You are on page 1of 8

Nutrition 9192 (2021) 111472

Contents lists available at ScienceDirect

Nutrition
journal homepage: www.nutritionjrnl.com

Applied nutritional investigation

A partially hydrolyzed formula with synbiotics supports adequate


growth and is well tolerated in healthy, Chinese term infants: A
double-blind, randomized controlled trial
Ying Wang M.D., Ph.D. a,*, Zailing Li M.D., Ph.D. b, Jie-ling Wu M.D. c, Lili Zhang M.D., Ph.D. d,
Min Liu M.D., Ph.D. e, Meizhen Tan M.D. f, Akke Botma Ph.D. g, Mengjin Liu Ph.D. h, Kelly A. Mulder Ph.D. g,
Marieke Abrahamse-Berkeveld Ph.D. g, Wei Cai M.D., Ph.D. a
a
Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
b
Peking University Third Hospital, Department of Pediatrics, Peking, China
c
Guangdong Women and Children Hospital, Department of Children Health Care, Guangzhou, China
d
Wuxi Children’s Hospital, Department of Children Health Care, Wuxi, China
e
Shanghai Public Health Clinical Center, Obstetrics Department, Shanghai, China
f
Guangzhou Women and Children’s Medical Centre, Obstetrics Department, Guangzhou, China
g
Danone Nutricia Research, Utrecht, The Netherlands
h
Danone Open Science Research Centre, Shanghai, China

A R T I C L E I N F O A B S T R A C T

Article History: Objectives: The aim of this study was to evaluate growth and gastrointestinal tolerance in infants fed a par-
Received 27 May 2021 tially hydrolyzed protein formula (pHF) with a synbiotic mixture of short-chain galacto-oligosaccharides and
Received in revised form 20 August 2021 long-chain fructooligosaccharides (scGOS/lcFOS; 9:1) and Bifidobacterium breve M-16V (test formula) com-
Accepted 24 August 2021
pared with an intact protein infant formula (IF) with scGOS/lcFOS (control formula).
Methods: This randomized, double-blind, controlled, multicenter trial enrolled healthy, fully formula-fed Chinese
Keywords:
infants (44 d) who received either the test (n = 112) or control formula (n = 112) until 17 wk of age. Fully
Tolerance
breastfed infants served as a reference (n = 60). Anthropometrics, gastrointestinal symptoms, and adverse events
Prebiotic
Probiotic
were assessed monthly. Primary outcome was weight gain in grams per day from baseline to 17 wk of age.
Infant feeding Results: Equivalence in daily weight gain (primary outcome) was demonstrated between the test and control
Safety groups (estimated mean difference [SE]: 0.36 [0.93] g/d, 90% confidence interval [CI], 1.90 to 1.18) as well
as between each IF group and the breastfed reference group (test: 0.02 [1.05] g/d, 90% CI, 1.71 to 1.75; con-
trol: 0.36 [1.04] g/d, 90% CI, 1.35 to 2.08). There were no clinically relevant differences in gastrointestinal
tolerance or adverse events between the formula groups.
Conclusion: A pHF with synbiotics supports adequate growth and is well tolerated in healthy, term-born Chi-
nese infants. Additionally, infant growth and gastrointestinal tolerance measures of both IF groups were
comparable to the breastfed group and can be considered suitable and well tolerated for use.
© 2021 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/by/4.0/)

Introduction

Human milk (HM) provides the best nutrition for infants, sup-
Danone Open Science Research Center Shanghai provided the funding to conduct
the study. YW, ZL, LZ, ML, MT, and WC were responsible for the conceptualization porting them through critical periods of early life. Notably, HM
of the study and the investigation. AB, ML, KM, and MAB were involved in the study contains bioactive and immunologic components that may modu-
methodology. KM, AB, and MAB were involved in the writing of the original draft. late the infant immune system and have benefits to growth, brain,
All authors were involved in the writing, review, and editing of the final manuscript. and gastrointestinal (GI) development [13]. For infants not
AB, KM, and MAB are employees of Danone Nutricia Research, Utrecht, The Nether-
receiving HM, specifically designed infant formulas (IFs) are
lands. ML is an employee at Danone Open Science Research Centre, Shanghai, China.
The remaining authors have no conflicts of interest to declare. required, and continuous efforts are made to improve the composi-
*Corresponding author: Fax: 86-21-65791316. tion of IF to bring functionality closer to HM.
E-mail address: 13611884226@126.com (Y. Wang).

https://doi.org/10.1016/j.nut.2021.111472
0899-9007/© 2021 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/)
2 Y. Wang et al. / Nutrition 9192 (2021) 111472

Cow’s milk protein is the most common source of protein used sequences, which were then coded onto the IF tins. The unique code corresponded
in IF and may be present as intact protein or as partially or exten- to either the test or control IFs, which were stored at the study site. Upon study
enrollment, the investigator accessed a central interactive web-response system
sively hydrolyzed protein, specifically designed for infants with
to obtain the randomly assigned code for each infant. For twin or sibling infants
cow’s milk allergy [46]. Initially, partially hydrolyzed protein for- both participating in the study, one infant was randomized and the other was
mulas (pHFs) were developed to reduce the allergenicity of and assigned to receive the same IF. The tins with the corresponding code were dis-
risk for allergic sensitization by cow’s milk protein-based formulas pensed to the parent of the infant during study visits with the instruction to feed
only the assigned study IF ad libitum during the intervention period. Parents were
[7,8]. More recently, pHF has been suggested to have potential ben-
offered the opportunity to continue with the study product up to 6 mo of age on a
efits in functional GI disorders [7,911]. Due to limited data evalu- voluntary basis, at which point they would switch to a follow-on formula of their
ating growth adequacy and GI tolerance of pHFs in healthy IF-fed choice.
infants, expert groups have called for studies evaluating both the All parents and study staff were blinded to IF assignment. The study was con-
short- and long-term health outcomes [7,9]. ducted in compliance with the principles of the Declaration of Helsinki and all pro-
cedures were approved by Ethical Review Boards of participating hospitals. Before
Increasing evidence suggests that GI development including the screening, written informed consent was obtained from parent(s)/legal guardians
gut microbiota has a role in supporting optimal infant develop- (parents) 18 y of age for each infant.
ment [12,13]. Environmental factors, including diet, influence
microbiota in early life, with well-characterized differences
Study products
between HM-fed and standard IF-fed infants [1417]. Inclusion of
prebiotics, probiotics, or a combination of both (termed synbiotics) The test and control IFs were compliant with the China guideline: National
in IF has resulted in stool microbiota closer to that of HM-fed Food Safety standard General Guideline for Formula for Special Medical Purposes
Intended for infants (GB 25596-2010; test) and National Food Safety Standard
infants and may provide a benefit to nutrient digestion and absorp-
Infant Formula (GB10765-2010; control) and manufactured according to divisional
tion, as well as the development of the immune system [1620]. quality standards (ISO 22000; Table 1). Both IFs were provided in identical 800-g
To our knowledge, this prospective, randomized, double-blind, tins.
controlled trial was the first aimed to demonstrate suitability of a The test pHF was a partially hydrolyzed non-ultrafiltrated cow’s milk protein-
whey protein-based pHF with a specific synbiotic composition based IF containing a synbiotic mixture of 0.8 g/100 mL scGOS/lcFOS (9:1 ratio)
and B. breve M-16V (3 £ 107 CFU/g). The control was a commercially available
comprised of the prebiotic mixture of short-chain galacto-oligosac- standard IF (Aptamil Pronutra) and is an intact cow’s milk protein-based IF con-
charides and long-chain fructooligosaccharides (scGOS/lcFOS, 9:1) taining a prebiotic mixture of 0.8 g/100 mL scGOS/lcFOS (9:1 ratio). Although
and Bifidobacterium breve M-16V in healthy, term-born infants. energy content was very similar in both products, it should be noted that the test
The primary objective of the present study was to investigate pHF contained a higher concentration of carbohydrates (7.2 versus 6.7 g/100 mL)
compensated by a lower level of fat content (3.4 versus 3.5 g/100 mL) compared
growth adequacy, defined as equivalence in daily weight gain, of with the control IF.
infants who received the pHF compared with those fed a standard
IF containing intact protein and scGOS/lcFOS. Secondary outcomes
Measurements
included other anthropometrics, parent-reported GI tolerance,
stool characteristics, and adverse events (AEs). As a reference, a The primary objective of this study was to evaluate equivalence of daily
group of breastfed infants was included. weight gain (g/d) from baseline to 17 wk of age between the test and control
groups, with a secondary objective to compare each IF group with the breastfed
Participants and methods group. Additional secondary objectives included equivalence of daily length and
head circumference gain, anthropometric measure z-scores, GI tolerance, and
Participants occurrence of AEs.
Infants were assessed during study visits (V) at baseline (V1), 4 (V2), 8 (V3), 13
This multicenter study included infants recruited from six study centers in (V4), and 17 (V5) wk of age. For infants randomized at 28 to 33 d of age, V2 coin-
four cities in China; Shanghai (Xinhua Hospital, School of Medicine, Shanghai Jiao cided with V1, and for infants randomized >33 d of age, V2 was not applicable.
Tong University; Shanghai Public Health Clinical Center), Beijing (Peking Univer- After written informed consent and checking of eligibility, the baseline character-
sity Third Hospital), Guangzhou (Guangdong Province Women and Children Hos- istics, including relevant medical history, were collected by interview at V1. Fully
pital; Guangzhou Women and Children's Medical Centre), and Wuxi (Wuxi formula-fed infants were randomized to one of the two study products and fully
Children's Hospital). breastfed infants enrolled in the reference group. Anthropometric measurements
Healthy, term-born infants, 44 d of age whose parent(s) autonomously were taken at each visit. Infants were weighed naked to the nearest gram on a cali-
decided to fully formula feed were invited to participate. Inclusion criteria was as brated electronic weighing scale (Seca 276, Seca, Germany). Recumbent length of
follows: the infants was measured to the nearest 0.1 cm with a length board (Seca 416,
Seca). A non-stretchable measuring tape (Seca 201, Seca) was used to measure
head circumference. Anthropometric measurements were taken twice, with a
 Chinese ethnicity,
third taken if a substantial difference between measurements was recorded
 Birth weight within the 10th to 90th percentile of INTERGROWTH-21st Inter-
(>50 g for weight and >5 mm for length and head circumference). The two closest
national Standards [21],
measurements were averaged for the statistical analysis.
 Head circumference at enrollment within normal range for age and sex (within
§2 SD World Health Organization [WHO] Child Growth Standards) [22],
 No current or previous illness/condition, and
 No participation in any other study involving investigational/marketed prod- Table 1
ucts. Composition of intervention infant formulas.

Unit Test Control


Fully breastfed infants were enrolled in the breastfed reference group if they
met these inclusion criteria and additionally their mothers had the intention to Energy kcal/100 mL 66 65
fully breastfeed until 17 wk of age and were not participating in any other clinical Fat g/100 mL 3.4 3.5
study involving other products during pregnancy/lactation. Saturates g/100 mL 1.6 1.6
Monounsaturates g/100 mL 1.2 1.3
Polyunsaturates g/100 mL 0.5 0.7
Study design
Protein g/100 mL 1.5 1.4
Whey g/100 mL 1.5 0.9
This was a double-blind, randomized controlled, two-arm parallel group
Casein g/100 mL  0.5
growth equivalence trial. This trial was registered in ClinicalTrials.gov (as
Carbohydrates g/100 mL 7.2 6.7
NCT03520764). Fully IF-fed infants were block randomized (block size of four) on
scGOS/lcFOS (9:1) g/100 mL 0.8 0.8
a 1:1 basis stratified by sex, to receive the test or control formula until 17 wk of
Bifidobacterium breve M-16V CFU/g 3 £ 107 
age. The PLAN procedure of SAS statistical software was used to create the ran-
domization sequences. A study-independent statistical programmer generated the scGOS/lcFOS, short-chain galacto-oligosaccharides/long-chain fructooligosaccharides
Y. Wang et al. / Nutrition 9192 (2021) 111472 3

After V1, parents completed a 7-d paper diary before each visit, which col- volume prepared minus the volume leftover and the mean daily IF intake per day
lected information on IF intake, GI symptoms, any illness or symptoms, and use of (mL/d) and body weight (mL/kg) were calculated for each visit. For both study IF
medication or supplements. For GI symptoms, stool consistency and frequency as intake and parent-reported GI tolerance parameters, three of seven completed
well as regurgitation and vomiting were recorded. During the 7-d period, stool diary days with no missing data was required. Stool characteristics included mean
consistency was scored for each defecation by the parents according to the stool frequency (number of stools per day) and stool consistency (percentage of
Amsterdam Stool Scale (watery, soft, formed, hard) [23] and frequency was the stools with certain consistency over all diary days within the visit week). The
total daily number of defecations. Number of occurrences was recorded for both occurrence of frequent watery stools (passage of three or more watery stools in a
regurgitation (return of the milk into the mouth without force) and vomiting day) and the occurrence of infrequent hard stools (two or less defecations per
(return of milk into the mouth with force). The investigator reviewed the diary week and [if any] with a hard consistency) were also calculated from the stool con-
and documented all AEs and any concomitant medications at each visit. A postin- sistency and frequency data for each visit. Regurgitation and vomiting were ana-
tervention follow-up visit was planned at 12 mo of age, which is beyond the scope lyzed as the occurrence of regurgitation or vomiting at least once on 1 d and the
of the present study and will be presented in a subsequent publication. occurrence of frequent regurgitation or vomiting (2 to 3 d with three or more
regurgitation or vomiting episodes).
Statistical analysis Unless otherwise specified, parameters were summarized with descriptive
statistics and for comparison between groups; two sample t tests or MannWhit-
The a priori assumptions of the primary objective included a margin of equiva- ney U tests were used for continuous data. Dichotomous outcome parameters
lence of §3 g/d [24], an equal within-group SD of 6 g/d in weight gain in both were compared with the MiettinenNurminen Score test. For the AE analyses,
intervention groups, and a difference between the intervention groups of 0.5 g/d. estimates of the risk differences between IF groups were also provided along with
A required sample size of 224 (112 per group) was calculated with the two one- the corresponding MiettinenNurminen 95% confidence intervals (CIs) for all
sided tests (a = 0.05; power of 0.80; dropout/non-compliance rate of 30%). An events that occurred in at least four participants in either IF group. For all second-
interim analysis to review AEs and confirm the sample size estimation was con- ary parameters, P-values are unadjusted. All statistical analyses were conducted
ducted after 109 randomized infants completed the visit at 17 wk of age and was according to a predefined statistical analysis plan finalized before data unblinding
evaluated by an independent data monitoring committee. It was recommended using SAS version 9.4 (SAS, Cary NC, USA).
that the study continue without modification.
The equivalence analyses for daily weight gain (primary outcome) was per-
Results
formed with the parametric growth curve mixed-effects model with a quadratic
function of time and included sex, sex £ time, sex £ time2, group, group £ time,
group £ time2, and birth weight as fixed effects and each infant’s intercept, time Participant characteristics
and time2 as random effects. The following sensitivity analyses were performed to
evaluate the robustness of the results:
From April 2018 to November 2019, we enrolled 285 healthy,
 Excluding second twin/sibling;
term-born, Chinese infants (IF fed: 224, breastfed: 60; one infant
 Excluding outliers or influential participants; withdrew before randomization; Fig. 1). In the IF groups, 190
 Adding imbalanced covariates to the model (e.g., mode of delivery, pooled infants completed V5 with 175 PP completers. For the breastfed
site). reference group, 53 infants completed V5 with 51 PP completers.
Baseline characteristics among the randomized groups were rela-
Equivalence analyses for growth outcomes used the per protocol (PP) as the pri-
tively well balanced with a few exceptions (Supplementary Table
mary analysis data set. Eligibility of data for the PP population was assessed on
blinded data before analyses and defined on a per visit level (where applicable) 1, PP). Compared with the control group, the test group had more
with major protocol deviations including not meeting inclusion criteria, a duration girls, more twins, a lower median baseline age, more Cesarean
of >7 d post-baseline to transition fully to the study IF, missing all post-baseline deliveries, and a lower incidence of familial allergy. Infant expo-
anthropometric measurements, and >3 d of non-study IF (or any IF for the breastfed sure to cigarette smoke was only reported for 5 IF-fed infants and
group) or complementary food use. In the case of infant withdrawal from the study,
all available data was considered, and no data imputation was performed.
1 breastfed infant.
A comparison of the IF groups with the breastfed reference group was also done Due to the severe acute respiratory syndrome coronavirus 2
as a secondary objective using the above primary parameter analysis model includ- (SARS-CoV-2) pandemic, some parents were not allowed or not
ing all three groups. For comparison with the WHO growth standard, anthropomet- willing to attend the study site. Ten infants, of which 9 were part
ric z-scores were calculated using a macro provided by the WHO [22].
of the PP population (6 in the test and 3 in the control group) had
The all subjects treated population (infants who received some study IF) was
used for all analyses of parent-reported GI tolerance parameters (stool characteris- anthropometric measurements taken by their parents using house-
tics, regurgitation, vomiting), and AEs. Study product intake was derived as the hold measuring tools after providing clear instructions and

Fig. 1. Participant flow through the study from enrollment to study completion for the ASE, ASR, completed, and PP populations. One IF-fed infant withdrew immediately
after enrollment before randomization. An AE was the reason for early termination for three infants in the test group, all of which were cows’ milk protein allergy or intoler-
ance, and two infants in the control group, one for lactose intolerance and one for eczema and rotavirus gastroenteritis. AE, adverse event; ASE, all subjects enrolled; ASR, all
subjects randomized; AST, all subjects treated; BF, breastfed; IF, infant formula; PP, per protocol.
4 Y. Wang et al. / Nutrition 9192 (2021) 111472

Table 2
Daily parent-reported infant formula intake per day and per kilogram body weight in the per-protocol population*

mL/d mL¢kg¢d1

Test Control Test Control


n (missingy) Mean (SD) n (missing) Mean (SD) n (missing) Mean (SD) n (missing) Mean (SD)

All subjects randomized population


Visit 2 83 (7) 783 (221) 69 (6) 804 (175) 83 (7) 182 (43.9) 69 (6) 185 (39.2)
Visit 3 93 (0) 927 (210) 96 (2) 894 (189) 93 (0) 172 (33.4) 96 (2) 162 (30.5)
Visit 4 93 (1) 941 (246) 96 (0) 916 (205) 93 (1) 147 (34.6) 96 (0) 141 (30.4)
Visit 5 94 (0) 970 (251) 96 (0) 974 (197) 94 (0) 138 (31.4) 96 (0) 137 (27.2)
Per-protocol population
Visit 2 73 (6) 809 (215) 68 (3) 803 (176) 73 (6) 187 (41.8) 68 (3) 185 (39.4)
Visit 3 86 (0) 922 (212) 94 (1) 894 (188) 86 (0) 171 (34.1) 94 (1) 162 (30.3)
Visit 4 86 (0) 927 (243) 93 (0) 914 (206) 86 (0) 146 (34.8) 93 (0) 141 (30.6)
Visit 5 85 (0) 961 (239) 90 (0) 975 (194) 85 (0) 137 (30.6) 90 (0) 137 (25.8)
*Average daily consumed volume per visit was calculated if at least three diary days were filled out completely, including volume of water and number of scoops used and the
volume of the left over reported for each day.
y
Missing includes those who were participating in the study but did not provide 3 d of completed study product intake data in the diary. Infants who withdrew from the
study are not included as missing.

guidance documents how to measure their infant as accurate as (0.93) g/d (90% CI, 1.90 to 1.18 g/d) for the PP population. Also
possible. During only 11 of the 873 observations in these random- after exclusion of home measurements, equivalence was demon-
ized groups, anthropometric measurements were collected by strated for the PP population with estimated differences in means
parents (1.3%). The blinded measurements were discussed by the (SE) of daily weight gain between the IF groups of 0.28 (0.94) g/d
study team using the longitudinal data plotted in growth charts (90% CI, 1.83 to 1.28). The equivalence was confirmed with the
before statistical analysis and considered accurate and reliable and, sensitivity analyses and the all subjects randomized population.
therefore, included in the analyses. It is of note that none of the Equivalence in daily weight gain was also achieved between the
outliers or influential participants identified during statistical anal- breastfed reference and both the test and control groups. The esti-
ysis had any home measurements taken. However, acknowledging mated mean difference (SE) between the test and breastfed refer-
the potential higher inaccuracy of home measurements and the ence groups was 0.02 (1.05) g/d (90% CI, 1.71 to 1.75 g/d) and
importance of standardization of measurements, an additional between the control and breastfed reference groups was 0.36
(post hoc) analysis was performed excluding these home measure- (1.04) g/d (90% CI, 1.35 to 2.08 g/d).
ments for confirmation. Equivalence in daily length gain (mm/d) was achieved between
the test and control groups, but not between either IF groups or
Study product intake the breastfed reference group. The difference in estimated means
(SE) between the test and control group was 0.01 (0.02) mm/d
The average daily IF intake was similar among IF-fed groups (90% CI, 0.05 to 0.03 mm/d). After exclusion of home measure-
over the intervention period (Table 2). The mean (SD) daily IF ments, equivalence was confirmed with a difference of estimated
intake at V2 was 809 (215) mL/d and 803 (176) mL/d for the test means (SE) between both formula groups of 0.001 (0.02) mm/d
and control groups, respectively, increasing to 961 (239) mL/d in (90% CI, 0.04 to 0.04 mm/d). Between the test and breastfed refer-
the test group and 975 (194) mL/d in the control group at V5 (PP). ence groups the difference in estimated means (SE) was 0.08 (0.03)
mm/d (90% CI, 0.040.13 mm/d) and between the control and
Growth outcomes breastfed reference groups it was 0.09 (0.03) mm/d (90% CI,
0.040.14 mm/d). For daily head circumference gain, equivalence
Growth outcomes were highly similar among both IF groups was achieved between the test and control groups, also when
and the breastfed reference group (Table 3). An equivalent daily home measurements were excluded, as well as between each IF
weight gain (g/d) between the test and control groups from base- group and the breastfed reference group (data not shown).
line to 17 wk was demonstrated. The difference in estimated Compared with the WHO growth standards, the mean z-scores
means (SE) of daily weight gain between the IF groups was 0.36 for weight-for-age, length-for-age, head-circumference-for-age,

Table 3
Gain in weight, length, and head circumference from baseline to visit 5 (17 wk of age) in the per-protocol population*

Test (n = 89) Control (n = 97) Breastfed (n = 56)

Mean Estimate (SE) 95% CI Mean Estimate (SE) 95% CI Mean Estimate (SE) 95% CI

Weight gain
g 3242 (67.9) 31083376 3279 (64.9) 31503407 3209 (83.5) 30443373
g/d 32 (0.67) 30.733.3 32.4 (0.64) 31.133.6 31.9 (0.83) 30.233.5
Length gain
mm 119.4 (1.76) 115.9122.9 120 (1.74) 116.6123.5 109.9 (2.27) 105.4114.4
mm/d 1.18 (0.02) 1.141.21 1.19 (0.02) 1.151.22 1.09 (0.02) 1.051.14
Head circumference gain
mm 55.3 (1.09) 53.157.4 54.9 (1.09) 52.757 54.1 (1.40) 51.356.8
mm/d 0.55 (0.01) 0.520.57 0.54 (0.01) 0.520.56 0.54 (0.01) 0.510.56
*Data were modeled with a parametric growth curve mixed model with a quadratic function of time.
Y. Wang et al. / Nutrition 9192 (2021) 111472 5

Fig. 2. Estimated mean (§95% CI) WHO Growth Standard z-scores weight-for-age (A), length-for-age (B), BMI-for-age (C), head circumference-for-age (D), mid-upper arm cir-
cumference-for-age (E), per age category for the test, control, and the breastfed reference per-protocol population. BMI, body mass index; WHO, World Health Organization.

and body mass index-for-age were all within or very close to the § breastfed reference groups at all timepoints (Supplementary Table
0.5 SD bandwidth for the test, control, and breastfed reference 3). In general, the occurrence of both regurgitation and vomiting
groups, suggesting adequate infant growth (Fig. 2). Highly similar decreased over the intervention period from V2 to V5 for all
growth z-score patterns were observed after exclusion of home infants.
measurements (data not shown). The mid-upper arm circumfer-
ence was measured at 13 and 17 wk of age and the corresponding Adverse events
median z-score was between the +0.5 and +1 SD bandwidth at 13
wk and close to +1 SD at 17 wk. The occurrence of at least one AE was reported for 100 (47.8%)
IF-fed infants (test: 52.9%; control: 43.0%, P = 0.151) and 16 (26.7%)
Parent-reported gastrointestinal tolerance breastfed infants. There were no statistically significant nor clini-
cally relevant differences between the number or type of AEs or
Average stool frequency decreased slightly over the study serious AE (SAEs) between the IF groups. The most common AEs
period from a median (Q1, Q3) of 1.9 (1.13.3) stools per day at V2 were skin and subcutaneous tissue disorders (test: 22 [21.6%]; con-
to 1 (0.71.4) at V5 among the IF groups (Fig. 3). There was only trol: 20 [18.7%]; breastfed: 6 [10%]), with eczema being the most
one minor statistically significant difference between the IF reported for 18 (17.6%), 17 (15.9%), and 5 (8.3%) of infants in the
groups; the median (Q1, Q3) of 1 (0.71.7) stool per day in the test test, control, and breastfed groups, respectively. Infections and
group was slightly higher than in the control group of 0.9 (0.61.3; infestations (test: 16 [15.7%]; control: 20 [18.7%]; breastfed group:
P = 0.049) at V5. In the breastfed reference group, the median (Q1, 5 [8.3%]) and GI disorders (test: 16 [15.7%]; control: 13 [12.1%];
Q3) stool frequency was 4.3 (2.86) stools per day at V2 and 1.6 breastfed: 6 [10%]) were also commonly reported AEs. The number
(0.92.3) at V5. of AEs related to the study product was comparable between IF
Most stools were scored as soft, with no statistically significant groups (test: 24 [23.5%]; control: 26 [24.3%]), with skin and subcu-
differences between IF groups at any time point (Fig. 3). The per- taneous tissue disorders being the most reported as related to the
centage of watery stools among IF-fed infants was very low (Sup- study product (test: 14 [13.7%]; control: 16 [15.9%]), followed by GI
plementary Table 2). Some watery stools were reported in the disorders (test: 9 [8.8%]; control: 9 [8.4%]). None of the AEs
breastfed reference group with a median (Q3) of 0% (23.1%) and 0% reported as related to the study product were classified as severe.
(66.7%) of stools at V2 and V5, respectively. The occurrence of fre- The occurrence of at least one SAE was reported for 8 (3.8%) infants
quent watery stools was also low among IF-fed infants from V2 (test: 5 [4.9%]; control: 3 [2.8%], P = 0.430; breastfed: 0). All SAEs
(test: 8.4%, control: 7.2%, P = 0.788) to V5 (test: 2.1%; control: 4.2%, were classified as an infection and infestation and none were
P = 0.423). For the breastfed reference group, frequent watery reported as related to the study products.
stools were 34.6% and 18.9% of infants at V2 and V5, respectively.
Hard stools were rarely reported during the intervention period Discussion
among all infants (Supplementary Table 2). The occurrence of
infrequent hard stools was also rare (<1%) among all of the infants To our knowledge, this prospective, randomized, double-blind,
with no statistically significant differences between the IF groups controlled trial was the first to evaluate the suitability of a whey
at any time point. protein-based pHF with a specific synbiotic composition comprised
There were no statistically significant differences in the occur- of the prebiotic mixture scGOS/lcFOS (9:1) and B. breve M-16V
rence of parent-reported regurgitation or vomiting between the IF compared with a standard intact cow’s milk protein-based for-
groups at any time point. The occurrence of any or frequent regur- mula. Equivalence in daily weight gain was demonstrated (primary
gitation or vomiting were similar between the IF infants and outcome), and both IFs were well tolerated without any
6 Y. Wang et al. / Nutrition 9192 (2021) 111472

Fig. 3. Average stool frequency (n/d) (A) and mean percentage of stool consistency (B) for the all subjects treated population and the breastfed reference group. For stool fre-
quency, only visit 5 was statistically significantly different between the test and control groups (P = 0.049), by MannWhitney test. No statistically significant differences in
stool consistency between IF groups at any time point. Percentage of stool consistency was calculated by patient per visit, defined per consistency category as the number of
stools with certain consistency over all diary days within that visit divided by the total number of stools (of any consistency) over all diary days within that visit.

statistically significant or clinically relevant differences in number infants receiving HM had a slightly lower length between 6 and
or type of AEs. Additionally, infant growth and GI tolerance meas- 12 mo of age, and that infants in the Chinese study population
ures of both IF groups were comparable to the breastfed group and had slightly higher weight-for-age and length-for-age z-scores
can be considered as suitable for use in healthy term infants. compared with WHO standards from 2 to 12 mo of age [31].
The growth of Chinese infants in this study was comparable to Although WHO growth standards included breastfed infants
infant growth observed in studies evaluating IFs in both China from six countries, China was not represented and studies show
and Europe [2528]. We demonstrated equivalence in daily that Chinese infants and/or children may have a higher median
weight gain between both the test and control groups with the weight, length, and head circumference than WHO reference val-
breastfed reference group. In contrast, we observed a slightly ues, although not all infants in those studies were exclusively
higher length gain in both IF groups compared with the breastfed breastfed [3133]. In the present study, the weight of infants
reference group. This is consistent with other IF studies in China was only slightly higher than the WHO growth standards, with
[29,30]. One study showed that compared with IF-fed infants, mean weight-for-age z-scores around 0.5 SD among all infants by
Y. Wang et al. / Nutrition 9192 (2021) 111472 7

17 wk of age. The length-for-age z-score for the IF-fed infants sensitivity analyses and did not influence the conclusions of our
was also around 0.5 SD; whereas for the breastfed infants it was growth analyses (primary outcome). The study was conducted in
closer to 0. Together, the equivalence in daily weight gain part during the SARS-CoV-2 pandemic and as a result, several
between the IF-fed and breastfed infants in the present study as infant anthropometrics were being taken by the parents at home,
well as the comparison with the WHO growth standards con- potentially affecting the accuracy of these measurements.
firms the growth adequacy in infants of both the pHF (test) and Although the number was limited (1.3% of all measurements), and
the commercial IF (control). visual assessment of the unblinded measurements did not reveal
Several previous studies have shown that the addition of any signs for unreliability of the collected data (and therefore
scGOS/lcFOS and/or B. breve M-16 V was well tolerated [25,3436] remained part of the prespecified statistical analysis), additional
and this was confirmed in the present study combining the synbi- analyses were performed excluding these home measurements,
otic mixture with a pHF. At 17 wk of age, the median reported stool which did not reveal any significant effect on the outcomes. Addi-
frequency was slightly higher for the test participants than for the tionally, it is possible that due to the pandemic, the usual environ-
control group (1 versus 0.9 stools per day, P = 0.049, unadjusted). ment of infants was affected, for example with reduced exposure
The clinical relevance of this observation in healthy infants to external environments due to a lockdown situation. However, it
remains to be elucidated. Most notably, the median percentage of is anticipated that this has not substantially affected the key out-
stool consistency was reported as soft for 100% of stools at V3 to comes of this study.
V5 for infants in both the IF and the breastfed reference groups.
Consistent with this was the rare occurrence of infrequent hard Conclusion
stools (<1%) among all infants. It has long been documented that
breastfed infants have softer stools than IF-fed infants [3739]. In The present study demonstrated that a pHF with a synbiotic
the present study, both the test and control products contained mixture of scGOS, lcFOS, and B. breve M-16 V supports adequate
scGOS/lcFOS (0.8 g/100 mL), and the test pHF additionally con- growth, is well tolerated, and is suitable for use in healthy, term-
tained B. breve M-16V. The stool softening effect of both scGOS/ born infants. A follow-up of these infants at 12 mo of age aims to
lcFOS and B. breve M-16V has previously been attributed to the evaluate the intervention effects on longer-term outcomes.
bifidogenic effect of these pre- and probiotics [35,40,41]. Although
a similar effect might have occurred in the present study, explain-
Author Roles and Responsibilities
ing the soft stools observed, this requires further investigation to
confirm. In a study comparing pHF with an intact protein IF, both
Conceptualisation, Y.W., Z.L., L.Z., M.L., M.T., W.C.; Investigation,
containing scGOS/lcFOS, »80% of infants had a stool consistency
Y.W., Z.L., L.Z., M.L., M.T., W.C.; Methodology, A.B., M.L., K.M., and
reported by parents as soft, and hard stools were nearly absent
M.A-B.; Writing  original draft, K.M., A.B., and M.A-B.; Writing 
[25]. A previous epidemiologic survey in China showed functional
review & editing, Y.W., Z.L., L.Z., M.L., M.T., A.B., M.L., K.M., M.A-B.,
constipation prevalence in 6.2% in infants 0 to 3 mo of age [42].
W.C.
Additionally, there appeared to be no difference in intake between
the test and control groups, indicating a similar acceptability of
both products by infants and their parents. Overall, GI tolerance Declaration of interests
was comparable between infants in both the test and control
groups with the breastfed reference group, particularly with The authors declare that they have no known competing finan-
respect to stool consistency and regurgitation. cial interests or personal relationships that could have appeared to
In the present study, number and type and relatedness of AEs influence the work reported in this paper.
were not statistically significantly nor clinically relevantly different
between IF groups. The nature of AEs were those typically Acknowledgments
expected in young infants [42,43]. Eczema was the most commonly
reported AE in the present study (test: 17.6%; control: 15.9%, P > The authors acknowledge the families who participated in this
0.05), and was higher than what has been reported by other study. They also acknowledge the research staff at the study sites
growth and safety trials of pHF and IF in European infants for their hard work and dedication. The contribution of the Dragon
[25,26,44]. However, in the present study, eczema was also study team, especially Stephanie Song from Danone Open Science
reported as an AE for 8.3% of the breastfed infants, which might Research Center (Shanghai), Jasmine Ho from Danone Research
indicate a population-specific increased sensitivity to such AEs. (Singapore), and Maya Marintcheva-Petrova, Ramona Grigorescu,
Infant cohort studies in other countries have shown that several Meint-Jan Elzinga, Berdine Draijer, Marion Kaspers, and Sonakshi
factors are associated with eczema development in infants, includ- Shankar from Danone Nutricia Research (Utrecht) is also acknowl-
ing maternal history of allergy or eczema, cesarean delivery, antibi- edged.
otic use during pregnancy, and East Asian ethnicity [4547]; the
present study population did have a number of these risk factors. Supplementary materials
Eczema development is clearly multifactorial and given the simi-
larity in prevalence between intervention groups and prevalence Supplementary material associated with this article can be
in the breastfed reference group, the occurrence in our study is not found in the online version at doi:10.1016/j.nut.2021.111472.
considered a safety concern.
This study had several key strengths, including its randomized, References
double-blind, controlled design, the confirmed powered sample
size, and the rigorous, strict high-quality standards applied during [1] Anderson CE, Whaley SE, Crespi C, Wang MC, Chaparro MP. Every month mat-
conduct and evaluation. The authors also had several limitations to ters: longitudinal associations between exclusive breastfeeding duration, child
address. Despite the randomized design, there were some notable growth and obesity among WIC-participating children. J Epidemiol Commu-
nity Health 2020. https://doi.org/10.1136/jech-2019213574.
differences in baseline characteristics between the IF groups. The [2] Horta BL, de Sousa BA, de Mola CL. Breastfeeding and neurodevelopmental
potential effect of these differences was investigated in the outcomes. Curr Opin Clin Nutr Metab Care 2018;21:174–8.
8 Y. Wang et al. / Nutrition 9192 (2021) 111472

[3] Victora CG, Bahl R, Barros AJ, Franca GV, Horton S, Krasevec J, et al. Breastfeed- specific oligosaccharides, 20 -FL, and milk fat supports adequate growth, is safe
ing in the 21st century: epidemiology, mechanisms, and lifelong effect. Lancet and well-tolerated in healthy term infants: a double-blind, randomised, con-
2016;387:475–90. trolled, multi-country trial. Nutrients 2020:12.
[4] Muraro A, Werfel T, Hoffmann-Sommergruber K, Roberts G, Beyer K, Bindslev- [27] Wu SL, Ding D, Fang AP, Chen PY, Chen S, Jing LP, et al. Growth, gastrointestinal
Jensen C, et al. EAACI food allergy and ana-phylaxis guidelines: diagnosis and tolerance and stool characteristics of healthy term infants fed an infant for-
management of food allergy. Allergy 2014;69:1008–25. mula containing hydrolyzed whey protein (63%) and intact casein (37%): a ran-
[5] Fleischer DM, Spergel JM, Assa'ad AH, Pongracic JA. Primary prevention of domized clinical trial. Nutrients 2017;9.
allergic disease through nutritional interventions. J Allergy Clin Immunol Pract [28] Li X, Peng Y, Li Z, Christensen B, Heckmann AB, Stenlund H, et al. Feeding
2013;1:29–36. infants formula with probiotics or milk fat globule membrane: a double-blind,
[6] Osborn DA, Sinn J. Formulas containing hydrolysed protein for prevention of randomized controlled trial. Front Pediatr 2019;7:347.
allergy and food intolerance in infants. Cochrane Database Syst Rev 2006. [29] Tian Q, Gao X, Sha T, Chen C, L L, He Q, et al. Effect of feeding patterns on
10.1002/14651858.CD003664.pub3, CD003664. growth and nutritional status of children aged 024 months: a Chinese cohort
[7] Vandenplas Y, Alarcon P, Fleischer D, Hernell O, Kolacek S, Laignelet H, et al. study. PLoS One 2019;14:e0224968.
Should partial hydrolysates be used as starter infant formula? A Working [30] Zong XN, Li H, Zhang YQ, Wu HH. Growth performance comparison of exclu-
Group Consensus. J Pediatr Gastroenterol Nutr 2016;62:22–35. sively breastfed infants with partially breastfed and formula fed infants. PLoS
[8] Iskedjian M, Szajewska H, Spieldenner J, Farah B, Berbari J. Meta-analysis of a One 2020;15:e0237067.
partially hydrolysed 100%-whey infant formula vs. extensively hydrolysed [31] Ouyang F, Jiang F, Tao F, Xu S, Xia Y, Qiu X, et al. Growth patterns from birth to
infant formulas in the prevention of atopic dermatitis. Curr Med Res Opin 24 months in Chinese children: a birth cohorts study across China. BMC
2010;26:2599–606. Pediatr 2018;18:344.
[9] Vandenplas Y, Latiff AHA, Fleischer DM, Gutierrez-Castrellon P, Miqdady MS, [32] Zong XN, Li H. Construction of a new growth references for China based on
Smith PK, et al. Partially hydrolyzed formula in non-exclusively breastfed urban Chinese children: comparison with the WHO growth standards. PLoS
infants: a systematic review and expert consensus. Nutrition 2019;57:268–74. One 2013;8:e59569.
[10] Nguyen TT, Bhandari B, Cichero J, Prakash S. In vitro digestion of infant formu- [33] Tian Q, Gao X, Sha T, He Q, Cheng G, Wu X, et al. Differences between WHO
lations with hydrolysed and non-hydrolysed proteins from dairy and soybean. growth standards and China growth standards in assessing the nutritional sta-
Food Funct 2016;7:4908–19. tus of children aged 036 months old. Int J Environ Res Public Health 2019;17.
[11] Vandenpla Y, Gutierrez-Castrellon P, Velasco-Benitez C, Palacios J, Jaen D, [34] Huet F, Abrahamse-Berkeveld M, Tims S, Simeoni U, Beley G, Savagner C, et al.
Ribeiro H, et al. Practical algorithms for managing common gastrointestinal Partly fermented infant formulae with specific oligosaccharides support ade-
symptoms in infants. Nutrition 2013;29:184–94. quate infant growth and are well-tolerated. J Pediatr Gastroenterol Nutr
[12] Ihekweazu FD, Versalovic J. Development of the pediatric gut microbiome: 2016;63:e43–53.
impact on health and disease. Am J Med Sci 2018;356:413–23. [35] Abrahamse-Berkeveld M, Alles M, Franke-Beckmann E, Helm K, Knecht R,
[13] Nauta AJ, Ben Amor K, Knol J, Garssen J, van der Beek EM. Relevance of pre- and Kollges R, et al. Infant formula containing galacto-and fructo-oligosaccharides
postnatal nutrition to development and interplay between the microbiota and and Bifidobacterium breve M-16V supports adequate growth and tolerance in
metabolic and immune systems. Am J Clin Nutr 2013;98:586S–93S. healthy infants in a randomised, controlled, double-blind, prospective, multi-
[14] Fallani M, Young D, Scott J, Norin E, Amarri S, Adam R, et al. Intestinal micro- centre study. J Nutr Sci 2016;5:e42.
biota of 6-week-old infants across Europe: geographic influence beyond deliv- [36] Chua MC, Ben-Amor K, Lay C, Neo AG, Chiang WC, Rao R, et al. Effect of synbi-
ery mode, breast-feeding, and antibiotics. J Pediatr Gastroenterol Nutr otic on the gut microbiota of cesarean delivered infants: a randomized, dou-
2010;51:77–84. ble-blind, multicenter study. J Pediatr Gastroenterol Nutr 2017;65:102.
[15] Ma J, Li Z, Zhang W, Zhang C, Zhang Y, Mei H, et al. Comparison of gut micro- [37] Weaver LT, Ewing G, Taylor LC. The bowel habit of milk-fed infants. J Pediatr
biota in exclusively breast-fed and formula-fed babies: a study of 91 term Gastroenterol Nutr 1988;7:568–71.
infants. Sci Rep 2020;10:15792. [38] Tham EB, Nathan R, Davidson GP, Moore DJ. Bowel habits of healthy Australian
[16] Peroni DG, Nuzzi G, Trambusti I, Di Cicco ME, Comberiati P. Microbiome com- children aged 02 years. J Paediatr Child Health 1996;32:504–7.
position and its impact on the development of allergic diseases. Front Immu- [39] Moretti E, Rakza T, Mestdagh B, Labreuche J, Turck D. The bowel movement
nol 2020;11:700. characteristics of exclusively breastfed and exclusively formula fed infants dif-
[17] Hascoet JM, Hubert C, Rochat F, Legagneur H, Gaga S, Emady-Azar S, et al. fer during the first three months of life. Acta Paediatr 2019;108:877–81.
Effect of formula composition on the development of infant gut microbiota. J [40] MoroG Minoli I, Mosca M, Fanaro S, Jelinek J, Stahl B, et al. Dosage-related bifi-
Pediatr Gastroenterol Nutr 2011;52:756–62. dogenic effects of galacto- and fructooligosaccharides in formula-fed term
[18] Cukrowska B, Bierla JB, Zakrzewska M, Klukowski M, Maciorkowska E. The infants. J Pediatr Gastroenterol Nutr 2002;34:291–5.
relationship between the infant gut microbiota and allergy. The role of Bifido- [41] Wong CB, Iwabuchi N, Xiao JZ. Exploring the science behind Bifidobacterium
bacterium breve and prebiotic oligosaccharides in the activation of anti-aller- breve M-16V in infant health. Nutrients 2019:11.
gic mechanisms in early life. Nutrients 2020:12. [42] Wenjing J, Aimin L, Chengyi Q, Ruiyun S, Zhuang W, Yang M. Epidemiologic
[19] Vandenplas Y, De Greef E, Veereman G. Prebiotics in infant formula. Gut survey on the prevalence and distribution of infants' common gastrointestinal
Microbes 2014;5:681–7. symptoms in 7 cities in China: a population-based study. Chin J Epidemiol
[20] Salminen S, Stahl B, Vinderola G, Szajewska H. Infant formula supplemented 2018;39:1179–83.
with biotics: current knowledge and future perspectives. Nutrients 2020:12. [43] Vandenplas Y, Abkari A, Bellaiche M, Benninga M, Chouraqui JP, Cokura F, et al.
[21] Villar J, Cheikh Ismail L, Victora CG, Ohuma EO, Bertino E, Altman DG, et al. Prevalence and health outcomes of functional gastrointestinal symptoms in
International standards for newborn weight, length, and head circumference infants from birth to 12 months of age. J Pediatr Gastroenterol Nutr
by gestational age and sex: the Newborn Cross-Sectional Study of the INTER- 2015;61:531–7.
GROWTH-21st Project. Lancet 2014;384:857–68. [44] Rodriguez-Herrera A, Mulder K, Bouritius H, Rubio R, Munoz A, Agosti M, et al.
[22] WHO Multicentre Growth Reference Study Group. WHO Child growth stand- Gastrointestinal tolerance, growth and safety of a partly fermented formula
ards based on length/height, weight and age. Acta Paediatr 2006;450:76–85. with specific prebiotics in healthy infants: a double-blind, randomized, con-
[23] Bekkali N, Hamers SL, Reitsma JB, Van Toledo L, Benninga MA. Infant stool form trolled trial. Nutrients 2019:11.
scale: development and results. J Pediatr 2009;154:521–6. e521. [45] Gao X, Yan Y, Zeng G, Sha T, Liu S, He Q, et al. Influence of prenatal and early-
[24] American Academy of Pediatrics. Clinical testing of infant formula with respect life exposures on food allergy and eczema in infancy: a birth cohort study.
to nutritional suitability for term infants. Elk Grove Village, IL: Author 1988. BMC Pediatr 2019;19:239.
[25] Picaud JC, Pajek B, Arciszewska M, Tarczon I, Escribano J, Porce R, et al. An [46] Martin PE, Koplin JJ, Eckert JK, Lowe AJ, PonsonbyA.L Osborne NJ, et al. The
infant formula with partially hydrolyzed whey protein supports adequate prevalence and socio-demographic risk factors of clinical eczema in infancy: a
growth and is safe and well-tolerated in healthy, term infants: a randomized, population-based observational study. Clin Exp Allergy 2013;43:642–51.
double-blind, equivalence trial. Nutrients 2020:12. [47] Rehbinder EM, Advocaat Endre KM, Lodrup Carlsen K, Asarnoj A, Stensby Bains
[26] Vandenplas Y, de Halleux V, ArciszewskaM Lach, P Pokhylko V, Klymenko V, KE, Berents TL, et al. Predicting Skin barrier dysfunction and atopic dermatitis
et al. A partly fermented infant formula with postbiotics including 30 -GL, in early infancy. J Allergy Clin Immunol Pract 2020;8:664–73. e665.

You might also like