

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Reference

Vaisman et
al. 2010

Study Design &


Objective
To determine if
prebiotics modulate
inflammatory
processes of acute
diarrhea.
Double-blind,
placebo-controlled,
randomized,
prospective.

van der Aa
et al. 2010

To evaluate the effect


of synbiotics on the
severity of atopic
dermatitis in infants.
Double-blind,
placebo-controlled,
randomized.

van der Aa
et al. 2011

To investigate the
effect of synbiotics on
the prevalence of
asthma-like
symptoms in infants
with atopic dermatitis.
Double-blind,
placebo-controlled,
randomized.

Safety Data

Treatment
Population a

n = 104;
age = 9 24
months; suffering
from acute diarrhea

n = 90;
age < 7 mo; with
atopic dermatitis

n = 75;
mean age = 17
mos; with atopic
dermatitis

Test Substance and


Dose

6 g/day of 80%
GOS/lc-inulin (9:1
ratio), 20% pAOS

scGOS/lc-inulin (9:1
ratio) mixture in a
hydrolyzed wheybased formula that
included
Bifidobacterium breve
9
M-16V (1.3x10
CFU/100mL)
Source: Nutricia
scGOS/lc-inulin (9:1
ratio) mixture in a
hydrolyzed wheybased formula that
included
Bifidobacterium breve
9
M-16V (1.3x10
CFU/100mL)

Treatment
Duration

Results of GOS/lc-inulin Ingestion

12 days

Growth: Data not measured/reported.


Tolerance: Data not measured/reported.
Dropouts: 15; failed to receive intervention.
Other Endpoints:
-No difference in stool characteristics.
-No difference in cytokine levels, except TNF- (but not
clinically relevant).

12 weeks

Growth: Not evaluated.


Tolerance: Well tolerated no adverse events.
Dropouts: 8; none related to treatment.
Other Endpoints:
-No effect on severity of atopic dermatitis or serum IgE.
-Increased bifidobacteria, reduced pathogenic bacteria.
-Lowered fecal pH, increased fecal lactate, lowered
SCFAs.
-Fewer infants with dry stools, less constipation.

12 weeks

Growth: Not evaluated.


Tolerance: Well tolerated no adverse events.
Dropouts: 8; none related to treatment.
Other Endpoints:
-Lower prevalence of frequent wheezing & wheezing
and/or noisy breathing apart from colds.
-No difference in IgE levels.

Source: Nutricia

64

000080

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Reference

van der Aa
et al. 2012

Study Design &


Objective
To determine the
effects of synbiotics
on atopic markers,
cytokine production,
and circulating T cells
in infants with atopic
dermatitis.
Double-blind,
placebo-controlled,
randomized.
To measure the
modulation of
response to cows
milk protein and
vaccinations by
prebiotics.

van Hoffen
et al. 2009

van
Stuijvenber
g et al.
2011

Prospective, doubleblind, randomized,


placebo-controlled.
To assess the effect
of prebiotics on the
number of fever
episodes in infants.
Randomized, doubleblind, placebocontrolled.

Safety Data

Treatment
Population a

n = 90;
age < 7 mos; with
atopic dermatitis

Test Substance and


Dose
scGOS/lc-inulin (9:1
ratio) mixture in a
hydrolyzed wheybased formula that
included
Bifidobacterium breve
9
M-16V (1.3x10
CFU/100mL)

Treatment
Duration

12 weeks

Results of GOS/lc-inulin Ingestion

Growth: Not evaluated.


Tolerance: Well tolerated no adverse events.
Dropouts: 8; none related to treatment.
Other Endpoints:
-No difference in ex vivo cytokine production by
peripheral blood mononuclear cells.
-No difference in circulating regulatory T cell
percentages.

Source: Nutricia

n = 84; (subset of
Moro et al. 2006)

Hypoallergenic whey
formula with 8 g/L
scGOS/lc-inulin
(9:1 ratio)
Source: Numico

n = 830
(subset of Grber
et al. 2010 and
Piemontese et al.
2011)

6.8 g/L of scGOS/lcinulin (9:1 ratio) and


1.2 g/L pAOS in a
non-hydrolyzed cows
milk-based formula
Source: Nutricia

65

3 months
postvaccination
(6 months
of age)

Growth: Data not measured/reported.


Tolerance: See Moro et al. (2006).
Dropouts: See Moro et al. (2006).
Other Endpoints:
-Significant reduction in plasma IgE, IgG1, IgG2, IgG3
and cows milk protein-specific IgG1.
-No difference in IgG4 and diphtheria, tetanus, & poliospecific Ig.

1 year

Growth: No difference in growth (see Piemontese et al.


2011).
Tolerance: See Piemontese et al. 2011.
Dropouts: See Piemontese et al. 2011.
Other Endpoints:
-No difference in the number of fever episodes between
groups.

000081

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-4. Studies of 90% GOS/10% Long-Chain Inulin in Term Infant Formulas and Weaning Foods
Reference

VeeremanWauters et
al. 2011

Vivatvakin
et al. 2010

Study Design &


Objective
To assess safety,
tolerance, and
bifidogenic effects of
different doses of
prebioticsupplemented
formula
Prospective,
randomized, doubleblind, controlled.
To investigate the
effects of prebiotics
on gastrointestinal
comfort in infants.
Prospective,
randomized, doubleblind, placebocontrolled.

Treatment
Population a

n = 110

Test Substance and


Dose
Formula with 0.4 g/dL
of a 50:50 mixture of
lc-inulin and FOS;
formula with 0.8 g/dL
lc-inulin/FOS; or
formula plus 0.8 g/L
GOS/FOS (9:1 ratio).

Treatment
Duration

28 days

Source: Beneo

n = 144
(completed);
age = 30 days

Whey-based formula
containing LCPUFAs
and 4 g/L of a
GOS/FOS mixture
(9:1 ratio).

3 months

Results of GOS/lc-inulin Ingestion


Growth: No differences between groups.
Tolerance: No serious adverse events;
gastroesophageal reflux, regurgitation, hunger; similar
across groups.
Dropouts: 33; similar across groups; not attributed to
prebiotic use.
Other Endpoints:
-0.8 g/L supplementation had bifidobacteria counts
comparable to breast fed group.
-0.8 g/L supplementation had softer stools, consistency
comparable to breast fed group.
Growth: No difference in anthropometric
measurements.
Tolerance: No difference in adverse events between
groups.
Dropouts: 55; no difference across groups.
Other Endpoints:
-Fewer hard stools, more soft stools.
-No difference in bacterial composition, gastric emptying
time.

AOS = acidic oligosaccharides; cfu = colony forming units; d = day; f = female; FOS = fructooligosaccharides; g = grams; GOS = galacto-oligosaccharides; HDL = highdensity lipoprotein; HM = human milk; Ig-fLC = immunoglobulin free light-chain; lc-inulin = long-chain inulin; LDL = low-density lipoprotein; m = male; pAOS = pectin-derived
acidic oligosaccharides (identical ingredient to AOS); sc = short-chain; SCFA = short-chain fatty acids; SCORAD = SCORing Atopic Dermatitis; sIga = secretory
immunoglobulin A; wk = week.
a
Age at enrollment into the study.

(The remainder of this page intentionally left blank.)

Safety Data

66

000082

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

5.7.3 Water Balance


In 2003, the EU Scientific Committee on Food assessed the safety of adding GOS/FOS to infant
formula in Report of the Scientific Committee on Food on the Revision of Essential
Requirements of Infant Formulae and Follow-on Formulae (SCF 2003). The evaluation noted
the potential adverse effects on infant water balance for hypernatremia due to a high dietary
renal solute load. The Committee found that based on the information available at the time the
addition of up to 0.8 g/100mL 9:1 GOS:FOS did not demonstrate adverse effects, particularly to
growth or markers of water balance. EFSA considered the issue and in a 2004 opinion based
on five studies available at the time did not find the information sufficient to eliminate the
possibility of a dehydration risk.
Notably, EFSA considered loose stools an adverse event. In many of the studies in sections
5.7 and 5.8, researchers reported that infants fed combinations of GOS and FOS had softer or
looser stools (Boehm et al. 2003; Veereman-Wauters et al. 2011; Westerbeek et al. 2010) but
these were not considered adverse events. Other studies noted less constipation in the
prebiotic-fed groups and constipation could be considered an adverse event (Mihatsch et al.
2006; Puccio et al. 2007; Savino et al. 2006; Van der Aa et al. 2010). Still other researchers
reported less or no diarrhea (an adverse event) in the prebiotic-fed group (Mihatsch et al. 2006;
Moro et al. 2002; Vivatvakin et al. 2010) while several studies found no difference in stool
frequency or consistency (Modi et al. 2010; Piemontese et al. 2011; Scholtens et al. 2006b).
Food Standards Australia New Zealand (FSANZ) addressed the EFSA opinion directly in the
following excerpt from the Final Assessment Report Proposal P306 Addition of Inulin/FOS &
GOS to Food:
In regard to the use of other ratios of oligosaccharides, or inulin-derived substances
alone in infant formula, FSANZ has considered a concern expressed by EFSA relating to
water balance. The available evidence that oligofructose and inulin are fermented by
colonic microflora in formula fed infants as described above (presence of SCFA in stool,
in vitro stool studies, and evidence from adult toleration studies), reduces the concern
that water balance could be adversely affected by an increase in osmotic potential due
to undigested inulin-derived substances in the colon. In addition, a 12-week study in
term infants which indicated oligofructose at 3 g/L had no significant effect on growth,
blood chemistry, or reported adverse events, supports the safety of inulin-derived
substances in infant formula products. It has been shown that 8 g/L of GOS:long-chain
inulin is safe, levels as high as 25 g HMOs [human milk oligosaccharides]/L in breast
milk is safe, and inulin-derived substances are likely to be fermented to a similar degree
to GOS and HMOs, therefore, FSANZ concludes up to 8 g/L added inulin derived
substances will be safe for young infants. This is supported by a recent 28-day study on
up to 8 g/L of a 1:1 oligofructose and long-chain inulin combination. This conclusion
applies equally to the use of GOS alone or any ratio of GOS:inulin-derived substances
so long as the total level of oligosaccharides is no greater than 8 g/L (FSANZ 2008,
p160).
These issues is addressed in greater detail in FSANZ (2008) and in GRN 392, but to summarize
here, studies and opinions published since the EFSA opinion in 2004 support the safety of the

Safety Data

67

000083

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

prebiotic-supplementation in infant formula and do not suggest a risk of hypernatremic


dehydration.

5.8

Long-Chain Inulin and Galacto-Oligosaccharides in Preterm Infant Formulas

Thirteen clinical trials involving the administration of infant formulas containing added nondigestible carbohydrates comprised of 90% GOS and 10% FOS (lc-inulin) to preterm infants
were identified. The studies are summarized below and in Table 5-5.

5.8.1 Human Studies of Inulin and Oligofructose in Preterm Infant Formulas


In the trial conducted by Boehm and colleagues (2002), preterm infants who could tolerate a
formula volume of 80 mL/kg-bw/d (approximately 8 days postpartum) were randomly assigned
to consume a standard preterm formula or a formula supplemented with 10 g GOS/lc-inulin per
L formula (9 g GOS per L, 1 g lc-inulin per L). Fifteen infants consumed each formula for a
period of 28 days. A group of 12 breast-fed infants was followed as a reference group. At the
end of 28 days, fecal bifidobacteria was increased in the GOS/lc-inulin group as compared to
the control group (10.0 2.05 vs. 7.9 0.83 log 10 CFU/g wet stools) and levels in infants
consuming the supplemented formula were reported to be in the upper range of the reference
group of infants, which was 10.7 log 10 CFU/g wet stools (Boehm et al. 2003). No other
differences in fecal bacteria were observed. The consistency of stools from infants consuming
the supplemented formula was softer than the consistency of infants consuming the control
formula, and comparable to the stool consistency of breast-fed infants. At study completion, the
hardest stools were observed in the control group, which had a significantly higher score of
3.55 0.8 compared to the supplemented and reference groups which were 2.74 0.7 and
2.33 0.6, respectively (Boehm et al. 2003). Infants consuming the supplemented formula also
had a higher stool frequency as compared to infants in the control group, but comparable to the
frequency in breast-fed infants. Weight and length gain and incidence of crying, regurgitation,
and vomiting were reported to be similar among the formula groups. The GOS/lc-inulinsupplemented formula had no effect on plasma concentrations of calcium and phosphorus,
plasma activity of alkaline phosphatase, or urinary phosphate concentrations, though urinary
calcium concentrations in infants fed the supplemented formula tended to be higher than those
in the group fed the standard formula (1.63 1.15 mmol/L vs 0.93 0.72 mmol/L; p=0.055)
(Lidestri et al. 2003). In a subset of the infants, the investigators examined the sum of selected
pathogens in fecal samples (Knol et al. 2005b) and found that the sum of pathogens and
pathogens as a percentage of total bacteria were lower in fecal samples from infants fed the
supplemented formula as compared to the control formula.
In another clinical trial, 20 preterm infants were randomly assigned to consume a formula
containing 10 g GOS/lc-inulin per L (9 g GOS per L) or a standard formula (Mihatsch et al.
2006). The study formulas were prepared once a day by combining a standard formula with
sachets containing the oligosaccharide mixture or a placebo (maltodextrin). Infants consumed
the assigned formulas for a period of two weeks. Infants consuming the GOS/lc-inulinsupplemented formula had decreased stool viscosity, change in fecal transit time, and fecal pH.
During the trial the groups did not differ in clinical characteristics, formula intake or weight gain,
and adverse events such as weight loss, constipation, diarrhea, abdominal discomfort or
flatulence were not reported.

Safety Data

68

000084

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Indrio et al. (2009a) performed a double-blind, placebo-controlled study involving 49 preterm


infants with normal Apgar scores during their first week of life. Of the 49 infants, 17 were
exclusively breast fed, 12 received a placebo formula, 10 received a prebiotic-supplemented
formula with 0.8 g/dL scGOS/lc-inulin (9:1 ratio), and 10 received a probiotic-supplemented
formula (Lactobacillus reuteri at 1 x 108 cfus/day ). Indrio et al. (2009a) stated that the
objective of the study was to compare the effects of prebiotic, probiotic, and breast milk on GI
motility respect to a standard formula. The authors evaluated GI motility by non-invasive
cutaneous electrogastrography and ultrasound gastric emptying recording. Indrio et al. (2009a)
also recorded anthropometric measurements (weight, length, and head circumference)
throughout the study.
Indrio et al. (2009a) stated that no adverse events were reported. The authors found no
significant difference in the daily increase of body weight (average of 35 g/d). The authors
found that after the 30-d intervention period, gastric half-emptying time was faster in the
prebiotic, probiotic and breast milk groups compared to the placebo group. Indrio et al. (2009a)
stated that faster gastric emptying times may result in decreased retention times of luminal
nutrients. The authors suggest this may assist in the prevention of illness in preterms; however,
no data regarding subjects health during this study are presented and the hypothesized benefit
was not established. This study was of a very short duration with an unspecified formula and
unknown sources for the prebiotic and probiotic supplements.
Indrio et al. (2009b) reports a study very similar to Indrio et al. (2009a). Indrio et al. (2009b)
performed a double-blind, placebo-controlled, randomized feeding study with 20 preterm infants
for a period of 15 days. Ten infants received a maltodextrin-based placebo formula while ten
received a prebiotic-supplemented formula with 0.8 g/dL scGOS/lc-inulin (9:1 ratio). The
authors stated that Numico partially funded the study, but reported no conflicts of interest. The
authors performed the same analyses and report the same results as in Indrio et al. (2009a)
with respect to the prebiotic and placebo formulas (i.e., faster gastric half-emptying times and
comparable daily increase in body weight). The authors stated that no adverse events were
reported.
Modi et al. (2010) performed a randomized, double-blind, controlled trial that compared a
preterm formula that contained 0.8 g/100 mL scGOS/lc-inulin in a 9:1 ratio with an identical
formula that did not contain the scGOS/lc-inulin mixture. The authors stated objective was to
determine if the scGOS/lc-inulin supplemented formula, when used to supplement insufficient
maternal milk, improved enteral tolerance in preterms infants. The evaluated outcomes
included the primary outcome (PO) time to establish a total milk intake of 150 mL/kg/d a
principal secondary outcome (PSO) proportion of time between birth and 28-d discharge that
a total milk intake of 150 mL/kg/d was tolerated as well as growth, fecal characteristics,
gastrointestinal signs, necrotizing enterocolitis, and bloodstream infection. The authors
declared that Danone research provided the lead author, Neena Modi, an unrestricted research
donation and lecture honorarium. Danone Research also produced and supplied the formula
(Nutriprem 1). Of 160 preterm infants enrolled, 150 infants completed the study. Seventy nine
infants from the control group and 71 from the scGOS/lc-inulin group were available for
comparison at the end of the study. The enteral feeding began as early as possible within the
first 24 h of birth. A predefined protocol defined the rate of increase in milk volume: infants born

Safety Data

69

000085

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

28-w 6-d gestation began feeding at 0.5 mL/kg for every 2 h and advanced by 15 mL/kg/d to
15 mL/kg twice daily when 60 mL/kg was reached. Infants born 29 to 32 weeks 6-d gestation
were fed 15 mL/kg/d and increased to 30 mL/kg/d on day 2 and by 15 mL/kg twice daily from
then on. Breastfeeding was encouraged. The authors reported that infants were monitored
until 40 wk postmenstrual age or discharge home. There were no significant differences
between groups at the trial start.
Modi et al. (2010) found no significant differences in the PO or the PSO; however, the authors
note that increasing infant immaturity demonstrated differences in the PSO (2.9% improved
tolerance for infants of 28-w gestation and 9.9% for infants of 26-w gestation; P < 0.001).
Infants of > 31-w gestation has decreased or no benefit. The authors recognize that the
inclusion of maternal milk affected the ability to discern differences. Modi et al. (2010) reported
no statistically significant difference in weight gain, length, or head circumference; fecal flora; GI
signs; number of daily stools or stool characteristics. The authors found no significant
difference in cases of necrotizing enterocolitis (control = 1, scGOS/lc-inulin = 2; p = 0.6) or
episodes of bloodstream infection (control = 10, scGOS/lc-inulin = 9; p = 0.18). The authors
noted that the mechanism by which prebiotics influence enteral tolerance is through the
promotion of colonization of bifidogenic microbial species and increasing GI transit time.
However, Modi et al. (2010) did not find statistically significant differences in fecal flora or stool
characteristics. The authors note that the high intake of maternal milk by the study groups may
be the reason for a lack of differentiation. The authors reported six adverse events including
three deaths. None of the deaths were attributed to the study and only one of the events was
considered related to the trial (from the control formula group abdominal distension and
tenderness associated with a Streptococcal infection). Modi et al. (2010) concluded that the
scGOS/lc-inulin supplementation appears safe and may benefit enteral tolerance in preterm
infants.
Underwood et al. (2009) performed a randomized, blinded, placebo-controlled trial with
90 preterm infants (age < 7 d) to compare the effect of two prebiotic/probiotic supplement
products on weight gain, stool microbiota, and stool short chain fatty acid content. The infants
were placed on one of three groups and administered the one mL of the treatment twice daily
for 28 days or discharge, if earlier. Treatments consisted of a saline solution with lactobacilli
and bifidobacteria (5 x 108 organisms; n = 30), the same solution plus FOS (inulin; unknown
quantity; n = 31), and a dilute preparation of a placebo formula with no pro- or prebiotic activity
and minimal caloric value (n = 29). Underwood et al. (2009) found no differences in weight
gain, stool SCFA content, or lactobacilli colonization between groups. The authors observed
that the bifidobacteria content of the infants receiving the lactobacilli and bifidobacteria plus
FOS treatment had a significantly higher bifidobacteria content in their stools (Kruskal-Wallis
p = 0.011). The authors noted that there were no adverse reactions.
Westerbeek et al. (2010; 2011a; 2011b; 2011c) performed a series of studies based on a
randomized, double-blind, controlled trial that included 113 preterm infants with a gestational
age of < 32 weeks and/or a birth weight of < 1.5 kg. From day three of life to day 30, the infants
received either an enteral supplementation of a mixture of 80% scGOS/lc-inulin and 20% AOS
(1.5 g/kg/d; n = 55 enrolled, 43 completed) or a maltodextrin placebo (n = 59 enrolled,
51 completed). Infants received the treatments in breast milk or a preterm formula free of

Safety Data

70

000086

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

oligosaccharides. Danone Research provided the scGOS/lc-inulin/AOS and maltodextrin


powders. The authors reported that the baseline nutritional and patient characteristics did not
differ between groups.
In Westerbeek et al. (2010), the authors objective was to determine the effect of the prebiotic
mixture on serious infectious morbidity, feeding tolerance, and short-term outcome in preterm
infants. The authors evaluated incidence of serious infectious morbidity, time to full enteral
feeding, age when parenteral feeding was discontinued, occurrence of necrotizing enterocolitis,
growth, need for mechanical ventilation, presence of periventricular-intraventricular hemorrhage,
patent ductus ateriosus treated with ibuprofen, indomethacin and/or surgical ligation,
bronchopulmonary dysplasia, retinopathy of prematurity, age at discharge from neonatal
intensive care, age at discharge from the hospital, and death. Westerbeek et al. (2010) found
no statistically significant difference between the prebiotic group and the placebo group in any of
these outcomes except one. The authors found that the group receiving the prebiotic mixture
had a lower incidence of bronchopulmonary dysplasia (0% vs. 23%, p = 0.003). Westerbeek et
al. (2010) concluded that the prebiotic supplementation by scGOS/lc-inulin/AOS did not
significantly reduce the risk of serious infectious morbidity in preterm infants though the authors
did observe a trend toward decreased serious infectious morbidity that should be investigated in
a larger study.
In Westerbeek et al. (2011a), the authors objective was to determine the effect of enteral
supplementation of the prebiotic mixture on fecal interleukin-8 (f-IL-8) and fecal calprotectin
(f-calprotectin) in preterm infants and to determine host- and treatment-related factors
associated with f-IL-8 and f-calprotectin in these preterm infants. The authors measured IL-8
and calprotectin in fecal samples taken from the diaper at 48 h after birth, postnatal days 7,
14 and 30. The authors observed that f-IL-8 and f-calprotectin were strong correlated at all time
points (p < 0.001) but they did not observe a difference between the prebiotic supplemented
group and the placebo group. Westerbeek et al. (2011a) concluded that the enteral
supplementation of the prebiotic mixture did not affect f-IL-8 and f-calprotectin.
In Westerbeek et al. (2011b), the authors objective was to determine the effect of the prebiotic
mixture on intestinal permeability in the first week of life in preterm infants and to determine
host-and treatment-related factors associated with intestinal permeability in these preterm
infants. The authors measured intestinal permeability by a sugar absorption test preformed at
approximately 36 15 h after birth and days 4 and 7 post-birth. Westerbeek et al. (2011b)
observed that while decreased intestinal permeability was correlated with increased body weight
(p = 0.002), the difference between the prebiotic supplemented group and the placebo group
was not significantly different. Westerbeek et al. (2011b) concluded that the enteral
supplementation of the prebiotic mixture did not enhance the postnatal decrease in intestinal
permeability in preterm infants in the first week of life.
In Westerbeek et al. (2011c), the authors objective was to determine the effect of the prebiotic
mixture on stool viscosity, stool frequency and stool pH in preterm infants. The authors
observed lower stool viscosity (p = 0.03) and lower stool pH (p = 0.009) at day 30 in the
prebiotics group but no significant difference in stool frequency.

Safety Data

71

000087

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Results from these thirteen clinical trials provide additional evidence that the addition of a
GOS/lc-inulin mixture in a 9:1 ratio to infant formula for preterm infants is well tolerated over the
2 to 12 week study periods. These preterm infants were closely monitored throughout the
studies, and no adverse effects were observed on measures of growth, fluid balance, formula
intake, or stool characteristics. The absence of reports of adverse effects in this highly sensitive
and closely monitored study population provides strong evidence for the suitability of GOS and
lc-inulin in a 9:1 ratio in infant formulas.

(The remainder of this page intentionally left blank.)

Safety Data

72

000088

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-5. Studies of 90% GOS/10% Long-Chain Inulin in Preterm Infant Formula
Reference

Boehm et al.
2002 and
2003; Lidestri
et al. 2003;
Knol et al.
2005b

Indrio et al.
2009a/b

Study Objective &


Design

Investigate effects
of preterm formula
containing GOS/lcinulin on fecal flora
and stool
characteristics of
infants born at 32
wk gestational age.

Study effect of
GOS/lc-inulin added
to an enteral milk
formula on the
maturation of
gastrointestinal
motility and
permeability in
preterm infants.

Treatment
Population

n = 15;
age = 7.9 - 8.3
d

Test Substance
and Dose

Preterm formula
with 9 g GOS/L &
1 g lc-inulin/L

Treatment
Duration

Results of GOS/lc-inulin ingestion*

28 d

Growth:
-No effect on weight or length gains.
Tolerance:
-Softer stool consistency and increased stool frequency.
-No effect on incidence of crying, regurgitation, vomiting.
Dropouts:
-None reported.
Other Endpoints:
-Increase in fecal bifidobacteria; no effects on lactobacilli, E. Coli.,
Clostridium spp. Bacteroides, Enterobacter, Citrobacter, Proteus,
Klebsiella and Candida.
-No effects on plasma concentrations of calcium and phosphorus or
plasma activity of alkaline phosphatase; trend toward higher calcium
concentrations in urine (p=0.055).
-Decrease in fecal counts and % of total selected pathogens (sum of
Staphylococcus aureus, S. epidermidis, S. haemolyticus,
Pseudomonas aeruginosa, Enterobacter, Klebsiella, Proteus,
Streptococcus group B, Clostridium difficile, Bacillus subtilis,
Acinetobacter).

Source: Numico
(now Danone)

n = 49;
GA = ~34
weeks

Preterm formula
with 7.2 g GOS/L
& 0.8 g lc-inulin/L
30 days
Source: Numico

Growth: No difference in daily increase of body weight.


Tolerance: No adverse events reported.
Dropouts: None reported.
Other Endpoints:
-Faster gastric emptying time.

Double-blind,
randomized,
controlled, parallel.

Safety Data

73

000089

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-5. Studies of 90% GOS/10% Long-Chain Inulin in Preterm Infant Formula
Reference

Mihatsch et al.
2006

Study Objective &


Design
Evaluate feeding
tolerance in preterm
infants given
formula containing
GOS/lc-inulin

Treatment
Population

Modi et al.
2010

Double-blind,
randomized,
controlled, parallel

Safety Data

Treatment
Duration

preterm formula
with 9 g GOS/L &
1 g lc-inulin/L
n = 10;
age = mean 42
d postpartum

Double-blind,
controlled
Study enteral
tolerance of
GOS/lc-inulin
formula in preterm
infants whose
mothers milk is in
shortage or
unavailable.

Test Substance
and Dose

n = 160;
age < 1 d

14 days
Source: Milupa
GmbH (fully
owned subsidiary
of Danone)
Nutriprem
formula with 7.2 g
GOS/L & 0.8 g lcinulin/L
~9 wk

Source: Numico

74

Results of GOS/lc-inulin ingestion*


Growth:
-No effects on daily formula intake or daily weight gains.
Tolerance:
-Decrease in stool viscosity, decreased change in gastrointestinal
transit time (decrease of 6 d vs. increase of 9 d in control group).
Dropouts:
-No adverse effects observed.
Other Endpoints:
-Lower fecal pH.
Growth: No difference between groups in mean weight gain,
increase in body length and head circumference.
Tolerance: Intake levels did not differ between study groups.
Dropouts: 10;
-6 infants withdrawn due to parental request (2 in control, 4 in
treatment group).
-5 infant deaths. 2 in control group, 3 in GOS/inulin group.
-For all adverse events, possible or presumable relationship to
treatment was excluded by clinician.
Other Endpoints:
-No difference in fecal flora, gastrointestinal signs or indices of fluid
balance, or in daily number of stools and stool characteristics.

000090

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-5. Studies of 90% GOS/10% Long-Chain Inulin in Preterm Infant Formula
Reference

Underwood et
al. 2009

Study Objective &


Design

To compare the
effect of prebiotics
& probiotics on
weight gain, stool, &
stool SCFAs in
premature infants.

Treatment
Population

n = 90

Randomized,
blinded, placebocontrolled trial

Westerbeek et
al. 2010;
2011a/b/c

To evaluate the
effect of prebiotics
on infections,
intestinal
inflammation,
intestinal
permeability, stool
viscosity, stool
frequency, stool pH.
Randomized,
double-blind,
controlled trial.

Test Substance
and Dose
Supplement
containing 2
lactobacillus
species plus FOS
(Culturelle); or a
supplement
containing
several species
of lactobacilli and
bifidobacteria
plus FOS
(ProBioPlus);
twice daily.

Treatment
Duration

Results of GOS/lc-inulin ingestion*

28 days or
discharge

Growth: No significant effect on growth.


Tolerance: No adverse reactions were noted.
Dropouts: None reported.
Other Endpoints:
-64% of infants receiving ProBioPlus became colonized with
bifidobacteria, compared to 18% of infants receiving Culturelle and
27% of infants receiving placebo.
-No differences were noted between groups in colonization rates for
lactobacilli, Gram-negative enteric bacteria or staphylococci.
-Bifidobacteria content in the stools of the infants receiving
ProBioPlus was higher than in the infants receiving Culturelle or
placebo.
-No significant differences in stool short chain fatty acid content were
detected between groups.

28 days

Growth: No significant effect on growth.


Tolerance: No adverse reactions were noted. No effect on feeding
tolerance.
Dropouts: None reported.
Other Endpoints:
-No reduction in risk of serious infectious morbidity, NEC, or other
complications.
-No effect on fecal interleukin-8 or fecal calprotectin.
-No effect on postnatal decrease in intestinal permeability.
-Decreased stool viscosity and decreased stool pH.
-No effect on stool frequency.

Source:
Culturelle ConAgra;
ProBioPlus UASLabs

n = 113;
GA < 32 weeks

enteral
supplementation
of 80%
scGOS/lc-inulin
and 20% AOS
(1.5 g/kg/d)
Source: Danone

AOS = acidic oligosaccharides; cfu = colony forming units; d = days; f = female; FOS = fructooligosaccharides; g = grams; GA = gestational age; GOS = galactooligosaccharides; HDL = high-density lipoprotein; HM = human milk; lc-inulin = long-chain inulin; LDL = low-density lipoprotein; m = male; NEC = necrotizing enterocolitis;
sc = short-chain; SCFA = short-chain fatty acids; wk = weeks; y = years.

Safety Data

75

000091

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

5.9 Use of Inulin and Oligofructose in Medical Foods


5.9.1 Potential Role of Fiber in Medical Foods
Medical foods are administered to many chronically ill individuals in order to improve nutritional
parameters (Cockram et al. 1998). Until recently, most enteral foods were intentionally
produced without added fiber, in part due to concerns about tube obstruction and in part to
provide rest for the intestinal tract (Lochs et al. 2006). However, individuals receiving fiber-free
medical foods were observed to have significantly prolonged gut transit times (Silk 2001) and
significant decreases in anaerobic bacteria (Schneider et al. 2000), leading to diarrhea,
constipation, and bloating. It is now recognized that consumption of fiber-containing enteral
formulae and tube feeds by patients improves stool consistency, gut function, and control of
glycemic and lipid parameters. The addition of fiber to enteral formulas therefore may have
important clinical benefits (Elia et al. 2007).
Soy polysaccharides were the first fiber source added to medical foods (Green 2001). Types of
fiber currently added to medical foods include soy polysaccharides, cellulose, resistant starch,
guar gum, acacia fiber, pectin, lignin, inulin, FOS, and other products. Tolerance to enteral
formulas containing soy polysaccharides has been established in a variety of patient
populations (Green 2001). However, data on tolerance to enteral formulas containing lc-inulin
or related 2-1 fructan products are more limited, and most of the available data on tolerance
have been obtained in studies testing a combination of fiber products (Table 5-6).

5.9.2 Tolerance of Inulin and Oligofructose in Medical Foods


Tolerance of 2-1 fructan products added to medical foods has been demonstrated in four
studies of chronically ill patients. The tolerance of lc-inulin in a supplemental medical food has
been studied in a population of non-symptomatic, antiretroviral-nave HIV seropositive adults
(Gori et al. 2011). Gori et al. (2011) administered supplements containing GOS/lc-inulin+pectin
hydrolysate-derived AOS in a 9:1:10 ratio in a randomized, double-blind and placebo-controlled
Phase I, multi-center study. The supplements were distributed to patients as individual sachets
containing lc-inulin, GOS, pectin hydrolysate, inert sugars and maltodextrin in powder form.
Subjects were instructed to dissolve the powder in water or juice or to mix it with yogurt prior to
consumption three times each day. Fifty seven patients were enrolled in the study. Patients
consumed a total of either 6.75 g GOS, 0.75 g lc-inulin, and 7.5 g pectin hydrolysate per day
(single dose group); 13.5 g GOS and 1.5 g lc-inulin, and 15 g pectin hydrolysate (double dose
group); or maltodextrin (placebo group). The daily total intakes were consumed in three equal
portions. The treatment interventions lasted 12 weeks. During the study, participants
completed questionnaires in which they ranked seven indices of gastrointestinal tolerance
(0 = no complaints, 1 = mild, 2 = moderate, 3 = severe), and total scores were calculated as a
measure of overall gastrointestinal tolerance. The seven indices included nausea, burping,
abdominal distension, flatulence, diarrhea, constipation, and urgent need for defecation.
Data from 51 patients were included in the intent-to-treat analysis (single dose, n=16; double
dose, n=18; placebo group, n=17). After 4 and 12 weeks of consumption, the overall
gastrointestinal tolerance reported by patients in the single dose group did not differ from
tolerance reported by the placebo group. Patients in the double dose group reported
significantly higher overall gastrointestinal distress at both 4 and 12 weeks versus the placebo
group (3.8 versus 0.8 and 2.6 versus 0.6, respectively; p < 0.025).

Safety Data

76

000092

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

At week 4, participants in the single and double dose groups were more likely to report the
presence of abdominal distension than participants in the placebo group (50% and 55.6%
versus 10.5%). At week 12, the presence of diarrhea was significantly greater in the double
dose group as compared to the placebo group (43.8% versus 0%); the presence of an urgent
need to defecate was also greater in the double dose group as compared to the placebo group
(31.3% versus 0%).
Flatulence and abdominal distension received the highest scores (i.e., the most complaints)
from patients in both the single and double dose groups after 4 weeks of treatment. At 4 weeks,
abdominal distension scores were significantly higher in both treatment groups (1.0 in each
group) as compared to the placebo group (0.11). Flatulence scores in the single and double
dose groups were 0.83 and 1.44, respectively; the score in the double dose group was
significantly different from the placebo group (0.32). Following 12 weeks of consumption,
participants in the double dose group reported significantly higher mean scores for diarrhea and
urgent need for defecation as compared to the placebo group (0.69 versus 0 and 0.56 versus
0). Severe diarrhea was reported by two participants in the high dose group during the feeding
period (one at day 3 and one at week 12) and by one participant in the single dose group (week
4). No participants in the placebo group reported moderate or severe diarrhea during the study.
At week 12, patients consuming the GOS/lc-inulin+acidic oligosaccharides had increased
proportions of fecal bifidobacteria and decreased fecal pathogenic loads.
Results from these assessments of tolerance indicate that a supplemental medical food
containing a total of 7.5 or 15 g of GOS/lc-inulin in a 9:1 ratio of GOS to lc-inulin (along with
7.5 or 15 g pectin hydrolysate) is generally well tolerated in a population of HIV positive adults.
The prebiotic-containing supplement was most commonly associated with flatulence and
abdominal distension after approximately one month of treatment, though the severity of these
symptoms decreased over time, indicating an adaptation by the gastrointestinal tract to the
non-digestible substances. The higher dose (15 g GOS/lc-inulin) may cause greater overall
gastrointestinal distress due to increased diarrhea or an urgent need to defecate in some
individuals, though overall the distress was mild in nature and not likely to be of clinical
significance.
Sobotka and colleagues (1997) added inulin to tube feedings for of nine chronically ill patients
for a period of one week. Patients included in the study included individuals suffering from
anorexia, pseudobulbar paralysis, ulcerative colitis, Crohns disease and/or chronic pancreatitis.
Daily inulin intake ranged from 30 to 37.5 g. No statistically significant change in stool
frequency or consistency was observed, and no change in fecal SCFA concentration was
observed. An increase in flatulence was reported in six of the nine patients. However, no
nausea or other side effects were associated with the inulin administration. Intestinal
permeability was not influenced by the inulin, as demonstrated by the 51Cr-EDTA absorption
test. All patients successfully completed the study. It should be noted that the 2-1 fructan
dose tested in this study was significantly higher than the 2 g lc-inulin (combined with 18 g
GOS) daily intake potentially resulting from the proposed use of these substances in medical
foods.

Safety Data

77

000093

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Zheng and colleagues (2006) administered an enteral formula containing a 70/30 combination
of Raftilose oligofructose and Raftiline inulin to 32 hospitalized pediatric cancer patients
undergoing chemotherapy (mean age of 7.5 years). A control group was included in the study.
All patients were allowed an oral mixed diet of their choice, ad libitum. The test formula was
consumed for at least 13 days, with continuation of the study from days 14 to 30 optional.
Twenty-nine patients continued on with the optional part of the study. The average combined
oligofructose and inulin intake was 1.22 g/day. Normal growth rates were observed throughout
the study. No changes in biochemical or immunological parameters were observed. Overall,
gastrointestinal complaints were mild in nature. In the oligosaccharide/inulin group, one patient
reported rectal discomfort, three reported abdominal pain and/or mild flatulence and one
reported mild diarrhea. Twelve patients in the control group and 11 in the treatment group
complained of nausea. One patient in the oligosaccharide/inulin group developed diarrhea and
was withdrawn from the study; this case was determined to not be treatment-related.
In a study conducted by Bunout and colleagues (2002), 20 healthy, elderly adults were
administered a daily 6 g supplement containing a 70/30 combination of Raftilose oligofructose
and Raftiline inulin for 28 weeks. The study also included a control group of 23 subjects. Full
clinical examinations, including measurement of anthropometric parameters and a mini nutrition
assessment (MNA), were performed at baseline. No differences were observed between the
two study groups. At week 2 of the study, subjects were immunized with influenza and
pneumococcal vaccines. Subjects were evaluated at the study center weekly for the first
8 weeks and biweekly thereafter for the remaining 20 weeks. Basal values for serum proteins,
immunoglobulins, C-reactive protein and secretory IgA did not differ between groups and did not
change significantly at week 2 or week 8 of the study. In total, 62 episodes of respiratory
infections or symptoms, 13 gastrointestinal infections, four skin infections and five genitourinary
infections were reported during the study. No differences in these reports were observed
between the treatment and control groups. Of the 66 subjects originally considered eligible for
the study, no participants were withdrawn or dropped-out due to treatment-related events. Nine
participants, however, were withdrawn from the analysis for non-compliance, and five of these
participants belonged to the treatment group.
Cockram and colleagues (1998) reported a study in which 79 end-stage renal disease patients
were enrolled in groups fed a standard enteral formula, a standard renal enteral formula, or a
FOS-supplemented (DP 2-4) renal enteral formula for a period of two weeks. The formula
containing added FOS was consumed by 26 stable, outpatient adults. One week prior to
supplementation onset, baseline gastrointestinal symptoms, bowel habits and biochemical
parameters were collected while participants ingested their usual diets. During the
supplementation phase, patients consumed 17, 28 and 35 kcal/kg actual weight/day on days 8,
9 and 10, respectively. Patients experiencing gastrointestinal intolerance had the rate at which
the product intake was increased delayed, but all patients were required to achieve at least
30 kcal/kg/day by day 12. The average FOS intake was 15.6 g/day during week 1 and
18.5 g/day during week 2. Formula intake did not statistically differ between treatment groups.
No changes in the number or severity of gastrointestinal symptoms including nausea, diarrhea
and flatulence were observed for any of the groups compared to baseline. A total of five
patients withdrew from the study due to gastrointestinal complaints; one of these patients was

Safety Data

78

000094

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

from the FOS group. The highest intake of FOS achieved in the study was 29.6 g/day for four
days by one patient; this did not result in any gastrointestinal complaints.
Results of studies conducted among healthy populations provide additional evidence for
tolerance to medical foods containing added 2-fructans at intake levels of up to 31 g/d. Garleb
and colleagues (1996) administered FOS-containing (DP 2-4) enteral formulas to 18 healthy,
male university students for two weeks to observe effects on participants tolerance, serum
chemistry profiles and fecal bifidobacteria. Two levels of FOS were used in the formulations,
5 g/L and 10 g/L FOS. The test substance was prepared in meals and administered three times
per day. The nine subjects in the low-dose FOS group consumed an average of 15 g/d FOS
and the nine in the high-dose FOS group consumed approximately 31 g/d FOS. Nine additional
subjects were included in the study as a control group. Formula intake within the two treatment
groups was consistent throughout the study and the amount of formula consumption did not
significantly differ from the control group. No changes in body weights were observed
throughout the trial nor were any clinically relevant effects on serum chemistry profiles identified
for any of the study participants. Fecal samples from the two FOS-groups revealed higher
acetate concentrations by the end of the treatment period compared to the control group. Both
treatments groups also had significantly higher fecal bifidobacteria levels at the end of the
treatment period compared to the control. Gastrointestinal complaints of nausea, cramping,
distension, vomiting and regurgitation did not differ between the study groups and the incidence
of such complaints was low. Furthermore, no complaints were severe in nature. Significantly
more flatulence was reported in the high-dose FOS group compared to the low-dose and control
groups. One participant withdrew from the study after day 1 due to intolerance to the liquid
product. This subject was replaced with an alternate. No other dropouts were reported in the
study.
In two separate crossover-designed studies (Whelan et al. 2005; Whelan et al. 2006) healthy
adults consumed a standard enteral formula for one 2-week period and an enteral formula
containing FOS (DP 2-4) and soy polysaccharides as a sole source of nutrition for another
2-week period. The first study (Whelan et al. 2005) included ten adults (four men and six
women). The test formula in this study contained 5.1 g/L FOS and 8.9 g/L soy polysaccharides.
Daily FOS intake averaged 9.5 g. The amount of daily formula ingestion prescribed for each
participant was calculated based on body weight to achieve a daily energy intake of
approximately 9414 kJ. Compliance was derived based on whether or not this objective was
met. Intake compliance during the trial period was 83%, significantly lower than the 90%
compliance observed during the control period. Despite this decrease in formula intake, no
difference in weight loss was observed. At the end of the two week period in which the FOScontaining formula was consumed, the proportion of bifidobacteria was significantly higher than
that observed at the end of the control period and at baseline. Investigators noted that the
magnitude of the bifidogenic effect was negatively correlated with the baseline concentration of
bifidobacteria. Compared to baseline, total SCFA, acetate and propionate concentrations were
significantly higher after consumption of the FOS-containing formula while butyrate
concentrations decreased. During the control period, all fecal concentrations of SCFAs
decreased compared to baseline. Fecal pH increased with administration of both formulas
compared to baseline, although the increase was significantly lower during the trial period.
No difference in the incidence or severity of stomach rumbling, stomach cramps, acid reflux,

Safety Data

79

000095

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

belching, vomiting, gut rumbling or gut cramps were observed during any phase of the study.
Although a higher incidence and severity of nausea and flatulence was observed while
consuming the FOS-containing formula versus the standard enteral formula, no difference was
observed when compared to baseline levels. Four of the original 14 subjects recruited to the
study dropped out, two of which claimed they could not consume enteral formula as a sole
source of nutrition. The other two dropouts were due to personal or medical reasons and not
related to FOS treatment.
In the second study reported by Whelan and colleagues (2006), 11 subjects (five males and six
females) consumed an average of 9.8 g/d FOS (DP 2-4) during the test phase. The test formula
contained 5.0 g/L FOS and 10 g/L pea fiber. Study participants lost weight during both the FOS
period and control period compared to baseline levels, although there was no significant
difference in mean weight loss when these two periods were compared. There was also no
difference in macronutrient intake during the two periods and consumption of the FOS enteral
formula resulted in significantly higher mean fullness, minimum fullness and minimum satiety
compared to the control enteral formula. Three of the original 14 subjects dropped out either
due to personal reasons or dislike of the formula. No FOS-related dropouts were reported.
Guimber et al. (2010) evaluated the effect of a pediatric tube feed supplemented with a
multifiber mixture on the gut microbiota, bowel function, GI tolerance and nutritional and
micronutrient status of children over seven years old on long-term enteral nutrition (EN). The
cross-over study was randomized, controlled, double-blind, and involved 27 children (20 male,
7 female; average age of 11.9 3.9 years) who were on long term EN (4.8 3.9 years).
The fiber-free pediatric tube feed product was otherwise nutritionally complete and the multifiber product contained 10.3% FOS, 22.2% inulin, 30.1% soya polysaccharides, 11.3%
cellulose, 15.0% gum arabic, and 11.1% resistant starch. Nutricia manufactured and distributed
the study products. Six of the ten study authors are affiliated with Nutricia and Danone
Research. The subjects received one of the products for a three month period, and then
received the other product for three months. The subjects had a one month run-in phase prior
to the study and a one month washout phase of fiber-free feed between each intervention
phase. The authors relied on 48 h dietary recalls and reported that overall the products provided
80 85% of the subjects total energy intake. The authors evaluated anthropometric measures,
fecal pH, microbiota, bowel movements and markers of blood micronutrient status. Guimber et
al. (2010) observed a 16.6% increase in stool bifidobacteria (p < 0.05) and a reduction in stool
pH (p < 0.001) during the multi-fiber period. Guimber et al. (2010) also observed a significant
increase in plasma ferritin (p < 0.05) during the fiber-free period but the levels were within
normal ranges. The authors did not observe a difference in other blood parameters,
anthropometric measures, stool frequency or consistency, or other diet effects between the
treatments.
Seven of the subjects did not finish the study including four due to GI issues (one postgastrostomy digestive fistula requiring surgery and three disturbed GI functions with diarrhea)
(Guimber et al. 2010). The authors did not explicitly state that these issues were not related to
the study treatments, though their conclusion that the treatments were well tolerated suggests
that they were not. The authors noted that several variables may have had a carry-over effect

Safety Data

80

000096

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

due to an insufficient length of the washout period. The study population was small with only
20 subjects completing the study, and the population had a variety of pathologies that may have
impacted the observed effects. Regardless, Guimber et al. (2010) concluded that a pediatric
tube feed supplemented with multiple dietary fibers, including FOS/inulin, was well tolerated,
reduces gut pH, and promotes the growth of bifidobacteria.
(The remainder of this page intentionally left blank.)

Safety Data

81

000097

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-6. Studies with FOS/Inulin/Oligofructose in Enteral Formulas


Reference

Treatment
Population
n = 20;
age = mean 76.2 y

Bunout et
al. 2002

Healthy, elderly
adults administered
influenza &
pneumococcal
vaccines

n = 26;
age = mean 46 y
Cockram
et al. 1998

Free-living adults
with end-stage
renal disease

n = 18;
Garleb et
al. 1996

Safety Data

Healthy, male
university students

Dose

Treatment
Duration

6 g/d
oligofructose & inulin
Source:
70% Raftilose, 30%
Raftiline, Orafti

28 wk

15.6 g/d (wk 1)


18.5 g/d (wk 2) FOS
Source:
reformulated Nepro;
Ross Products.
(DP=2-4)

2 wk

15 g/d or 31 g/d
FOS
Source: Golden
Technologies Co,
Inc.; Colorado.
(DP=2-4)

2 wk

Effects
Safety/Tolerance:
-No difference in episodes of infections, including gastrointestinal.
Dropouts:
-3 withdrawn from study due to adverse events; not treatment-related.
-9 subjects withdrawn for non-compliance; 5 in treatment group.
Microbiological:
-Not measured in this study.
Other Endpoints:
-No effect on serum proteins, immunoglobulins, C-reactive protein, secretory IgA,
antibodies, or IL-4 and interferon- secretion by cultured monocytes.
Safety/Tolerance:
-No changes in number or severity of GI symptoms including nausea, diarrhea,
flatulence, cramping, distention; no change in stool frequency or consistency.
-No change in urea kinetics; no difference in mean energy or protein intakes.
Dropouts:
-5 withdrawn due to GI symptoms; 1 from treatment group.
Microbiological:
-Not measured in this study.
Other Endpoints:
-Improved serum phosphorus and calcium phosphorus product at 15 and 22 d.
Safety/Tolerance:
-No change in body weights.
-No effect on serum chemistry profiles.
-No difference in complaints of nausea, cramping, distension, or vomiting.
-Comparable incidence of diarrhea; slightly more complaints in high-dose
treatment group of diarrhea, distension and flatus.
Dropouts:
-1 withdrawn from high-dose group after 1 d due to intolerance of the liquid
product; subject replaced with an alternate.
Microbiological:
-Increase in fecal bifidobacteria levels for both treatment groups.
-Increase in SCFA concentration by d 14.
-No difference in fecal pH.

82

000098

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-6. Studies with FOS/Inulin/Oligofructose in Enteral Formulas


Reference

Treatment
Population

n = 40; >18 y
20 per treatment
group
Gori et al.
2011

Non-symptomatic,
antiretroviral-nave
HIV seropositive
adults

n = 27;

Guimber
et al. 2010

age = 11.9 3.9 y;

Long-term enteral
nutrition patients.

Sobotka et
al.
1997

Safety Data

n = 9;
age = mean 45 y
Adult patients
requiring enteral
nutrition.

Dose

Treatment
Duration

13.5 g GOS/d & 1.5


lc-inulin g/d
or
6.75 g GOS/d &
0.75 g lc-inulin/d
dissolved in water or
juice or mixed with
yogurt

12 wk

Sources: scGOS
Borculo Domo; lcinulin Orafti; AOS Sudzucker
Nutritionally
complete, multi-fiber
product containing
10.3% FOS & 22.2%
inulin

3 months
with 1
month runin, 1 month
washout

Source: Nutricia

30-37.5 g/d inulin


Source: Raffinerie,
Belgium

1 wk

Effects
Safety:
-Increase in overall G.I. symptoms in high-dose group at wk 4 and 12 compared
to low-dose group and control.; decrease in complaints observed at wk 12
compared to wk 4.
-Increase in abdominal distension and flatulence at wk 12.
-Complaints overall were mild and not clinically relevant.
-No unexpected clinical adverse events, or serious adverse events observed.
Dropouts: 10; 4 due to adverse events; 2 withdrew consent; 4 lost to follow-up.
Microbiological:
-Increased bifidobacteria.
-decreased Clostridium coccoides/Eubacterium rectale cluster.
-Decreased C. lituseburnense/C. histolyticum groups levels.
Other Effects:
-Reduction of soluble CD14.
+
+
-Activated CD4 /CD25 T cells.
-Increased natural killer cell activity.
Safety: Well tolerated; no changes in stool frequency or consistency, no change
in blood micronutrient status.
Dropouts: 7; not related to the study treatments.
Microbiological:
-Increase in stool bifidobacteria.
Other Effects:
-Reduction in stool pH.
Safety:
-Increase in flatulence.
-No change in stool frequency or consistency.
-No change in intestinal permeability.
Dropouts:
-No dropouts; all patients completed study.
Microbiological:
-No change in fecal SCFA concentration.

83

000099

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-6. Studies with FOS/Inulin/Oligofructose in Enteral Formulas


Reference

Treatment
Population

Dose

Treatment
Duration

9.5 g/d FOS


n = 10; 21-34 y

soy polysaccharides

Whelan et
al. 2005

2 wk
Healthy adults
consuming enteral
formula

n = 11; mean 28 y
Whelan et
al. 2006

Safety Data

Healthy adults
consuming enteral
formula

Source: Nestle
(DP=2-4)

9.8 g/d FOS


Pea fiber
Source: Nestle
(DP=2-4)

2 wk

Effects
Safety/Tolerance:
-No difference in weight loss.
-Intake compliance 83% for treatment group; lower than for control.
-Increase in stool frequency; decrease in daily fecal weight.
-No difference in incidence or severity of gastrointestinal complaints.
Dropouts:
-2 subjects unable to consume enteral formula as sole source of nutrition; 1
subject dropped out for personal reasons unrelated to study; 1 subject not
included in analysis due to a positive Giardia lamblia test.
Microbiological:
-Increase in bifidobacteria compared with baseline and control levels.
-Decrease in clostridia compared to baseline.
-Increase in total SCFA, acetate and propionate concentrations; decrease in
butyrate.
Safety/Tolerance:
-No difference in macronutrient intakes.
-No difference in mean body weight.
Dropouts:
-2 participants dropped out due to dislike of formula; 1 dropped out for personal
reasons.
-No treatment-related dropouts.
Microbiological:
-Not measured in study.
Other Endpoints:
-Increase in mean fullness, minimum fullness and satiety.

84

000100

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Table 5-6. Studies with FOS/Inulin/Oligofructose in Enteral Formulas


Reference

Treatment
Population

n = 32; mean 7.5 y


Zheng et
al. 2006

Pediatric cancer
patients consuming
enteral formula

Dose

Treatment
Duration

1.22 g/d
oligofructose & inulin
60 mg-kg-bw/d
Source: 70%
Raftilose, 30%
Raftiline

13-30 d

Effects
Safety/Tolerance:
-No effect on biochemical parameters (total bilirubin, alanine aminotransferase,
alkaline phosphatase, blood urea nitrogen, creatinine, glucose, etc.).
-Increase in hemoglobin and hematocrit; no effect on other hematological
parameters.
-Normal growth rates.
-1 report of rectal discomfort, 3 reports of mild flatulence, 1 report of mild
diarrhea; reports of flatulence included two occasions of abdominal pain.
-Twelve patients in control and 11 in treatment complained of nausea.
Dropouts:
-6 patients in the control group and 3 patients in treatment group voluntarily
discontinued enteral product between d 13 and 30 (Continuation was optional).
-1 patient developed diarrhea and was withdrawn from study at 3 d; nontreatment related.
Microbiological:
-No effect on bifidobacteria.
Other Endpoints:
-Normal immunological parameters: TNF-, IL-6, IL-2, LI(CD4/CD8).
-AGP levels different at 13 d of treatment; no difference in proportion of patients
with AGP increases above 1.4 g/L.
-Decrease in Prognostic Inflammatory and Nutritional Index at 30 d of treatment.

Abbreviations:
AOS = acidic oligosaccharides; cfu = colony forming units; d = day; DP = degree of polymerization; f = female; FOS = fructooligosaccharides; g = grams; GA =
gestational age; GI = gastrointestinal; GOS = galacto-oligosaccharides; HDL = high-density lipoprotein; HM = human milk; IL = interleukin; L = liter; lc-inulin = longchain inulin; LDL = low-density lipoprotein; m = male; NEC = necrotizing enterocolitis; sc = short-chain; SCFA = short-chain fatty acids; TNF = tumor necrosis factor;
wk = week; y = years.

Safety Data

85

000101

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

6 Summary and Conclusions


6.1

Summary and Conclusion on the Use of Long-Chain Inulin in Infant Formula

Numerous large, randomized, double-blind, placebo-controlled trials have been conducted to


assess the impact of formulas supplemented with a combination of GOS and lc-inulin on infant
growth, tolerance, and fecal microflora. Results from these studies consistently indicate that
formulas containing up to 8 g of the GOS/lc-inulin mixture support normal growth, are well
tolerated, and tend to produce fecal microflora and stool patterns more characteristic of infants
fed human milk as compared to infants fed standard formula. Results from studies of highly
sensitive and closely monitored preterm infants provide additional evidence that the GOS/lcinulin mixture is suitable for use in infant formulas.
None of the study populations in the clinical trials of GOS/lc-inulin supplementation included
infants with metabolic disorders. Given the rare occurrence of most inborn errors of
metabolism, it is difficult to conduct a clinical trial in this population. Trials in the general
population of infants, however, are appropriate to test the efficacy and safety of some novel
ingredients added to infant formula. Prebiotics such as GOS and lc-inulin are not digested and
exert their putative health effects primarily via fermentation in the colon. Gastrointestinal
function is not adversely affected in most infants requiring metabolic formula, therefore
tolerance of exempt metabolic infant formulas containing added lc-inulin and GOS ingredients
would be expected to be equivalent to tolerance of comparable doses in standard formulas.
A critical evaluation of the available evidence indicates that infant formulas containing up to 0.8
g lc-inulin, in combination with 7.2 g GOS, are well tolerated, safe, and suitable for term infants
from birth and that toddler formulas containing up to 1.2 g lc-inulin and 10.8 g GOS are well
tolerated and suitable for children 12 to 35 months of age.

6.2

Summary and Conclusion on the Use of Long-Chain Inulin in Medical Foods

The lc-inulin and related 2-1 fructans, in amounts of up to 37.5 g/day, are well-tolerated by
most individuals as demonstrated in studies conducted in a variety of populations, including
both healthy and chronically ill individuals. Initial mild discomfort tends to lessen with the
individuals adjustment to the 2-1 fructan intake. Some individuals, however, may not tolerate
higher levels of non-digestible carbohydrates. These medical foods will be administered under
medical supervision. The content of the GOS/lc-inulin mixture in the product will be clearly
labeled, which will enable health care professionals to administer the product in such a way that
ingestion of the product is well tolerated. Medical foods will be available in forms that do not
contain added lc-inulin and GOS, so it will be possible to customize fiber intakes as needed by
administering a combination of fiber-containing and fiber-free products.
A critical evaluation of the results of clinical studies of 2-1 fructan intake in populations
requiring medical foods demonstrates that the proposed typical uses of lc-inulin, in combination
with GOS are well tolerated, safe, and suitable for the tolerated by the target population. A
critical evaluation of the available evidence indicates that medical foods containing up to 2.5 g
lc-inulin per day, with a total GOS/lc-inulin intake of approximately 25 g per day, are well
tolerated, safe, and suitable for the general population under medical supervision.

Conclusions

86

000102

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

6.3

Conclusion on the Use of Long-Chain Inulin

The weight of the available evidence supports the safety of Danones proposed uses of lc-inulin
as a food ingredient in term infant formulas, toddler formulas and medical foods.
(The remainder of this page intentionally left blank.)

References

87

000103

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

7 References
Alliet P, Scholtens P, Raes M, Hensen K, Jongen H, Rummens JL, Boehm G, Vandenplas Y.
2007. Effect of prebiotic galacto-oligosaccharide, long chain fructo-oligosaccharide infant
formula on serum cholesterol and triacylglycerol levels. Nutr. 23:719-723.
Arslanoglu S, Moro GE, Boehm G. 2007. Early supplementation of prebiotic oligosaccharides
protects formula-fed infants against infections during the first 6 months of life. J Nutr.
137(11):2420-4.
Arslanoglu S, Moro GE, Schmitt J, Tandoi L, Rizzardi S and Boehm G. 2008. "Early dietary
intervention with a mixture of prebiotic oligosaccharides reduces the incidence of allergic
manifestations and infections during the first two years of life." J Nutr 138(6): 1091-5.
Bacchetta J, Villard F, Vial T, Dubourg L, Bouvier R, Kassai B and Cochat P. 2008a. 'Renal
hypersensitivity' to inulin and IgA nephropathy. Pediatr Nephrol 23(10): 1883-5.
Bacchetta J, Cochat P, Villard F, Astier M, Vial T, Dubourg L, Hadj-Aissa A and Kassai B.
2008b. Hypersensitivity to inulin: a rare and mostly benign event. Am J Kidney Dis 52(3):
632-3.
Bakker-Zierikzee AM, Alles M, Knol J, Kok FJ, Tolboom JJM, Bindels JG. 2005. Effects of infant
formula containing a mixture of galacto- and fructo-oligosaccharides or viable
Bifidobacterium animalis on the intestinal microflora during the first four months of life. Br J
Nutr. 94:783-790.
Bakker-Zierikzee AM, van Tol EAF, Kroes H, Alles MS, Kok FJ, Bindels JG. 2006. Fecal SIgA
secretion in infants fed on pre- or probiotic infant formula. Pediatr Allergy Immunol. 17:134140.
Balmer SE, Scott PH, Wharton BA. 1989. Diet and faecal flora in the newborn: casein and whey
proteins. Arch Dis Child. 64:1678-1684.
Ben Amor K, Rizzardini G, Torti C, Quirino T, Moroni M, Knol J, Bray D, Vriesema A, Clerici M,
Gori A. 2008a. Disturbed gut microbiota in HAART-nave HIV-positive adults: effect of
intervention with a specific prebiotic oligosaccharide mixture. Poster session presented at:
CROI 2008, 15th Conference on Retroviruses and Opportunistic Infections; 2008 Feb 3-6;
Boston, MA.
Ben Amor K, Rizzardini G, Torti C, Quirino T, Moroni M, Knol J, Bray D, Vriesema A, Clerici M,
Gori A. 2008b. Disturbed gut microbiota in HAART-nave HIV-positive adults: favourable
effect of intervention with a specific prebiotic oligosaccharide mixture. Paper presented at:
SOMED 2008. XVI Symposium of the International Association on Gnotobiology and XXXI
Congress of the Society of Microbial Ecology and Disease; 2008 May 28-30; Stockholm,
Sweden.
Beylot M. 2005. Effects of inulin-type fructans on lipid metabolism in man and in animal models.
Br J Nutr. 93 (Suppl 1):S163-S168.
Biedrzycka E, Bielecka M. 2004. Prebiotic effectiveness of fructans of different degrees of
polymerization. Trends in Food Science & Technology. 15:170-175.

References

88

000104

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Bisceglia M, Indrio F, Riezzo G, Poerio V, Corapi U and Raimondi F. 2009. The effect of
prebiotics in the management of neonatal hyperbilirubinaemia. Acta Paediatr 98(10): 157981.
Bode L. 2006. Recent advances on structure, metabolism, and function of human milk
oligosaccharides. J Nutr. 136(8):2127-30.
Boehm G, Lidestri M, Casetta P, Jelinek J, Negretti F, Stahl B, Marini A. 2002. Supplementation
of a bovine milk formula with an oligosaccharide mixture increases counts of fecal
bifidobacteria in preterm infants. Archives Diseases of Childhood- Fetal Neonatal Edition.
86: F178-F181.
Boehm G, Fanaro S, Jelinek J, Stahl B, Marini A. 2003. Prebiotic concept for infant nutrition.
Acta Paediatr Suppl. 441:64-67.
Boehm G, Stahl B, Jelinek J, Knol J, Miniello V, Moro GE. 2005. Prebiotic carbohydrates in
human milk and formulas. Acta Paediatr. 94:18-21.
Boehm G, Stahl B. 2007. Oligosaccharides from milk. J Nutr. 137(3 Suppl 2):847S-9S.
Bouhnik Y, Raskine L, Simoneau G, Vicaut E, Neut C, Flourie B, Brouns F, Bornet FR. 2004.
The capacity of nondigestible carbohydrates to stimulate fecal bifidobacteria in healthy
humans: a double-blind, randomized, placebo-controlled, parallel-group, dose-response
relation study. Am J Cin Nutr. 80:1658-1664.
Bovee-Oudenhoven IM, ten Bruggencate SJ, Lettink-Wissink ML, van der Meer R. 2003.
Dietary fructo-oligosaccharides and lactulose inhibit intestinal colonisation but stimulate
translocation of salmonella in rats. Gut. 52(11):1572-8.
Bruzzese E, Volpicelli M, Squeglia V, Bruzzese D, Salvini F, Bisceglia M, Lionetti P, Cinquetti M,
Iacono G, Amarri S and Guarino A. 2009. "A formula containing galacto- and fructooligosaccharides prevents intestinal and extra-intestinal infections: an observational study."
Clin Nutr 28(2): 156-61.
Bunout D, Hirsch S, Pia de la Maza M, Munoz C, Haschke F, Steenhout P, Klassen P, Barrera
G, Gattas V, Petermann M. 2002. Effects of prebiotics on the immune response to
vaccination in the elderly. J Parenter Enteral Nutr. 26:372-376.
Carabin IG and Flamm WG. 1999. Evaluation of safety of inulin and oligofructose as dietary
fiber. Reg Tox Pharm. 30:268-282.
Chandra R, Barron JL. 2002. Anaphylactic reaction to intravenous sinistrin (Inutest). Ann Clin
Biochem. 39(Pt 1):76.
Cherbut C. 2002. Inulin and oligofructose in the dietary fibre concept. Br J Nutr. 87:S159-S162.
Cockram DB, Hensley MK, Rodriguez M, Agarwal G, Wennberg A, Ruey P, Ashback D, Hebert
L, Kunau R. 1998. Safety and tolerance of medical nutritional products as sole sources of
nutrition in people on hemodialysis. J Ren Nutr. 8:25-33.
Codex. 2010. Guidelines on Nutrition Labelling. Codex Alimentarius. CAC/GL 2-1985.

References

89

000105

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Colome G, Sierra C, Blasco J, Garcia MV, Valverde E, Sanchez E. 2007. Intestinal permeability
in different feedings in infancy. Acta Paediatr. 96:69-72.
Conway PL. 1997. Development of Intestinal Microbiota. In Mackie RI, White BA & Isaacson
RE (Eds.), Gastrointestinal Microbiology (pp. 3-38). New York, NY: Chapman & Hall.
Corradini C, Bianchi F, Matteuzzi D, Amoretti A, Rossi M, Zanoni S. 2004. High-performance
anion-exchange chromatography coupled with pulsed amperometric detection and capillary
zone electrophoresis with indirect ultra violet detection as powerful tools to evaluate
prebiotic properties of fructooligosaccharides and inulin. J Chromatogr. 1054:165-173.
Costalos C, Kapiki A, Apostolou M and Papathoma E. 2008. "The effect of a prebiotic
supplemented formula on growth and stool microbiology of term infants." Early human
development 84(1): 45-49.
Coudray C, Demign C, Rayssiguier Y. 2003. Effects of dietary fibers on magnesium absorption
in animals and humans. J Nutr. 133:1-4.
Coussement PA. 1999. Inulin and oligofructose: safe intakes and legal status. J Nutr.
129:1412S-1417S.
Durieux A, Fougnies C, Jacobs H, Simon J-P. 2001. Metabolism of chicory
fructooligosaccharides by bifidobacteria. Biotechnology Letters. 23:1523-1527.
Edwards CA and Parrett AM. 2002. Intestinal flora during the first months of life: new
perspectives. Br J Nutr. 88:S11-S18.
European Food Safety Authority. 2004. Opinion of the Scientific Panel on Dietetic Products,
Nutrition and Allergies on a request from the Commission relating to the safty and suitability
for particular nutritional use by infants of fructooligosaccharides in infant formulae and
follow-on formulae. The EFSA Journal 31; 1-11.
Elia M, Engfer MB, Green CJ, Silk DB. 2007. Systematic review and meta-analysis: the clinical
and physiological effects of fiber-containing enteral formulas. Aliment Pharmacol Ther.
Postprint. doi: 10.1111/j.1365-2036.03544.x.
Ellegard L, Andersson H, Bosaeus I. 1997. Inulin and oligofructose do not influence the
absorption of cholesterol, or the excretion of cholesterol, Ca, Mg, Zn, Fe, or bile acids but
increases energy excretion in ileostomy subjects. Eur J Clin Nutr. 51:1-5.
Engfer MB, Stahl B, Finke B, Sawatzki G, Daniel H. 2000. Human milk oligosaccharides are
resistant to enzymatic hydrolysis in the upper gastrointestinal tract. Am J Clin Nutr. 71:15891596.
Fanaro S, Chierici R, Guerrini P, Vigi V. 2003. Intestinal microflora in early infancy: Composition
and development. Acta Paediatr Suppl. 441:48-55.
Fanaro S, Jelinek J, Stahl B, Boehm G, Kock R, Vigi V. 2005. Acidic oligosaccharides from
pectin hydrolysate as new component for infant formulae: Effect on intestinal flora, stool
characteristics, and pH. J Pediatr Gastroenterol Nutr. 41:186-190.

References

90

000106

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Firmansyah A, Dwipoerwantoro PG, Kadim M, Alatas S, Conus N, Lestarina L, Bouisset F and


Steenhout P. 2011. Improved growth of toddlers fed a milk containing synbiotics. Asia Pac J
Clin Nutr 20(1): 69-76.
Franck A. 2002. Technological functionality of inulin and oligofructose. Br J Nutr. 87 (Suppl
2):S287-91.
Franck P, Moneret-Vautrin DA, Morisset M, kanny G, Megret-Gabeaux ML, Olivier JL. 2005.
Anaphylactic reation to inulin: first identification of specific IgEs to an inulin protein
compound. Int Arch Allergy Immunol. 136:155-158.
FSANZ. 2008. Food Standards Australia New Zealand. Final Assessment Report; Proposal
P306; Addition of Inulin/FOS & GOS to Food.16 July.
http://www.foodstandards.gov.au/foodstandards/proposals/proposalp306addition3639.cfm
Fux R, Biedermann T, Sander-Wiecker T, Mrike K, Gleiter CH. 2004. Anaphylaxis to
intravenous sinistrin. Ann Pharmacother. 38:2175-2176. Epub 2004 Nov 9.
Garleb KA, Snook JT, Marcon MJ, Wolf BW, Johnson WA. 1996. Effect of fructooligosaccharide
containing enteral formulas on subjective tolerance factors, serum chemistry profiles, and
faecal bifidobacteria in healthy adult male subjects. Microb Ecol Health Dis. 9:279-285.
Gay-Crosier F, Schreiber G, Hauser C. 2000. Anaphylaxis from inulin in vegetables and
processed food. N Engl J Med. 342:1372.
Geboes KP, Luypaerts A, Rutgeerts P, Verbeke K. 2003. Inulin is an ideal substrate for a
hydrogen breath test to measure the orocaecal transit time. Aliment Pharmacol Ther.
18:721-729.
German JB, Freeman SL, Lebrilla CB and Mills DA. 2008. Human milk oligosaccharides:
evolution, structures and bioselectivity as substrates for intestinal bacteria. Nestle Nutr
Workshop Ser Pediatr Program 62: 205-18; discussion 218-22.
Gopal PK and Gill HS. 2000. Oligosaccharides and glycoconjugates in bovine milk and
colostrum. Br J Nutr. 84:S69-S64.
Gori A, Rizzardini G, van't Land B, Amor KB, van Schaik J, Torti C, Quirino T, Tincati C,
Bandera A, Knol J, Benlhassan-Chahour K, Trabattoni D, Bray D, Vriesema A, Welling G,
Garssen J and Clerici M. 2011. Specific prebiotics modulate gut microbiota and immune
activation in HAART-naive HIV-infected adults: results of the COPA pilot randomized trial.
Mucosal Immunol 4(5): 554-563.
Green CJ. 2001. Fiber in enteral nutrition. Clin Nutr 20:23-39.
Grber C, van Stuijvenberg M, Mosca F, Moro G, Chirico G, Braegger CP, Riedler J, Boehm G
and Wahn U. 2010. "Reduced occurrence of early atopic dermatitis because of
immunoactive prebiotics among low-atopy-risk infants." J Allergy Clin Immunol 126(4):
791-7.
Guarner F. 2007. Studies with inulin-type fructans on intestinal infections, permeability, and
inflammation. J Nutr. 137(11 Suppl):2568S-2571S.

References

91

000107

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Guimber D, Bourgois B, Beghin L, Neuville S, Pernes P, Ben Amor K, Goedhart A, Sijben J,


Knol J and Gottrand F. 2010. Effect of multifibre mixture with prebiotic components on
bifidobacteria and stool pH in tube-fed children. Br J Nutr 104(10): 1514-22.
Haarman M and Knol J. 2005. Quantitative real-time PCR assays to identify and quantify fecal
bifidobacterium species in infants receiving a prebiotic infant formula. Appl Environ
Microbiol. 71:2318-2324.
Haarman M and Knol J. 2006. Quantitative real-time PCR analysis of fecal lactobacillus species
in infants receiving a prebiotic infant formula. Appl Environ Microbiol. 72:2359-2365.
Harmsen HJM, Wildeboer-Veloo ACM, Raangs GC, Wagendorp AA, Klijn N, Bindels JG,
Welling GW. 2000. Analysis of intestinal flora development in breast-fed and formula-fed
infants by using molecular identification and detection methods. J Pediatr Gastroenterol
Nutr. 30:61-67.
Hond ED, Geypens B, Ghoos Y. 2000. Effect of high performance chicory inulin on constipation.
Nutr Res. 20:731-736.
Hopkins MJ, Sharp R, Macfarlane GT. 2001. Age and disease related changes in intestinal
bacterial populations assessed by cell culture, 16S rRNA abundance, and community
cellular fatty acid profiles. Gut. 48:198-205.
Hopkins MJ, Sharp R, Macfarlane GT. 2002. Variation in human intestinal microbiota with age.
Digest Liv Dis. 34:S12-18.
Indrio F, Riezzo G, Raimondi F, Bisceglia M, Cavallo L and Francavilla R. 2009a. Effects of
probiotic and prebiotic on gastrointestinal motility in newborns. J Physiol Pharmacol 60
Suppl 6: 27-31.
Indrio F, Riezzo G, Raimondi F, Francavilla R, Montagna O, Valenzano ML, Cavallo L and
Boehm G. 2009b. Prebiotics improve gastric motility and gastric electrical activity in preterm
newborns. J Pediatr Gastroenterol Nutr 49(2): 258-61.
Institute of Medicine (IOM). 2002. Dietary Reference Intakes (DRI) for Energy, Carbohydrates,
Fiber, Fat, Fatty Acids, Cholesterol, Protein, and Amino Acids. Washington, DC: National
Academies Press.
Institute of Medicine (IOM). 2006. Dietary Reference Intakes (DRI): the Essential Guide to
Nutrient Requirements. Washington, DC: National Academies Press.
Isolauri E, Sutas Y, Kankaanpaa P, Arvilommi H, Salminen S. 2001. Probiotics: Effects on
immunity. Am J Clin Nutr. 73:S444-S450.
Kaur N and Gupta K. 2002. Applications of inulin and oligofructose in health and nutrition.
J Biosci. 27:703-714.
Kim SH, Lee da H and Meyer D. 2007. Supplementation of baby formula with native inulin has a
prebiotic effect in formula-fed babies. Asia Pac J Clin Nutr 16(1): 172-7.

References

92

000108

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Knol J, Scholtens P, Kafka C, Steenbakkers J, Gross S, Helm K, Klarcyzk M, Schoopfer H,


Boockler HM, Wells, J. 2005a. Colon microflora in infants fed formula with galacto- and
fructo-oligosaccharides: More like breast-fed infants. J Pediatr Gastroenterol Nutr. 40:36-42.
Knol J, Boehm G, Lidestri M, Negretti F, Jelinek J, Agosti M, Stahl B, Marini A, and Mosca, F.
2005b. Increase of fecal bifidobacteria due to dietary oligosaccharides induces a reduction
of clinically relevant pathogen germs in the faeces of formula-fed preterm infants. Acta
Paediatr. 94:31-33.
Knudsen KE and Hessov I. 1995. Recovery of inulin from Jerusalem artichoke (Helianthus
tuberosus L.) in the small intestine of man. Br J Nutr. 74:101-113.
Kolbye Associates. Evaluation of the Food Safety Aspects of Inulin and Oligofructose. GRAS
Determination. Conducted by a Committee of Experts on Food Safety convened by Kolbye
Associates, 7313 Helmsdale Road, Bethesda, MD 20817. September 1992.
Langlands SJ, Hopkins MJ, Coleman N, Cummings JH. 2004. Prebiotic carbohydrates modify
the mucosa associated microflora of the human large bowel. Gut. 53:1610-1616.
Lidestri M, Agosti M, Marini A, Boehm G. 2003. Oligosaccharides might stimulate calcium
absorption in formula-fed preterm infants. Acta Paediatr. 441:91-91.
Lochs H, Allison SP, Meier R, Pirlich M, Kondrup J, Schneider S, van den Berghe G, Pichard C.
2006. Introductory to the ESPEN Guidelines on Enteral Nutrition: Terminology, definitions
and general topics. Clin Nutr. 25(2):180-6. Epub 2006 May 11.
Lugonja NM, Martinov OB, Rasovic MR, Spasic SD, Gojgic G and Vrvic MM. 2010. A
Comparative Investigation of an in vitro and Clinical Test of the Bifidogenic Effect of an
Infant Formula. J Clin Biochem Nutr 47(3): 208-16.
Lundequist B, Nord CE, Winberg J. 1985. The composition of the faecal microflora in breastfed
and bottle fed infants from birth to eight weeks. Acta Paediatr Scand. 74:45-51.
Mackie RI, Sghir A, Gaskins HR. 1999. Developmental microbial ecology of the neonatal
gastrointestinal tract. Am J Clin Nutr. 69:S1035-S1045.
Magne F, Hachelaf W, Suau A, Boudraa G, Bouziane-Nedjadi K, Rigottier-Gois L, Touhami M,
Desjeux JF and Pochart P. 2008. Effects on faecal microbiota of dietary and acidic
oligosaccharides in children during partial formula feeding. J Pediatr Gastroenterol Nutr
46(5): 580-8.
Mihatsch WA, Hoegel J, Pohlandt F. 2006. Prebiotic oligosaccharides reduce stool viscosity and
accelerate gastrointestinal transport in preterm infants. Acta Pediatr. 95:843-848.
Modi N, Uthaya S, Fell J and Kulinskaya E. 2010. A randomized, double-blind, controlled trial of
the effect of prebiotic oligosaccharides on enteral tolerance in preterm infants
(ISRCTN77444690). Pediatr Res 68(5): 440-5.
Molis C, Flourie B, Ouarne F, Gailing MF, Lartigue S, Guibert A, Bornet F, Galmiche JP. 1996.
Digestion, excretion, and energy value of fructooligosaccharides in healthy humans. Am J
Clin Nutr. 64:324-328.

References

93

000109

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Moro G, Minoli I, Mosca M, Fanaro S, Jelinek J, Stahl B, Boehm, G. 2002. Dosage-related


bifidogenic effects of galacto- and fructooligosaccharides in formula-fed term infants. J
Pediatr Gastroenterol Nutr. 34:291-5.
Moro G, Stahl B, Fanaro S, Jelinek J, Boehm G, Coppa GV. 2005. Dietary prebiotic
oligosaccharides are detectable in the faeces of formula-fed infants. Acta Paediatrica
94(Suppl 449):27-30.
Moro G, Arslanoglu S, Stahl B, Jelinek J, Wahn U, Boehm G. 2006. A mixture of prebiotic
oligosaccharides reduces the incidence of atopic dermatitis during the first six months of
age. Ach Dis Child. 91:814-819.
Moshfegh AJ, Friday JE, Goldman JP, Ahuja JK. 1999. Presence of inulin and oligofructose in
the diets of Americans. J Nutr. 129(7 Suppl):1407S-11S.
National Center for Health Statistics (NCHS). 2003-2004 National Health and Nutrition
Examination Survey Data. Hyattsville, MD: U.S. Department of Health and Human Services,
Centers for Disease Control and Prevention. 2006. Available at
http://www.cdc.gov/nchs/about/major/nhanes/nhanes2003-2004/nhanes03_04.htm.
Accessed January, 2007.
Newburg DS. 2000. Oligosaccharides in human milk and bacterial colonization. J Pediatr
Gastroenterol Nutr. 30:S8-S17.
Niness KR. 1999. Inulin and oligofrutose: What are they? J Nutr. 129:1402S-1406S.
Orrhage K and Nord CE. 1999. Factors controlling the bacterial colonization of the intestine in
breastfed infants. Acta Paediatr Suppl. 430:47-57.
Panigrahi P, Parida S, Pradhan L, Mohapatra SS, Misra PR, Johnson JA, Chaudhry R, Taylor
S, Hansen NI and Gewolb IH. 2008. Long-term colonization of a Lactobacillus plantarum
synbiotic preparation in the neonatal gut. J Pediatr Gastroenterol Nutr 47(1): 45-53.
Penders J, Thijs C, Vink C, Stelma FF, Snijders B, Kummeling I, van den Brandt PA,
Stobberingh EE. 2006. Factors influencing the composition of the intestinal microbiota in
early infancy. Pediatrics. 118:511-521.
Perrin S, Fougnies C, Grill JP, Jacobs H, Schneider F. 2002. Fermentation of chicory fructooligosaccharides in mixtures of different degrees of polymerization by three strains of
bifidobacteria. Can J Microbiol. 48:759-763.
Piemontese P, Gianni ML, Braegger CP, Chirico G, Gruber C, Riedler J, Arslanoglu S, van
Stuijvenberg M, Boehm G, Jelinek J and Roggero P. 2011. "Tolerance and safety evaluation
in a large cohort of healthy infants fed an innovative prebiotic formula: a randomized
controlled trial." PLoS One 6(11): e28010.
Pool-Zobel BL. 2005. Inulin-type fructans and reduction in colon cancer risk: review of
experimental and human data. Br J Nutr. 93 (Suppl 1):S73-S90.

References

94

000110

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Puccio G, Cajozzo C, Meli F, Rochat F, Grathwohl D and Steenhout P. 2007. Clinical evaluation
of a new starter formula for infants containing live Bifidobacterium longum BL999 and
prebiotics. Nutrition 23(1): 1-8.
Raes M, Scholtens PA, Alliet P, Hensen K, Jongen H, Boehm G, Vandenplas Y and Rummens
JL. 2010. Exploration of basal immune parameters in healthy infants receiving an infant milk
formula supplemented with prebiotics. Pediatr Allergy Immunol 21(2 Pt 2): e377-85.
Raghuveer TS, Garg U, Graf WD. 2006. Inborn errors of metabolism in infancy and early
childhood: an update. Am Fam Phys. 73:1981-1990.
Rao S, Srinivasjois R and Patole S. 2009. Prebiotic supplementation in full-term neonates: a
systematic review of randomized controlled trials. Arch Pediatr Adolesc Med 163(8): 755-64.
Rigo J, Pieltain C, Studzinski F. 2001. Growth, weight gain composition and mineral accretion in
term infants fed a new experimental formula containing hydrolysed protein, -palmitate and
prebiotics. Pediatrica. 21:387-396.
Rinne M, Gueimonde M, Kalliomaki M, Hoppu U, Salminen S and Isolauri E. 2005. Similar
bifidogenic effects of prebiotic-supplemented partially hydrolyzed infant formula and
breastfeeding on infant gut microbiota. FEMS Immunology and Medical Microbiology. 43:5965.
Roberfroid MB, Van Loo JA and Gibson GR. 1998. The bifidogenic nature of chicory inulin and
its hydrolysis products. J Nutr. 128:11-19.
Roberfroid MB. 2005. Introducing inulin-type fructans. Br J Nutr. 93:S13-S25.
Roberfroid M. 2007. Prebiotics: The concept revisited. J Nutr. 137:830S-837S.
Rodricks JV, Yates AA and Kruger CL. 2007. Gastrointestinal tract development and its
importance in toxicology. Gad SC (Ed.), Toxicology of the Gastrointestinal Tract. (pp. 81105). Florida: CRC Press.
Ross Nutrition. 2006. Ross 2006 Pocket Guide. Available at:
http://www.nbngroup.com/manuals/machine/RossProductsPocketGuide.pdf. Accessed
February 21, 2012.
Rossi M, Corradini C, Amaretti A, Nicolini M, Pompei A, Zanoni S, Matteuzzi D. 2005.
Fermentation of fructooligosaccharides and inulin by bifidobacteria: a comparative study of
pure and fecal cultures. Appl Environ Microbiol. 71:6150-6158.
Salvini F, Riva E, Salvatici E, Boehm G, Jelinek J, Banderali G and Giovannini M. 2011. A
specific prebiotic mixture added to starting infant formula has long-lasting bifidogenic effects.
J Nutr 141(7): 1335-9.
Savino F, Cresi F, Maccario S, Cavallo F, Dalmasso P, Fanaro S, Oggero R, Vigi V, Silvestro L.
2003. Minor feeding problems during the first months of life: effect of a partially hydrolysed
milk formula containing fructo- and galacto-oligosaccharides. Acta Paediatr Suppl. 441:8690.

References

95

000111

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Savino F, Palumeri E, Castango E, Cresi F, Dalmasso P, Cavallo F, Oggero R. 2006. Reduction


of crying episodes owing to infantile colic: A randomized controlled study on the efficacy of a
new infant formula. Eur J Clin Nutr. pp. 1-7.
Schmelzle H, Wirth S, Skopnik H, Radke M, Knol J, Boockler H, Broostrup A, Wells J, Fusch C.
2003. Randomized double-blind study of the nutritional efficacy and bifidogenicity of a new
infant formula containing partially hydrolyzed protein, a high -palmitic acid level, and
nondigestible oligosaccharides. J Pediatr Gastroenterol Nutr. 36:343-351.
Schneider AR, Jepp K, Murczynski L, Biniek U, Stein J. 2007. The inulin hydrogen breath test
accurately reflects orocaecal transit time. Eur J Clin Invest. 37:802-807.
Schneider SM, Le Gall P, Girard-Pipau F, Piche T, Pompei A, Nano JL, Hebuterne X, Rampal
P. 2000. Total artificial nutrition is associated with major changes in the fecal flora. Eur J
Nutr. 39:248-255.
Scholtens P, Alles M, Willemsen L, Braak C, Bindels J, Boehm G, Govers M. 2006a. Dietary
fructo-oligosaccharides in healthy adults do not negatively affect faecal cytotoxicity: a
randomised, double-blind, placebo-controlled crossover trial. Br J Nutr. 95:1143-1149.
Scholtens P, Alles M, Bindels J, van der Linde EGM, Tolboom JM, Knol J. 2006b. Bifidogenic
effects of solid weaning foods with added prebiotic oligosaccharides: A randomized
controlled clinical trial. J Pediatr Gastroenterol Nutr. 42:553-559.
Scholtens PA, Alliet P, Raes M, Alles MS, Kroes H, Boehm G, Knippels LM, Knol J and
Vandenplas Y. 2008. "Fecal secretory immunoglobulin A is increased in healthy infants who
receive a formula with short-chain galacto-oligosaccharides and long-chain fructooligosaccharides." J Nutr 138(6): 1141-7.
Scholz-Ahrens KE, Ade P, Marten B, Weber P, Timm W, Asil Y, Gluer CC, Schrezenmeir J.
2007. Prebiotics, probiotics, and synbiotics affect mineral absorption, bone mineral content,
and bone structure. J Nutr. 137:838S-846S.
Schouten B, Van Esch BC, Kormelink TG, Moro GE, Arslanoglu S, Boehm G, Knippels LM,
Redegeld FA, Willemsen LE and Garssen J. 2011. "Non-digestible oligosaccharides reduce
immunoglobulin free light-chain concentrations in infants at risk for allergy." Pediatr Allergy
Immunol 22(5): 537-42.
Scientific Committee on Food (SCF). European Commission: Health and Consumer Protection
Directorate-General. 2003. Report of the Scientific Committee on Food on the revision of
essential requirements of infant formulae and follow-on formulae: Adopted April 4, 2003.
Available at: http://ec.europa.eu/food/fs/sc/scf/out199_en.pdf. Accessed February 22, 2012.
Shadid R, Haarman M, Knol J, Theis W, Beermann C, Rjosk-Dendorfer, Schendel DJ, Koletzko
BV and Krauss-Etschmann. 2007. Effects of galactooligosaccharide and long-chain
fructooligosaccharide supplementation during pregnancy on maternal and neonatal
microbiota and immunitya randomized, double-blind, placebo-controlled trial. Am J Clin
Nutr. 86:1426-1437.
Sherman PM, Cabana M, Gibson GR, Koletzko BV, Neu J, Veereman-Wauters G, Ziegler EE
and Walker WA. 2009. Potential roles and clinical utility of prebiotics in newborns, infants,

References

96

000112

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

and children: proceedings from a global prebiotic summit meeting, New York City, June 2728, 2008. J Pediatr 155(5): S61-70.
Silk DB, Walters ER, Duncan HD, Green CJ. 2001. The effect of a polymeric enteral formula
supplemented with a mixture of six fibres on normal human bowel function and colonic
motility. Clin Nutr. 20:49-58.
Sobotka L, Bratova M, Slemrova M, Manak J, Vizda J, Zadak Z. 1997. Inulin as the soluble fiber
in liquid enteral nutrition. J Nutr. 13:21-25.
Stam J, van Stuijvenberg M, Garssen J, Knipping K and Sauer PJ. 2011. "A mixture of three
prebiotics does not affect vaccine specific antibody responses in healthy term infants in the
first year of life." Vaccine 29(44): 7766-72.
Stark PL and Lee A. 1982. The microbial ecology of the large bowel of breast-fed and formulafed infants during the first year of life. J Med Microbiol. 15:189-203.
Stone-Dorshow T and Levitt MD. 1987. Gaseous response to ingestion of a poorly absorbed
fructooligosaccharide sweetener. Am J Clin Nutr. 46:61-65.
Ten Bruggencate SJ, Bovee-Oudenhoven IM, Lettink-Wissink ML, Van der Meer R. 2003.
Dietary fructo-oligosaccharides dose-dependently increase translocation of salmonella in
rats. J Nutr. 133(7):2313-8.
Ten Bruggencate SJ, Bovee-Oudenhoven IM, Lettink-Wissink ML, Katan MB, Van Der Meer R.
2004. Dietary fructo-oligosaccharides and inulin decrease resistance of rats to salmonella:
protective role of calcium. Gut. 53(4):530-5.
Ten Bruggencate SJ, Bovee-Oudenhoven IM, Lettink-Wissink ML and Van der Meer R. 2005.
Dietary fructooligosaccharides increase intestinal permeability in rats. J Nutr. 135(4):837-42.
Ten Bruggencate SJ, Bovee-Oudenhoven IM, Lettink-Wissink ML, Katan MB and Van der Meer
R. 2006. Dietary fructooligosaccharides affect intestinal barrier function in healthy men. J
Nutr. 136(1):70-4.
Tuohy KM, Finlay RK, Wynne AG and Gibson GR. 2001. A human volunteer study on the
prebiotic effects of HP-inulinfaecal bacteria enumerated using fluorescent in situ
hybridization (FISH). Anaerobe. 7:113-118.
Underwood MA, Salzman NH, Bennett SH, Barman M, Mills DA, Marcobal A, Tancredi DJ,
Bevins CL and Sherman MP. 2009. A randomized placebo-controlled comparison of two
prebiotic/probiotic combinations in preterm infants: impact on weight gain, intestinal
microbiota, and fecal short-chain fatty acids. J Pediatr Gastroenterol Nutr 48(2): 216-25.
Vaisman N, Press J, Leibovitz E, Boehm G and Barak V. 2010. "Short-term effect of prebiotics
administration on stool characteristics and serum cytokines dynamics in very young children
with acute diarrhea." Nutrients 2(7): 683-92.
van der Aa, LB, Heymans HS, van Aalderen WM, Sillevis Smitt JH, Knol J, Ben Amor K,
Goossens DA and Sprikkelman AB. 2010. "Effect of a new synbiotic mixture on atopic
dermatitis in infants: a randomized-controlled trial." Clin Exp Allergy 40(5): 795-804.

References

97

000113

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

van der Aa LB, van Aalderen WM, Heymans HS, Henk Sillevis Smitt J, Nauta AJ, Knippels LM,
Ben Amor K and Sprikkelman AB. 2011. "Synbiotics prevent asthma-like symptoms in
infants with atopic dermatitis." Allergy 66(2): 170-7.
van der Aa LB, Lutter R, Heymans HS, Smids BS, Dekker T, van Aalderen WM, Sillevis Smitt
JH, Knippels LM, Garssen J, Nauta AJ and Sprikkelman AB. 2012. "No detectable beneficial
systemic immunomodulatory effects of a specific synbiotic mixture in infants with atopic
dermatitis." Clin Exp Allergy 42(4): 531-9.
van Hoffen E, Ruiter B, Faber J, M'Rabet L, Knol EF, Stahl B, Arslanoglu S, Moro G, Boehm G
and Garssen J. 2009. "A specific mixture of short-chain galacto-oligosaccharides and longchain fructo-oligosaccharides induces a beneficial immunoglobulin profile in infants at high
risk for allergy." Allergy 64(3): 484-7.
Van Loo J, Coussement P, Leenheer LD, Hoebregs H, Smits G. 1995. On the presence of inulin
and oligofructose as natural ingredients in the western diet. Crit Rev Food Sci Nutr. 35:525552.
Van Loo J, Cummings J, Delzenne N, Englyst H, Franck A, Hopkins M, Kok N, Macfarlane G,
Newtone D, Quigley M, Roberfroid M, van Vliet T, van den Heuvel E. 1999. Functional food
properties of non-digestible oligosaccharides: a consensus report from the ENDO project
(DGXII AIRII-CT94-1095). Br J Nutr. 81:121-132.
Van Loo J. 2004. The specificity of the interaction with intestinal bacterial fermentation by
prebiotics determines their physiological efficacy. Nutr Res Rev. 17:89-98.
van Stuijvenberg, M., A. M. Eisses, C. Gruber, F. Mosca, S. Arslanoglu, G. Chirico, C. P.
Braegger, J. Riedler, G. Boehm and P. J. Sauer. 2011. "Do prebiotics reduce the number of
fever episodes in healthy children in their first year of life: a randomised controlled trial." Br J
Nutr 106(11): 1740-8.
Vandenplas Y, Veereman-Wauters G, De Greef E, Peeters S, Casteels A, Mahler T, Devreker T
and Hauser B. 2011. Probiotics and prebiotics in prevention and treatment of diseases in
infants and children. J Pediatr (Rio J) 87(4): 292-300.
Veereman G. 2007. "Pediatric applications of inulin and oligofructose." J Nutr 137(11 Suppl):
2585S-2589S.
Veereman-Wauters G, Staelens S, Van de Broek H, Plaskie K, Wesling F, Roger LC,
McCartney AL and Assam P. 2011. "Physiological and bifidogenic effects of prebiotic
supplements in infant formulae." J Pediatr Gastroenterol Nutr 52(6): 763-71.
Vivatvakin B, Mahayosnond A, Theamboonlers A, Steenhout PG and Conus NJ. 2010. "Effect
of a whey-predominant starter formula containing LCPUFAs and oligosaccharides
(FOS/GOS) on gastrointestinal comfort in infants." Asia Pac J Clin Nutr 19(4): 473-80.
Ward RE, Nionuevo M, Mills DA, Lebrilla CB, German JB. 2006. In vitro fermentation of breast
milk oligosaccharides by Bifidobacterium infantis and Lactobacillus gasseri. Appl Environ
Microbiol. 72(6):4497-9.

References

98

000114

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Ward RE, Nionuevo M, Mills DA, Lebrilla CB, German JB. 2007. In vitro fermentability of
human milk oligosaccharides by several strains of bifidobacteria. Mol Nutr Food Res.
51(11):1398-405.
Watzl B, Girrbach S, Roller M. 2005. Inulin, oligofructose and immunomodulation. Br J Nutr.
93(Suppl 1):S49-S55.
Weaver CM. 2005. Inulin, oligofructose and bone health: experimental approaches and
mechanisms. Br J Nutr. 93 (Suppl 1):S99-S103.
Westerbeek EAM, van den Berg JP, Lafeber HN, Fetter WP, Boehm G, Twisk JW and van
Elburg RM. 2010. "Neutral and acidic oligosaccharides in preterm infants: a randomized,
double-blind, placebo-controlled trial." Am J Clin Nutr 91(3): 679-86.
Westerbeek EAM, Morch E, Lafeber HN, Fetter WP, Twisk JW and Van Elburg RM. 2011a.
"Effect of neutral and acidic oligosaccharides on fecal IL-8 and fecal calprotectin in preterm
infants." Pediatr Res 69(3): 255-8.
Westerbeek EAM, van den Berg A, Lafeber HN, Fetter WP and van Elburg RM. 2011b. "The
effect of enteral supplementation of a prebiotic mixture of non-human milk galacto-, fructoand acidic oligosaccharides on intestinal permeability in preterm infants." Br J Nutr 105(2):
268-74.
Westerbeek EA, Hensgens RL, Mihatsch WA, Boehm G, Lafeber HN and van Elburg RM.
2011c. "The effect of neutral and acidic oligosaccharides on stool viscosity, stool frequency
and stool pH in preterm infants." Acta Paediatr 100(11): 1426-31.
Whelan K, Judd PA, Preedy VR, Simmering R, Jann A, Taylor MA. 2005.
Fructooligosaccharides and fiber partially prevent the alterations in fecal microbiota and
short-chain fatty acid concentrations caused by standard enternal formula in healthy
humans. J Nutr. 135:1896-1902.
Whelan K, Efthymiou K, Judd PA, Preedy VR, Taylor MA. 2006. Appetite during consumption of
enteral formula as a sole source of nutrition: the effect of supplementing pea-fibre and
fructo-oligosaccharides. Br J Nutr. 96:350-356.
Zheng S, Steenhout P, Kuiran D, Qihong W, Weiping W, Hager C, Haschke F, Clemens RA.
2006. Nutritional support of pediatric patients with cancer consuming an enteral formula with
fructooligosaccharides. Nutr Res. 26:154-162.

References

99

000115

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Safety Evaluation of
Long-Chain Inulin
for Use in
Term Infant Formulas,
Toddler Formulas,
and Medical Foods

Appendix A
Batch Data and Methods of Analysis for Orafti HP

Prepared for:
Danone Trading B.V.
Schiphol, The Netherlands
Prepared by:
ENVIRON International Corporation
Phoenix, Arizona
Date:
April 30, 2013
Project Number:
28-26968B

Appendix A

000116

Appendix A: Lc-Inulin Safety Evaluation


Prepared for Danone

Table A-1. Annual Analyses of Product: Orafti HP lc-inulin


Batches and Production Date
Parameter
Physical/Chemical
Inulin (% CHO)
Inulin DP 5 (% CHO)
Average DP
Glucose, fructose, sucrose (% CHO)
Sulfated ash (% at 525C)
Conductivity (S/cm at 28 Brix)
Contaminant
Lead (mg/kg)
Arsenic (mg/kg)
Microbiological
Total mesophilic bacteria (cfu/g)
Yeasts (cfu/g)
Molds (cfu/g)
Enterobacteriaceae (cfu/1 g)
Staphylococcus aureus (cfu/0.1 g)
Salmonella spp. (cfu/250 g)

28-26968B

HPBNN
6DNN6
10/27/06

HGQEO
7CEO7
04/19/07

HGQEQ
7CEQ7
04/21/07

HPHOT
7DOT7
11/26/07

HGQEC
8CEC8
04/08/08

HGQEE
8CEE8
04/10/08

HPQER
9CER9
04/22/09

HPQEX
9OEX9
04/27/09

HPBNK
9DNK9
10/24/09

HGBLT
0HLV0
09/15/10

99.8
99.3
30
0.2
0.18
182

99.9
99.9
31
0.1
0.05
54

99.9
99.8
32
0.1
0.07
77

99.8
99.4
27
0.2
0.14
242

99.9
99.8
33
0.1
0.13
133

99.9
99.8
34
0.1
0.11
137

99.7
99.2
34
0.3
0.05
84

99.8
99.4
35
0.2
0.10
150

99.9
99.6
30
0.1
0.07
136

99.7
99.3
27
0.3
0.12
156

<0.01
<0.03

<0.01
<0.03

<0.01
<0.03

<0.01
<0.03

<0.01
<0.03

<0.01
<0.03

<0.01
<0.03

<0.01
<0.03

<0.01
<0.03

<0.01
<0.03

0
0
0
negative
negative
negative

0
0
0
negative
negative
negative

0
0
0
negative
negative
negative

0
0
0
negative
negative
negative

0
0
0
negative
negative
negative

0
0
0
negative
negative
negative

0
0
0
negative
negative
negative

0
0
0
negative
negative
negative

0
0
0
negative
negative
negative

0
0
0
negative
negative
negative

000117

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Safety Evaluation of
Long-Chain Inulin
for Use in
Term Infant Formulas,
Toddler Formulas,
and Medical Foods

Appendix B
GRAS Notifications for the Use of Inulin, Oligofructose and
Fructooligosaccharides as Food Ingredients

Prepared for:
Danone Trading B.V.
Schiphol, The Netherlands
Prepared by:
ENVIRON International Corporation
Phoenix, Arizona
Date:
April 30, 2013
Project Number:
28-26968B

Appendix B

000118

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

List of GRAS Notifications Contained on CD


GRN044 FOS (GTC Nutrition) FDA response letter 2000
GRN044 FOS (GTC Nutrition) FDA response letter 2007
GRN044 FOS (GTC Nutrition)
GRN118 Inulin (Sensus) FDA response letter 2003
GRN118 Inulin (Sensus) FDA response letter 2008
GRN118 Inulin (Sensus)
GRN236 GOS (Friesland Foods) FDA response letter 2008
GRN236 GOS (Friesland Foods)
GRN285 GOS (GTC Nutrition)
GRN286 GOS (GTC Nutrition) FDA response letter 2009
GRN286 GOS (GTC Nutrition)
GRN334 GOS (Yakult) FDA response letter 2010
GRN334 GOS (Yakult)
GRN392 GOS Beneo-Orafti FDA response letter 2012
GRN392 GOS Beneo-Orafti

Appendix B

000119

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Safety Evaluation of
Long-Chain Inulin
for Use in
Term Infant Formulas,
Toddler Formulas,
and Medical Foods

Appendix C
References

Prepared for:
Danone Trading B.V.
Schiphol, The Netherlands
Prepared by:
ENVIRON International Corporation
Phoenix, Arizona
Date:
April 30, 2013
Project Number:
28-26968B

Appendix C

000120

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

List of References Contained on CD


Alliet et al. 2007.
Arslanoglu et al. 2007.
Arslanoglu et al. 2008.
Bacchetta et al. 2008a.
Bacchetta et al. 2008b.
Bakker-Zierikzee et al. 2005.
Bakker-Zierikzee et al. 2006.
Balmer et al. 1989.
Ben Amor et al. 2008a.
Ben Amor et al. 2008b.
Beylot 2005.
Biedrzycka and Bielecka 2004.
Bisceglia et al. 2009.
Bode 2006.
Boehm et al. 2002.
Boehm et al. 2003.
Boehm et al. 2005.
Boehm and Stahl 2007.
Bouhnik et al. 2004.
Bovee-Oudenhoven et al. 2003.
Bruzzese et al. 2009.
Bunout et al. 2002.
Carabin and Flamm 1999.
Chandra and Barron 2002.
Cherbut 2002.
Cockram et al. 1998.
Codex 2010.
Colome et al. 2007.
Conway 1997.
Corradini et al. 2004.
Costalos et al. 2008.
Coudray et al. 2003.
Coussement 1999.
Durieux et al. 2001.
Edwards and Parrett 2002.
European Food Safety Authority 2004.
Elia et al. 2007.
Ellegard et al. 1997.
Engfer et al. 2000.

Appendix C

000121

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Fanaro et al. 2003.


Fanaro et al. 2005.
Firmansyah et al. 2011.
Food Standards Australia New Zealand (FSANZ) 2008.
Franck 2002.
Franck et al. 2005.
Fux et al. 2004.
Garleb et al. 1996.
Gay-Crosier et al. 2000.
Geboes et al. 2003.
German et al. 2008.
Gopal and Gill 2000.
Gori et al. 2011.
Green 2001.
Grber et al. 2010.
Guarner 2007.
Guimber et al. 2010.
Haarman and Knol 2005.
Haarman and Knol 2006.
Harmsen et al. 2000.
Hond et al. 2000.
Hopkins et al. 2001.
Hopkins et al. 2002.
Indrio et al. 2009a.
Indrio et al. 2009b.
Institute of Medicine (IOM). 2002.
Institute of Medicine (IOM). 2006.
Isolauri et al. 2001.
Kaur and Gupta 2002.
Kim et al. 2007.
Knol et al. 2005a.
Knol et al. 2005b.
Knudsen and Hessov 1995.
Kolbye Associates 1992.
Langlands et al. 2004.
Lidestri et al. 2003.
Lochs et al. 2006.
Lugonja et al. 2010.
Lundequist et al. 1985.
Mackie et al. 1999.
Magne et al. 2008.

Appendix C

000122

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Mihatsch et al. 2006.


Modi et al. 2010.
Molis et al. 1996.
Moro et al. 2002.
Moro et al. 2005.
Moro et al. 2006.
Moshfegh et al. 1999.
National Center for Health Statistics (NCHS) 2006.
Newburg 2000.
Niness 1999.
Orrhage and Nord 1999.
Panigrahi et al. 2008.
Penders et al. 2006.
Perrin et al. 2002.
Piemontese et al. 2011.
Pool-Zobel 2005.
Puccio et al. 2007.
Raes et al. 2010.
Raghuveer et al. 2006.
Rao et al. 2009.
Rigo et al. 2001.
Rinne et al. 2005.
Roberfroid et al. 1998.
Roberfroid 2005.
Roberfroid 2007.
Rodricks et al. 2007.
Ross Nutrition. 2006.
Rossi et al. 2005.
Salvini et al. 2011.
Savino et al. 2003.
Savino et al. 2006.
Schmelzle et al. 2003.
Schneider et al. 2007.
Schneider et al. 2000.
Scholtens et al. 2006a.
Scholtens et al. 2006b.
Scholtens et al. 2008.
Scholz-Ahrens et al. 2007.
Schouten et al. 2011.
Scientific Committee on Food (SCF) 2003.
Shadid et al. 2007.

Appendix C

000123

lc-Inulin Safety Evaluation


Prepared for Danone Trading B.V.

Sherman et al. 2009.


Silk et al. 2001.
Sobotka et al. 1997.
Stam et al. 2011.
Stark and Lee 1982
Stone-Dorshow and Levitt 1987.
Ten Bruggencate et al. 2003.
Ten Bruggencate et al. 2004.
Ten Bruggencate et al. 2005.
Ten Bruggencate et al. 2006.
Tuohy et al. 2001
Underwood et al. 2009.
Vaisman et al. 2010.
van der Aa et al. 2010.
van der Aa et al. 2011.
van der Aa et al. 2012.
van Hoffen et al. 2009.
Van Loo et al. 1995.
Van Loo et al. 1999.
Van Loo 2004.
van Stuijvenberg et al. 2011.
Vandenplas et al. 2011.
Veereman 2007.
Veereman-Wauters et al.. 2011.
Vivatvakin et al. 2010.
Ward et al. 2006.
Watzl et al. 2005.
Weaver. 2005.
Westerbeek et al. 2010.
Westerbeek et al. 2011a.
Westerbeek et al. 2011b.
Westerbeek et al. 2011c.
Whelan et al. 2005.
Whelan et al. 2006.
Zheng et al. 2006.

Appendix C

000124

Pages 000125-001465 containing GRAS notices 44, 118, 236, 285, 286, 334, and 392
have been removed. They can be found in the "GRAS Notice Inventory" at
http://www.fda.gov/Food/IngredientsPackagingLabeling/GRAS/NoticeInventory/default.h
tm

Pages 001466-005452 have been removed in accordance with copyright


laws. Please see the bibliography list of the references that have
been removed from this request on pages 000104-000115.

SUBMISSION END

005453

You might also like

pFad - Phonifier reborn

Pfad - The Proxy pFad of © 2024 Garber Painting. All rights reserved.

Note: This service is not intended for secure transactions such as banking, social media, email, or purchasing. Use at your own risk. We assume no liability whatsoever for broken pages.


Alternative Proxies:

Alternative Proxy

pFad Proxy

pFad v3 Proxy

pFad v4 Proxy