Skip to main content
Erschienen in: Molecular and Cellular Pediatrics 1/2023

Open Access 01.12.2023 | Research

Real-world evidence study on tolerance and growth in infants fed an infant formula with two human milk oligosaccharides vs mixed fed and exclusively breastfed infants

verfasst von: Frank Jochum, Martina Meyer-Krott, Tina Hübler, Maja Lorenz, Raffi Bedikian, Joseph Zakarian, Anja Litzka, Guido Judex, Holger Hertzberg, Daniela Klee, Lothar Maurer, Martin Schacht, Adnan Al-Radhi, Jan Maier, Alexander Kröckel, Christian Faustmann, Luca Lavalle, Samir Dahbane

Erschienen in: Molecular and Cellular Pediatrics | Ausgabe 1/2023

Abstract

Introduction

Human milk oligosaccharides (HMOs) are important components of human milk having diverse functions in the development of infants. Randomized controlled trials (RCTs) have demonstrated that infant formulas with the HMOs 2′-fucosyllactose (2′FL) and lacto-N-neotetraose (LNnT) are safe, well-tolerated, and support normal growth. This study aimed to generate real-world evidence (RWE) on growth and gastrointestinal (GI) tolerance in infants consuming a formula with 1 g/L 2′FL and 0.5 g/L LNnT, including a mixed feeding group not studied before in RCTs.

Participants and methods

This 8-week open-label prospective multicenter study was conducted in Germany and Austria, and included groups of healthy, exclusively breastfed infants (BF), exclusively formula-fed infants (FF) who received the HMO-formula, and infants mixed fed with both HMO formula and human milk (MF). Co-primary outcomes were anthropometry and gastrointestinal tolerance via validated Infant Gastrointestinal Symptom Questionnaire (IGSQ). Secondary outcomes included formula satisfaction and adverse events (AEs).

Results

One-hundred six infants completed the study (46 FF, 22 MF, and 38 BF). Mean anthropometric z-scores were comparable between groups and generally within ± 0.5 of WHO medians at week 8. IGSQ composite scores demonstrated good GI tolerance in all groups with no significant group differences at week 4 or 8. IGSQ composite scores in FF improved during the course of the study and parents provided high satisfaction ratings for the HMO-formula. Four potentially product-related AEs were reported in FF (no in MF).

Conclusions

In this RWE study examining an infant formula with HMOs, growth and GI tolerance outcomes were confirming the good tolerance and safety of this early feeding option previously reported in RCTs.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Human milk oligosaccharides (HMOs) are found in abundance in human milk and make up the largest solid component after lactose and lipids [14]. HMOs can become characterized as complex group of more than 200 different, nondigestible, and non-nutritional carbohydrates, providing an energy source for beneficial intestinal bacteria. There is evidence that HMOs improve the host defense by strengthening the gut barrier and immune-modulating effects and other mechanisms. Bovine milk, in contrast to human milk, contains relatively low levels of oligosaccharides, and the prevalence of fucosylated oligosaccharides, in particular, is quite low [5]. 2′-fucosyllactose (2′FL) is a trisaccharide composed of glucose, galactose, and fucose and is one of the most abundant HMOs. Levels of 2′FL vary depending on the secretor blood group status of an individual woman as well as ethnicity and stage of lactation, with 2′FL levels from about 0.9 to above 4 g/L in mature milk among secretors [614]. Another predominant HMO in human milk is lacto-N-neotetraose (LNnT) at levels ranging from 0.1 to 0.6 g/L with higher levels within the first month of lactation [710, 1517].
Advancements in manufacturing technology now enable the synthesis of HMOs, and preclinical studies have established their safety for the purposes of supplementation of infant formulas [18, 19]. Safety, tolerance, and adequate growth as well as potential clinical benefits have been demonstrated in randomized controlled trials (RCTs) of term infant formulas supplemented with 2′FL alone and in combination with LNnT [2022]. An RCT in the United States of America (USA) found that infants receiving formula supplemented with either galacto-oligosaccharides (GOS) or GOS + 2′FL demonstrated adequate growth and good tolerance [21]. Another RCT conducted in Belgium and Italy examined a study formula containing 1.0 g/L 2′FL and 0.5 g/L of LNnT in the test arms, while the control arm received standard formula without HMOs [20]. The HMO-supplemented formula was again well-tolerated and supported age-appropriate growth. A third study in the USA compared tolerance in infants receiving a 100% whey, partially hydrolyzed infant formula with the probiotic Bifidobacterium lactis with and without the further addition of 2’FL and found that the HMOs-supplemented formula was well-tolerated [22].
Evidence is emerging that HMOs play an important role in the development of a balanced intestinal microbiota and in supporting immune protection in breastfed infants [2325]. Preclinical models have found that both 2’FL and LNnT promote the growth of Bifidobacterium species [26, 27]. Additionally, in a RCT of a term infant formula supplemented with 2’FL and LNnT, lower rates of parent-reported morbidity (particularly lower respiratory tract illnesses such as bronchitis) and lower use of antipyretics and antibiotics in the group receiving HMOs-supplemented formula were reported compared to the control infants [20]. Stool samples collected for microbiota assessment and metabolic signature at 3 months showed that the addition of 2’FL and LNnT shifted the stool microbiota closer to that observed in breastfed infants both in composition and function [28]. Collectively, these findings, in conjunction with the documented differences in HMOs composition between human and bovine milk, have provided a solid rationale for the benefits of bovine milk-based infant formulas with HMOs.
While the evidence provided to date in RCTs is supportive of the safety and tolerance of HMOs-supplemented infant formula, studies are needed in a real-world setting because results from a highly controlled RCT do not always translate outside of the trial setting [29]. Additionally, a relatively large proportion of infants in real-world settings are fed with both human milk and formula [3032], a mixed feeding regimen not studied in RCTs. The current study was thus designed to complement and enhance existing RCTs by assessing the growth, safety, and tolerance of healthy term infants, consuming an infant formula supplemented with HMOs either exclusively or mixed with human milk in a real-world setting.

Participants and methods

Study design

This was a three group, non-randomized, open-label, prospective study in healthy, term (37–42 weeks of gestation) infants enrolled at age 7 days to 2 months. The study was conducted between 08/07/2019 and 24 July 2020, in 12 centers (pediatrician and adolescent doctors) throughout Germany and Austria. One study group included infants who were exclusively formula fed (FF), while a second group included infants who were mixed fed, i.e., received both formula and human milk (MF). The third group included exclusively breastfed infants (BF) serving as a reference population. Formula-fed infants were eligible to participate if their parent(s) had independently elected, before study enrollment, to formula feed. Breastfed infants were eligible if the infants had been exclusively breastfed since birth, and their parent(s) had decided to continue exclusively breastfeeding until at least 4 months of age. Exclusion criteria included any known intolerance/allergy to cow’s milk (formula-fed group only), conditions requiring infant feedings other than those specified in the protocol, and evidence of significant systemic disorders (cardiac, respiratory, endocrinological, hematologic, gastrointestinal, or other).
At study enrollment, FF and MF infants received the study formula and were fed for 8 weeks (56 days). Formula was prepared and fed at home and was given ad libitum. Infants completed an in-person clinic visit at enrollment (baseline) and again at day 56 ± 3 days (week 8 visit). A telephone visit with the parents was also conducted on day 28 ± 3 days (week 4 visit).

Study product

Commercially available in Germany and Austria since autumn 2018, the study formula was provided to the participants at no charge. It was a partially hydrolyzed 100% whey, term infant formula with 67 kcal/100 mL consisting of 1.9-g protein, 11.5-g carbohydrates, and 5.1 g of lipids per 100 kcal powder, and with two HMOs: 1.0 g/L of 2′FL and 0.5 g/L of LNnT.

Ethical approval and informed consent

This study protocol was approved by the ethics committee of the Berlin Chamber of Physicians. Prior to the conduct of any screening tests, informed consent was obtained from each participant’s parent. Good clinical practice was followed by all sites throughout the study. The study was registered with ClinicalTrials.gov NCT05150288.

Study measures

At baseline and again at the clinic visit at week 8, anthropometry measures were obtained including weight, length, and head circumference using standardized procedures. Infant weight was measured without clothing or diaper on a calibrated electronic scale to the nearest 10 g. Recumbent length was measured on a pediatric length board to the nearest 1 mm. Head circumference was measured to the nearest 1 mm using a nonelastic plastic-coated measuring tape. Body mass index (BMI) was calculated as weight (kg)/(length (m))2. Z-scores for weight for age, length for age, head circumference for age, and BMI for age were calculated using the World Health Organization (WHO) Child Growth Standards [33].
The infant’s gastrointestinal (GI) symptom burden was assessed via the Infant Gastrointestinal Symptom Questionnaire (IGSQ) [34], a validated 13-item questionnaire that assesses GI-related signs and symptoms as observed by parents over the previous week in 5 domains: stooling, spitting up/vomiting, gassiness, crying, and fussing. Each item is scored on a scale of 1 to 5 with higher values indicating greater GI distress. A composite IGSQ score is derived from summing the individual scores with a possible range of 13 to 65 where higher values indicate greater GI distress and values ≤ 23 indicate no digestive distress [34]. The IGSQ was administered at baseline, week 4, and week 8.
A formula satisfaction questionnaire was administered to parents of infants in the formula-fed groups at week 4 and week 8 including three questions regarding the parent(s)’ experience with the study formula. Questions included the following: “Did your child like what he/she consumed?”, “How satisfied are you overall with the study product?”, and “Would you continue to provide the study formula to your child?”.
Adverse events (AE) were captured from the time of enrollment through the end of study. All AEs were assessed by the site investigator for duration, intensity, frequency, and relationship to study formula. AEs were classified by system, organ, and class (MedDRA SOC codes). In relation to published data of other studies, we expected AEs like the following: atopic diseases (f.e., eczema or cow’s milk protein allergy) infectious diseases; gastrointestinal symptoms, use of medication, and others.

Statistical methods

Demographics and other baseline characteristics were compared between all pairwise combinations of feeding groups using two-sided Wilcoxon rank-sum tests for continuous variables and Fisher’s exact tests for categorical variables. Fisher’s exact tests were computed from contingency tables. For tables larger than 2 × 2, a Monte Carlo estimation of the exact p-value was performed with 20,000 samples; otherwise, a direct exact p-value computation was performed. Missing values were excluded before performing the aforementioned tests.
The co-primary outcomes were growth and composite IGSQ score. Feeding group comparisons were assessed individually at each time point (baseline visit and week 8 visit) for all growth measures using the analysis of covariance (ANCOVA) controlling for baseline value, age, gender, and study center. Listwise deletion was performed to handle missing values in the models. Tolerance was assessed via the IGSQ scores. The 13 individual questions in the IGSQ as well as the five domain scores and the composite IGSQ score were tabulated for each feeding group at each time point (baseline visit, week 4 visit, and week 8 visit). These scores were compared between the feeding groups individually at each time point using ANCOVA controlling for the baseline scores and age at baseline. The derived inferential statistics on the IGSQ scores were based on the sandwich estimator of the variance–covariance matrix of the models’ parameters due to some heteroscedasticity observed in the models’ residuals. Listwise deletion was performed to handle missing values in the models.
All analyses were conducted using SAS BASE 9.4/SAS STAT 15.1 on the SAS Life Science Analytics Framework (SAS LSAF, SAS Institute Inc., Cary, NC, USA) version 5.2.2. Due to the descriptive nature of the trial, no adjustment for multiple testing was performed. The statistical significance was assessed using an α-level of 5%.
Being a real-world evidence (RWE) study, sample size was based on practical and logistical feasibility and on the experience of published RCTs investigating safety and tolerance of formula containing 2′FL/LNnt [2022]. The analysis set was defined by excluding infants who did not comply with the protocol (e.g., switched from breastfed group to formula-fed group and vice versa), were lost to follow-up, experienced tolerance issues, withdrew without explanations, or did not provide data due to other reasons. For BF, infants who were not exclusively breastfed and received other formula than the study product were excluded from the analysis set. All analyses of growth, tolerance, and satisfaction in this paper were conducted in the analysis set. AEs were reported for all enrolled infants.

Results

Subject disposition and demographics

In this study, 117 infants were enrolled including 51 FF, 22 MF, and 44 BF infants (Fig. 1). The number of subjects in the analysis set was 46, 22, and 38, respectively, in FF, MF, and BF, with primary exclusion reasons being major protocol deviations (namely, breastfed infants who were fed a non-study formula at least once during the study) and intolerance issues.
The demographics and baseline characteristics of the infants included in the analysis set are shown in Table 1. MF was slightly younger at enrollment as compared with FF and BF. The gender distribution was comparable between groups. There were no differences between the three groups in terms of mothers’ ethnicity or educational attainment. Mothers of the infants in FF were the youngest. Fathers in FF had a slightly lower level of education than those in MF and BF. Parents in FF had significantly higher proportions of smoking compared to those in BF.
Table 1
Demographics and baseline characteristics by feeding groups in the analysis set (N = 106)Subject characteristics
 
BF1
(N = 38)
MF1,2
(N = 22)
FF1,3,4
(N = 46)
p-value1
BF vs MF
p-value1
BF vs FF
p-value1
MF vs FF
Age at enrollment, days (median [IQR])
43 (30 − 49)
33 (29 − 39)
39 (32 − 49)
0.097
0.706
0.067
Gender
   
1.000
1.000
1.000
 Male
19 (50.0%)
11 (50.0%)
22 (47.8%)
-
-
-
 Female
19 (50.0%)
11 (50.0%)
24 (52.2%)
-
-
-
Ethnicity
   
1.000
0.202
0.324
 Caucasian
36 (94.7%)
21 (95.5%)
46 (100.0%)
-
-
-
 Other
2 (5.3%)
1 (4.6%)
0 (0.0%)
-
-
-
Days breastfed since birth (median [IQR])
43 (30 − 49)
33 (29 − 39)
0 (0 − 5)
0.100
< 0.001
< 0.001
Days formula-fed since birth (median [IQR])
0 (0 − 0)
30 (16 − 32)
38 (30 − 49)
< 0.001
< 0.001
0.007
Mother’s age, days (median [IQR])
33 (30 − 36)
32 (29 − 35)
29 (26 − 34)
0.299
0.004
0.150
Mother’s highest level of education
   
0.600
0.202
0.194
 Primary school
0 (0.0%)
0 (0.0%)
5 (10.9%)
-
-
-
 High school
9 (23.7%)
5 (22.8%)
10 (21.7%)
-
-
-
 College or above
18 (47.4%)
13 (59.1%)
17 (37.0%)
-
-
-
 Professional school
11 (29.0%)
4 (18.2%)
14 (30.4%)
-
-
-
Mother smoked during pregnancy
1 (2.6%)
2 (9.1%)
10 (21.7%)
0.548
0.010
0.311
Father’s age, days (median [IQR])
34 (31 − 37)
36 (33 − 39)
31 (29–36)
0.160
0.040
0.006
Father’s highest level of education
   
0.664
0.012
0.043
 Primary school
0 (0.0%)
0 (0.0%)
5 (10.9%)
-
-
-
 High school
6 (15.8%)
4 (18.2%)
7 (15.2%)
-
-
-
 College or above
18 (47.4%)
12 (54.6%)
10 (21.7%)
-
-
-
 Professional school
14 (36.8%)
5 (22.8%)
17 (37.0%)
-
-
-
 Missing, unknown, or less than primary school
0 (0.0%)
1 (4.6%)
7 (15.2%)
-
-
-
Father is a smoker
8 (21.1%)
9 (40.9%)
25 (54.4%)
0.132
0.002
0.430
Data shows as N and % unless otherwise noted. Percentages may not exactly add up to 100% due to rounding
BF exclusively breastfed, MF mixed fed, FF formula fed (exclusively), IQR interquartile range (Q1–Q3), SD standard deviation
1For continuous variables, p-values come from two-sided Wilcoxon rank-sum tests. For categorical variables, p-values correspond to Fisher’s exact tests
2Compared to BF, MF has been formula-fed longer (p < 0.001). There are no other significant differences between the two groups
3Compared to BF, FF has not been breastfed at all (p < 0.001) and formula-fed longer (p < 0.001). The mothers and the fathers in FF are younger (p = 0.004 and p = 0.040, respectively), and a greater number of mothers and fathers smoked more during pregnancy (p = 0.010 and p = 0.002). Fathers are also less educated in FF compared to MF (p = 0.012)
4Compared to MF, FF has not been breastfed at all (p < 0.001) and formula-fed longer (p = 0.007). The fathers in FF are younger (p = 0.006) and less educated (p = 0.043) compared to MF

Growth

Age-appropriate growth was observed in all three feeding groups. Baseline weight and length were slightly lower in MF. By week 8, there were no significant differences between any feeding groups for any of the anthropometric measures (all ANCOVA p-values between feeding groups > 0.05). Mean Z-scores for weight, length, head circumference, and BMI at baseline and week 8 are shown in Fig. 2. Weight-for-age, length-for-age, and BMI-for-age z-scores were comparable between all feeding groups at week 8. The mean z-scores were within ± 0.5 of the WHO medians at week 8. Head circumference-for-age z-scores were also comparable between groups and tracked closely with the WHO standards.

Gastrointestinal tolerance

Table 2 shows descriptive characteristics for the five IGSQ domains and the overall composite IGSQ score. Composite IGSQ scores demonstrated low GI distress in all feeding groups at all time points. At baseline, FF had significantly greater GI distress compared to BF (mean difference [95% confidence interval (CI)] FF-BF = 5.18 [2.44, 7.91], p = 0.0003). However, there were no significant differences in the composite IGSQ score between any of the feeding groups at week 4 or week 8 suggesting that the GI tolerance in FF improved after introduction of study formula. For one of the five domains of the IGSQ (gassiness), there were no significant differences in scores between the groups at baseline, week 4, or week 8. There was a minor difference in fussiness between FF and BF at week 4 (mean difference [95% CI] = 0.821 [0.089, 1.554], p = 0.028), but no differences were observed at baseline or week 8. For the stooling domain, FF had significantly higher scores (i.e., more stooling issues) than BF at baseline (mean difference [95% CI] = 1.335 [0.543, 2.126], p = 0.001) but showed significant improvement by week 8 (mean difference [95% CI] =  − 0.151 [− 0.704, 0.403], p = 0.59), with FF moving closer to the stooling profile of BF. For the spitting-up/vomiting domain, FF again had significantly greater distress compared with BF at baseline (mean difference [95% CI] = 1.476 [0.457, 2.469], p = 0.005) and showed significant improvement at week 8 (mean difference [95% CI] =  − 0.128 [− 0.865, 0.609], p = 0.731), moving closer to the spitting-up/vomiting profile of BF. Lastly, FF had significantly greater distress for the crying domain compared with BF at baseline (mean difference [95% CI] = 0.951 [0.189, 1.713], p = 0.015) and week 4 (mean difference [95% CI] = 0.639 [0.047, 1.232], p = 0.035) and showed significant improvement at week 8 (mean difference [95% CI] =  − 0.339 [− 0.877, − 0.199], p = 0.214), becoming more comparable to BF.
Table 2
Composite IGSQ and domain scoresa at baseline, week 4, and week 8, by feeding group, analysis set (N = 106)
 
BF
MF
 
FF
 
N
Mean
SD
Min–Max
N
Mean
SD
Min–Max
p-value2
N
Mean
SD
Min–Max
p-value2
Composite IGSQ score
 
Baseline
38
20.33
4.30
13–32
22
21.82
5.27
13–37
0.279
46
25.50
8.31
15–50
< 0.001
Week 4
38
19.20
4.05
13–29
22
20.45
4.73
13–32
0.563
43
21.94
5.64
13–37
0.127
Week 8
38
18.76
3.97
14–32
22
18.14
3.67
13–25
0.298
45
19.08
5.49
13–36
0.548
IGSQ domains
Stooling
Baseline
38
2.68
1.21
2–6
22
2.95
1.36
2–6
0.769
45
4.04
2.45
2–10
0.001
Week 4
37
2.54
0.99
2–6
22
3.00
1.41
2–6
0.364
43
3.05
1.66
2–8
0.549
Week 8
38
2.66
1.02
2–6
22
2.73
1.39
2–7
0.994
45
2.58
1.20
2–6
0.590
Spitting up/vomiting
Baseline
37
5.68
1.70
4–9
21
6.38
2.64
4–12
0.293
45
7.16
2.97
4–16
0.005
Week 4
38
5.84
2.05
4–14
22
5.91
1.85
4–11
0.758
42
6.12
2.25
4–12
0.949
Week 8
38
5.53
1.37
4–10
22
5.09
1.63
4–11
0.183
44
5.77
2.25
4–12
0.731
Crying
Baseline
38
4.16
1.22
3–7
22
4.32
1.46
3–8
0.659
46
5.11
2.28
3–13
0.015
Week 4
38
4.00
0.96
3–6
22
4.09
1.48
3–8
0.802
43
4.91
1.99
3–10
0.035
Week 8
38
4.37
1.20
3–8
22
3.95
1.33
3–7
0.143
45
4.24
1.65
3–11
0.214
Fussiness
Baseline
38
2.66
1.70
2–10
22
2.59
1.18
2–6
0.931
45
3.31
2.00
2–10
0.095
Week 4
38
2.45
0.89
2–6
22
2.59
1.26
2–7
0.522
43
3.40
2.27
2–10
0.028
Week 8
38
2.37
1.36
2–10
22
2.32
0.78
2–5
0.928
45
2.71
1.66
2–10
0.420
Gassiness
Baseline
38
5.11
1.75
2–9
22
5.45
2.18
2–9
0.387
45
5.82
2.06
2–10
0.078
Week 4
38
4.37
1.55
2–7
22
4.86
1.58
2–8
0.405
43
4.44
1.76
2–8
0.613
Week 8
38
3.84
1.75
2–8
22
4.05
1.65
2–8
0.976
45
3.80
1.80
2–7
0.452
BF exclusively breastfed group, MF mixed fed group, FF formula fed group (exclusively), SD standard deviation
2p-value compared to BF
aThe IGSQ consists of 13 individual questions (grouped into five domains). Each question is scored on a scale of 1 to 5 with higher values indicating greater GI distress. The composite IGSQ score is derived by summing the individual scores, resulting in a possible range of 13 to 65 where higher values indicate greater GI distress. A composite IGSQ ≤ 23 indicates no digestive distress
For the individual items in the 13-item IGSQ, FF passed more hard stools than BF and MF and had more difficulties in passing bowel movements than BF at baseline. Still at baseline, FF compared to BF showed a higher number of times the baby arched its back in pain when spitting up, was crying during feeding, or it was not possible to stop the baby from crying and also a higher number of fussy days in the past week. In FF, total time spent crying and number of times unable to soothe baby’s fussiness were more than in MF. There were no significant differences between the groups for the other IGSQ individual items at baseline. At week 8, there were no significant differences in any of the individual items between BF, MF, and FF except the total time the baby spent crying. FF cried less than BF (mean difference [95% CI] =  − 0.303 [− 0.614, 0.007], p = 0.056) at week 8, but there were no other differences for the other crying items. Total crying time did significantly improve in FF between baseline and 8 weeks (mean difference [95% CI] =  − 0.444 [− 0.799, − 0.090], p = 0.015), while there was no change between baseline and 8 weeks for BF and MF.

Formula satisfaction

Most parents in MF and FF reported that their child liked what he/she consumed, and that they would continue to provide the study formula to their child at both weeks 4 and 8 (Table 3).
Table 3
Formula satisfaction questionnaire results [N (%)], at weeks 4 and 8 among parents of infants receiving study formula, by feeding group, analysis set (N = 68 infants receiving formula)
 
MF
FF
N = 22
N = 22
N = 43
N = 46
Week 4
Week 8
Week 4
Week 8
How satisfied are you overall with the study formula?
 Satisfied
19 (86.4%)
17 (77.3%)
34 (79.1%)
39 (84.8%)
 Neutral
2 (9.1%)
4 (18.2%)
7 (16.3%)
5 (10.9%)
 Dissatisfied
0 (0.0%)
0 (0.0%)
2 (4.7%)
0 (0.0%)
 Missing
1 (4.6%)
1 (4.6%)
0 (0.0%)
2 (4.4%)
Would you continue to provide the study formula to your child?
 Yes
21 (95.5%)
21 (95.5%)
37 (86.1%)
38 (82.6%)
 No
0 (0.0%)
0 (0.0%)
6 (14.0%)
6 (13.0%)
 Missing
1 (4.6%)
1 (4.6%)
0 (0.0%)
2 (4.4%)
Did your child like what he/she consumed?a
 Yes
21 (95.5%)
21 (95.5%)
39 (90.7%)
42 (91.3%)
 No
0 (0.0%)
0 (0.0%)
4 (9.3%)
2 (4.4%)
 Missing
1 (4.6%)
1 (4.6%)
0 (0.0%)
2 (4.4%)
Percentages may not exactly add up to 100% due to rounding
MF mixed-fed group, FF formula-fed group (exclusively)
aThe question “Did your child like what he/she consumed?” was based on the parent’s observation when feeding their child (overall reaction, eagerness to consume the formula, refusal)

Adverse events

A total of 46 subjects experienced 69 AEs during the course of the study, and no serious AEs were reported (Table 4). A total of 7.8% (n = 4) of the AEs were potentially formula related and were only reported in the FF Group.
Table 4
Overview of adverse events, by feeding group, in the safety data set (n = 117)
 
BF (N = 44)
MF (N = 22)
FF (N = 51)
Occurrence
11 (25.0%) [18]
6 (27.3%) [10]
29 (56.9%) [41]
Severity
 Mildb
11 (25.0%) [18]
6 (27.3%) [9]
22 (43.1%) [32]
 Moderatec
0 (0.0%) [0]
1 (4.6%) [1]
8 (15.7%) [9]
Caused study discontinuation
 Yes
0 (0.0%) [0]
0 (0.0%) [0]
4 (7.8%) [4]a
 No
11 (25.0%) [18]
6 (27.3%) [10]
27 (52.3%) [37]
Data shown as N (%) [m]
N number of subjects with at least one AE, m total number of adverse events, BF exclusively breastfed group, MF mixed-fed group, FF formula-fed group (exclusively)
aThese four infants in FF experienced potentially product-related AEs including instances of lactose intolerance, hard feces, vomiting, and diarrhea
bMild AEs, negative clinical sign/situation which requires no intervention
cModerate AEs, negative clinical sign/situation which requires an intervention but without relevant or long-lasting limitations for the subject

Discussion

The results indicate that formula-fed infants, either exclusively or mixed fed, receiving the formula supplemented with 2′FL and LNnT, had age-appropriate growth in line with the WHO standards and comparable to BF infants. Growth was also comparable to that seen in previous studies with West and South European infant populations [35]. By week 8, GI tolerance as indicated by low IGSQ scores was comparable in the formula-fed infants with that in BF infants indicating the formula is well tolerated. The incidence of adverse events in all groups was low. As shown at Table 4, 7.8% (n = 4) of the AEs were potentially formula related. Despite the season of the year (fall-winter), cases of bronchitis were lower than expected from the literature [36]. Therefore, the results were not added as secondary outcomes.
The results of this RWE study are comparable to those from previous RCTs that have examined anthropometry and GI tolerance for term infant formulas supplemented with HMOs. One RCT was a multicenter, double-blind trial that enrolled 175 healthy term infants in Italy and Belgium at less than 14 days of age who were fed study formula for 6 months [20]. The HMO-supplemented formula (2′FL + LNnT) demonstrated age-appropriate growth as well as good tolerance as measured by parents. Another RCT included 189 term infants in the USA who were exclusively formula fed until 4 months of age [21]. Formulas with 2′FL (at two different dosages) and GOS were well-tolerated based on parental reports, and no significant differences were observed for growth compared to a control group. Notably, neither of those trials utilized a validated tool to assess tolerance, and thus, tolerance outcomes cannot easily be compared across studies. A recent RCT used the same validated IGSQ tool as in the current study to assess tolerance [22]. The HMO-supplemented formula was well tolerated as evidenced by similar IGSQ scores at week 6 between the groups with (mean [SD] = 20.9 [4.8]) and without (mean [SD] = 20.7 [4.3]) the addition of 2′FL. These scores are similar to those observed in FF in the current study at week 8 (mean [SD] = 19.1[4.5]). Additionally, a single-arm study of a formula supplemented with 2′FL fed to fussy infants showed significant improvement in IGSQ scores after 3 weeks of feeding (baseline mean [SD] = 34.1 [10.0]; week 3 mean [SD] = 21.4 [7.0]; p < 0.001) [37]. Although we did not limit our study to fussy infants, we also saw an improvement in IGSQ in FF in our study from baseline to week 8 (mean difference [95% confidence interval] =  − 6.639 [− 9.497, − 3.782], p < 0.0001). The improvement in GI tolerance in our study might be partially related to the natural evolvement of GI tolerance which improves with increasing age but could potentially also be attributed to the composition of the study formula including the two HMOs. Almost all infants switched to the study formula at the beginning of the study, i.e., they were receiving a different formula prior to enrollment (44 of 46 FF infants and 21 of 22 MF infants), suggesting that the HMO-containing study formula has better GI tolerance than the formulas without HMOs consumed prior to enrollment. Only one other real-world study has been conducted to our knowledge; a study with similar design to the current study was conducted in Spain and had very similar results for both growth and IGSQ scores [38]. The agreements between the previous RCTs, the Spanish real-world study, and the current real-world study are reassuring that growth, safety, and tolerance of HMO-supplemented formula are consistent and robust across different geographical populations.
This study has several strengths. First, GI burden was measured using a validated instrument, the 13-item IGSQ based on five separate domains of feeding tolerance. The use of a validated instrument provides information that is interpretable and meaningful to practicing clinicians. Second, this was an RWE study, a design distinct from the RCT, simpler, less restrictive, but still in line with current clinical practices, enhancing the generalizability of the results and providing complementary data to RCTs. The published prevalence of infants who are mixed fed indicates that at age 1 month, 30% of infants receive mixed feedings [30, 31], similar to that observed in this study. Thus, the demonstration of appropriate growth and good tolerance in the mixed feeding group of infants in this study provides important evidence not found in the RCTs conducted to date. Some limitations of the study should also be noted. An open-label, non-randomized design increases the risk for bias, in particular for response bias (even for validated questionnaires), and higher attrition rates and missing data. In a study with specifically defined feeding regimens such as ours, randomization is however not possible. The main aim of randomization is to have study groups with comparable characteristics. We therefore compared the baseline characteristics in our three groups, and there are no substantial differences except for infant age at enrollment and parents’ smoking status. Infant age at enrollment was slightly lower in MF compared with both BF and FF. As it may take longer for mothers to establish exclusive breastfeeding or formula-fed patterns, the younger age in MF at enrollment can be expected. To account for the slight difference in baseline age between the groups, we included baseline age in the statistical models to reduce potential risk of bias arising by the non-randomized nature of our trial. Smoking was higher among the parents in FF compared with BF which for the fathers could be linked to the slightly lower education level in FF compared with BF. Alternately, the parents in BF may have underreported their smoking habits to make a positive impression, a form of social-desirability bias. The study formula was supplemented with just a single level of 2′FL and LNnT, and thus, this study cannot assess whether the observed growth and tolerance effects might differ over a wider range of levels of these HMOs. Additionally, this study, while multicenter, took place within only two countries (Germany and Austria), and its results may not be generalizable outside of Northwest Europe. Furthermore, the authors want to point out that even if supplemented with HMOs, infant formula is not comparable with the gold standard “human breast milk” concerning multiple aspects. The aim of research in infant formula is of cause not to replace human breast milk — but to have the best possible substitute available in case breastfeeding fails or breast milk is not available.
In conclusion, this is one of the first studies to use real-world evidence to examine the supplementation of infant formula with HMOs. The results obtained were similar to those found in more tightly controlled RCTs, indicating robust effects for growth, safety, and tolerance in association with HMO-supplemented infant formulas.

Declarations

The ethical approval concerning the study protocol, the data handling, the publication of the data, the conduct of the study, and all additional study aspects was given by the ethics committee of the Ärztekammer Berlin (general medical council of the state Berlin, Germany) under the sign: 18.06.INF. Inclusion of patients in the study was only performed after informed consent of the parents. The study was registered at ClinicalTrials.gov under the study number NCT05150288.

Competing interests

-LL and SD are current employees of Société des Produits Nestlé S.A., Vevey, Switzerland.
-This study was supported by Société des Produits Nestlé S.A.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Ninonuevo MR, Lebrilla CB (2009) Mass spectrometric methods for analysis of oligosaccharides in human milk. Nutr Rev 67(Suppl 2):S216-226CrossRefPubMed Ninonuevo MR, Lebrilla CB (2009) Mass spectrometric methods for analysis of oligosaccharides in human milk. Nutr Rev 67(Suppl 2):S216-226CrossRefPubMed
2.
Zurück zum Zitat Gabrielli O, Zampini L, Galeazzi T et al (2011) Preterm milk oligosaccharides during the first month of lactation. Pediatrics 128(6):e1520-1531CrossRefPubMed Gabrielli O, Zampini L, Galeazzi T et al (2011) Preterm milk oligosaccharides during the first month of lactation. Pediatrics 128(6):e1520-1531CrossRefPubMed
3.
Zurück zum Zitat Zivkovic AM, German JB, Lebrilla CB, Mills DA (2011) Human milk glycobiome and its impact on the infant gastrointestinal microbiota. Proc Natl Acad Sci U S A 108(Suppl 1):4653–4658CrossRefPubMed Zivkovic AM, German JB, Lebrilla CB, Mills DA (2011) Human milk glycobiome and its impact on the infant gastrointestinal microbiota. Proc Natl Acad Sci U S A 108(Suppl 1):4653–4658CrossRefPubMed
4.
Zurück zum Zitat Hale TWaPEH (2007) Textbook of Human Lactation Hale TWaPEH (2007) Textbook of Human Lactation
5.
Zurück zum Zitat Urashima TMM, Oftedal OT (2016) Oligosaccharides in the milk of other mammals. In: McGuire MK, McGuire MA, Bode L (eds) Prebiotics and probiotics in human milk: origins and functions of milk-borne oligosaccharides and bacteria. pp 45–139 Urashima TMM, Oftedal OT (2016) Oligosaccharides in the milk of other mammals. In: McGuire MK, McGuire MA, Bode L (eds) Prebiotics and probiotics in human milk: origins and functions of milk-borne oligosaccharides and bacteria. pp 45–139
6.
Zurück zum Zitat Coppa GV, Pierani P, Zampini L, Carloni I, Carlucci A, Gabrielli O (1999) Oligosaccharides in human milk during different phases of lactation. Acta Paediatr Suppl 88(430):89–94CrossRefPubMed Coppa GV, Pierani P, Zampini L, Carloni I, Carlucci A, Gabrielli O (1999) Oligosaccharides in human milk during different phases of lactation. Acta Paediatr Suppl 88(430):89–94CrossRefPubMed
7.
Zurück zum Zitat Bao Y, Chen C, Newburg DS (2013) Quantification of neutral human milk oligosaccharides by graphitic carbon high-performance liquid chromatography with tandem mass spectrometry. Anal Biochem 433(1):28–35CrossRefPubMed Bao Y, Chen C, Newburg DS (2013) Quantification of neutral human milk oligosaccharides by graphitic carbon high-performance liquid chromatography with tandem mass spectrometry. Anal Biochem 433(1):28–35CrossRefPubMed
8.
Zurück zum Zitat Thurl S, Munzert M, Henker J et al (2010) Variation of human milk oligosaccharides in relation to milk groups and lactational periods. Br J Nutr 104(9):1261–1271CrossRefPubMed Thurl S, Munzert M, Henker J et al (2010) Variation of human milk oligosaccharides in relation to milk groups and lactational periods. Br J Nutr 104(9):1261–1271CrossRefPubMed
9.
Zurück zum Zitat Chaturvedi P, Warren CD, Altaye M et al (2001) Fucosylated human milk oligosaccharides vary between individuals and over the course of lactation. Glycobiology 11(5):365–372CrossRefPubMed Chaturvedi P, Warren CD, Altaye M et al (2001) Fucosylated human milk oligosaccharides vary between individuals and over the course of lactation. Glycobiology 11(5):365–372CrossRefPubMed
10.
Zurück zum Zitat Erney RM, Malone WT, Skelding MB et al (2000) Variability of human milk neutral oligosaccharides in a diverse population. J Pediatr Gastroenterol Nutr 30(2):181–192CrossRefPubMed Erney RM, Malone WT, Skelding MB et al (2000) Variability of human milk neutral oligosaccharides in a diverse population. J Pediatr Gastroenterol Nutr 30(2):181–192CrossRefPubMed
11.
Zurück zum Zitat Coppa GV, Gabrielli O, Zampini L et al (2011) Oligosaccharides in 4 different milk groups, Bifidobacteria, and Ruminococcus obeum. J Pediatr Gastroenterol Nutr 53(1):80–87CrossRefPubMed Coppa GV, Gabrielli O, Zampini L et al (2011) Oligosaccharides in 4 different milk groups, Bifidobacteria, and Ruminococcus obeum. J Pediatr Gastroenterol Nutr 53(1):80–87CrossRefPubMed
12.
Zurück zum Zitat Smilowitz JT, O’Sullivan A, Barile D, German JB, Lonnerdal B, Slupsky CM (2013) The human milk metabolome reveals diverse oligosaccharide profiles. J Nutr 143(11):1709–1718 Smilowitz JT, O’Sullivan A, Barile D, German JB, Lonnerdal B, Slupsky CM (2013) The human milk metabolome reveals diverse oligosaccharide profiles. J Nutr 143(11):1709–1718
13.
Zurück zum Zitat Nakhla T, Fu D, Zopf D, Brodsky NL, Hurt H (1999) Neutral oligosaccharide content of preterm human milk. Br J Nutr 82(5):361–367CrossRefPubMed Nakhla T, Fu D, Zopf D, Brodsky NL, Hurt H (1999) Neutral oligosaccharide content of preterm human milk. Br J Nutr 82(5):361–367CrossRefPubMed
14.
Zurück zum Zitat McGuire MK, Meehan CL, McGuire MA et al (2017) What’s normal? Oligosaccharide concentrations and profiles in milk produced by healthy women vary geographically. Am J Clin Nutr 105(5):1086–1100 McGuire MK, Meehan CL, McGuire MA et al (2017) What’s normal? Oligosaccharide concentrations and profiles in milk produced by healthy women vary geographically. Am J Clin Nutr 105(5):1086–1100
15.
Zurück zum Zitat Sumiyoshi W, Urashima T, Nakamura T et al (2003) Determination of each neutral oligosaccharide in the milk of Japanese women during the course of lactation. Br J Nutr 89(1):61–69CrossRefPubMed Sumiyoshi W, Urashima T, Nakamura T et al (2003) Determination of each neutral oligosaccharide in the milk of Japanese women during the course of lactation. Br J Nutr 89(1):61–69CrossRefPubMed
16.
Zurück zum Zitat Leo F, Asakuma S, Fukuda K, Senda A, Urashima T (2010) Determination of sialyl and neutral oligosaccharide levels in transition and mature milks of Samoan women, using anthranilic derivatization followed by reverse phase high performance liquid chromatography. Biosci Biotechnol Biochem 74(2):298–303CrossRefPubMed Leo F, Asakuma S, Fukuda K, Senda A, Urashima T (2010) Determination of sialyl and neutral oligosaccharide levels in transition and mature milks of Samoan women, using anthranilic derivatization followed by reverse phase high performance liquid chromatography. Biosci Biotechnol Biochem 74(2):298–303CrossRefPubMed
17.
Zurück zum Zitat Asakuma S, Urashima T, Akahori M et al (2008) Variation of major neutral oligosaccharides levels in human colostrum. Eur J Clin Nutr 62(4):488–494CrossRefPubMed Asakuma S, Urashima T, Akahori M et al (2008) Variation of major neutral oligosaccharides levels in human colostrum. Eur J Clin Nutr 62(4):488–494CrossRefPubMed
18.
Zurück zum Zitat Coulet M, Phothirath P, Allais L, Schilter B (2014) Pre-clinical safety evaluation of the synthetic human milk, nature-identical, oligosaccharide 2’-O-fucosyllactose (2’FL). Regul Toxicol Pharmacol 68(1):59–69 Coulet M, Phothirath P, Allais L, Schilter B (2014) Pre-clinical safety evaluation of the synthetic human milk, nature-identical, oligosaccharide 2’-O-fucosyllactose (2’FL). Regul Toxicol Pharmacol 68(1):59–69
19.
Zurück zum Zitat Coulet M, Phothirath P, Constable A, Marsden E, Schilter B (2013) Pre-clinical safety assessment of the synthetic human milk, nature-identical, oligosaccharide lacto-N-neotetraose (LNnT). Food Chem Toxicol 62:528–537CrossRefPubMed Coulet M, Phothirath P, Constable A, Marsden E, Schilter B (2013) Pre-clinical safety assessment of the synthetic human milk, nature-identical, oligosaccharide lacto-N-neotetraose (LNnT). Food Chem Toxicol 62:528–537CrossRefPubMed
20.
Zurück zum Zitat Puccio G, Alliet P, Cajozzo C et al (2017) Effects of infant formula with human milk oligosaccharides on growth and morbidity: a randomized multicenter trial. J Pediatr Gastroenterol Nutr 64(4):624–631CrossRefPubMedPubMedCentral Puccio G, Alliet P, Cajozzo C et al (2017) Effects of infant formula with human milk oligosaccharides on growth and morbidity: a randomized multicenter trial. J Pediatr Gastroenterol Nutr 64(4):624–631CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Marriage BJ, Buck RH, Goehring KC, Oliver JS, Williams JA (2015) Infants fed a lower calorie formula with 2’FL show growth and 2’FL uptake like breast-fed infants. J Pediatr Gastroenterol Nutr 61(6):649–658 Marriage BJ, Buck RH, Goehring KC, Oliver JS, Williams JA (2015) Infants fed a lower calorie formula with 2’FL show growth and 2’FL uptake like breast-fed infants. J Pediatr Gastroenterol Nutr 61(6):649–658
22.
Zurück zum Zitat Storm HM, Shepard J, Czerkies LM et al (2019) 2’-Fucosyllactose is well tolerated in a 100% whey, partially hydrolyzed infant formula with Bifidobacterium lactis: a randomized controlled trial. Glob Pediatr Health 6:2333794X19833995 Storm HM, Shepard J, Czerkies LM et al (2019) 2’-Fucosyllactose is well tolerated in a 100% whey, partially hydrolyzed infant formula with Bifidobacterium lactis: a randomized controlled trial. Glob Pediatr Health 6:2333794X19833995
23.
Zurück zum Zitat Kulinich A, Liu L (2016) Human milk oligosaccharides: the role in the fine-tuning of innate immune responses. Carbohydr Res 432:62–70CrossRefPubMed Kulinich A, Liu L (2016) Human milk oligosaccharides: the role in the fine-tuning of innate immune responses. Carbohydr Res 432:62–70CrossRefPubMed
24.
Zurück zum Zitat Lewis ZT, Totten SM, Smilowitz JT et al (2015) Maternal fucosyltransferase 2 status affects the gut bifidobacterial communities of breastfed infants. Microbiome 3:13CrossRefPubMedPubMedCentral Lewis ZT, Totten SM, Smilowitz JT et al (2015) Maternal fucosyltransferase 2 status affects the gut bifidobacterial communities of breastfed infants. Microbiome 3:13CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Bode L (2015) The functional biology of human milk oligosaccharides. Early Hum Dev 91(11):619–622CrossRefPubMed Bode L (2015) The functional biology of human milk oligosaccharides. Early Hum Dev 91(11):619–622CrossRefPubMed
26.
Zurück zum Zitat Yu ZT, Chen C, Newburg DS (2013) Utilization of major fucosylated and sialylated human milk oligosaccharides by isolated human gut microbes. Glycobiology 23(11):1281–1292CrossRefPubMedPubMedCentral Yu ZT, Chen C, Newburg DS (2013) Utilization of major fucosylated and sialylated human milk oligosaccharides by isolated human gut microbes. Glycobiology 23(11):1281–1292CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Marcobal A, Barboza M, Sonnenburg ED et al (2011) Bacteroides in the infant gut consume milk oligosaccharides via mucus-utilization pathways. Cell Host Microbe 10(5):507–514CrossRefPubMedPubMedCentral Marcobal A, Barboza M, Sonnenburg ED et al (2011) Bacteroides in the infant gut consume milk oligosaccharides via mucus-utilization pathways. Cell Host Microbe 10(5):507–514CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Alliet P, Puccio G, Janssens E et al (2016) Term infant formula supplemented with human milk oligosaccharides (2′-fucosyllactose and lacto-N-neotetraose) shifts stool microbiota and metabolic signatures closer to that of breastfed infants. J Pediatr Gastroenterol Nutr 63:S55CrossRef Alliet P, Puccio G, Janssens E et al (2016) Term infant formula supplemented with human milk oligosaccharides (2′-fucosyllactose and lacto-N-neotetraose) shifts stool microbiota and metabolic signatures closer to that of breastfed infants. J Pediatr Gastroenterol Nutr 63:S55CrossRef
30.
Zurück zum Zitat Cuadrón Andrés L, Samper Villagrasa MP, Álvarez Sauras ML, Lasarte Velillas JJ, Rodríguez Martínez G (2013) Breastfeeding prevalence during the first year of life in Aragon. CALINA study. An Pediatr (Barc). 79(5):312–318CrossRefPubMed Cuadrón Andrés L, Samper Villagrasa MP, Álvarez Sauras ML, Lasarte Velillas JJ, Rodríguez Martínez G (2013) Breastfeeding prevalence during the first year of life in Aragon. CALINA study. An Pediatr (Barc). 79(5):312–318CrossRefPubMed
31.
Zurück zum Zitat Karmaus W, Soto-Ramirez N, Zhang H (2017) Infant feeding pattern in the first six months of age in USA: a follow-up study. Int Breastfeed J 12:48CrossRefPubMedPubMedCentral Karmaus W, Soto-Ramirez N, Zhang H (2017) Infant feeding pattern in the first six months of age in USA: a follow-up study. Int Breastfeed J 12:48CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Zhang J, Himes JH, Guo Y et al (2013) Birth weight, growth and feeding pattern in early infancy predict overweight/obesity status at two years of age: a birth cohort study of Chinese infants. PLoS ONE 8(6):e64542CrossRefPubMedPubMedCentral Zhang J, Himes JH, Guo Y et al (2013) Birth weight, growth and feeding pattern in early infancy predict overweight/obesity status at two years of age: a birth cohort study of Chinese infants. PLoS ONE 8(6):e64542CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat WHO Multicentre Growth Reference Study Group (2006) WHO Child Growth Standards based on length/height, weight and age. Acta Paediatr Suppl 450:76–85CrossRef WHO Multicentre Growth Reference Study Group (2006) WHO Child Growth Standards based on length/height, weight and age. Acta Paediatr Suppl 450:76–85CrossRef
34.
Zurück zum Zitat Riley AW, Trabulsi J, Yao M, Bevans KB, DeRusso PA (2015) Validation of a Parent Report Questionnaire: the Infant Gastrointestinal Symptom Questionnaire. Clin Pediatr (Phila) 54(12):1167–1174CrossRefPubMed Riley AW, Trabulsi J, Yao M, Bevans KB, DeRusso PA (2015) Validation of a Parent Report Questionnaire: the Infant Gastrointestinal Symptom Questionnaire. Clin Pediatr (Phila) 54(12):1167–1174CrossRefPubMed
35.
Zurück zum Zitat Alexander DD, Yan J, Bylsma LC et al (2016) Growth of infants consuming whey-predominant term infant formulas with a protein content of 1.8 g/100 kcal: a multicenter pooled analysis of individual participant data. Am J Clin Nutr. 104(4):1083–1092CrossRefPubMed Alexander DD, Yan J, Bylsma LC et al (2016) Growth of infants consuming whey-predominant term infant formulas with a protein content of 1.8 g/100 kcal: a multicenter pooled analysis of individual participant data. Am J Clin Nutr. 104(4):1083–1092CrossRefPubMed
37.
Zurück zum Zitat Czerkies LA, Kineman B, Reichert H, Cohen SS, Carvalho RS (2019) Use of a partially hydrolyzed 100% whey-based infant formula with Lactobacillus reuteri in infants with caregiver-perceived intolerance. J Acad Nutr Diet 119:19–26CrossRef Czerkies LA, Kineman B, Reichert H, Cohen SS, Carvalho RS (2019) Use of a partially hydrolyzed 100% whey-based infant formula with Lactobacillus reuteri in infants with caregiver-perceived intolerance. J Acad Nutr Diet 119:19–26CrossRef
38.
Zurück zum Zitat Román E, Moreno Villares JM, Domínguez Ortega F et al (2020) Real-world study in infants fed with an infant formula with two human milk oligosaccharides. Nutr Hosp 37(4):698–706PubMed Román E, Moreno Villares JM, Domínguez Ortega F et al (2020) Real-world study in infants fed with an infant formula with two human milk oligosaccharides. Nutr Hosp 37(4):698–706PubMed
Metadaten
Titel
Real-world evidence study on tolerance and growth in infants fed an infant formula with two human milk oligosaccharides vs mixed fed and exclusively breastfed infants
verfasst von
Frank Jochum
Martina Meyer-Krott
Tina Hübler
Maja Lorenz
Raffi Bedikian
Joseph Zakarian
Anja Litzka
Guido Judex
Holger Hertzberg
Daniela Klee
Lothar Maurer
Martin Schacht
Adnan Al-Radhi
Jan Maier
Alexander Kröckel
Christian Faustmann
Luca Lavalle
Samir Dahbane
Publikationsdatum
01.12.2023
Verlag
Springer International Publishing
Erschienen in
Molecular and Cellular Pediatrics / Ausgabe 1/2023
Elektronische ISSN: 2194-7791
DOI
https://doi.org/10.1186/s40348-023-00162-6

Weitere Artikel der Ausgabe 1/2023

Molecular and Cellular Pediatrics 1/2023 Zur Ausgabe

„Übersichtlicher Wegweiser“: Lauterbachs umstrittener Klinik-Atlas ist online

17.05.2024 Klinik aktuell Nachrichten

Sie sei „ethisch geboten“, meint Gesundheitsminister Karl Lauterbach: mehr Transparenz über die Qualität von Klinikbehandlungen. Um sie abzubilden, lässt er gegen den Widerstand vieler Länder einen virtuellen Klinik-Atlas freischalten.

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.

Erstmanifestation eines Diabetes-Typ-1 bei Kindern: Ein Notfall!

16.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Manifestiert sich ein Typ-1-Diabetes bei Kindern, ist das ein Notfall – ebenso wie eine diabetische Ketoazidose. Die Grundsäulen der Therapie bestehen aus Rehydratation, Insulin und Kaliumgabe. Insulin ist das Medikament der Wahl zur Behandlung der Ketoazidose.

Frühe Hypertonie erhöht späteres kardiovaskuläres Risiko

Wie wichtig es ist, pädiatrische Patienten auf Bluthochdruck zu screenen, zeigt eine kanadische Studie: Hypertone Druckwerte in Kindheit und Jugend steigern das Risiko für spätere kardiovaskuläre Komplikationen.

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.