Safety and tolerability of Bifidobacterium longum subspecies infantis EVC001 supplementation in healthy term breastfed infants: a phase I clinical trial

Jennifer T Smilowitz, Jackelyn Moya, Melissa A Breck, Chelsea Cook, Annette Fineberg, Kathleen Angkustsiri, Mark A Underwood, Jennifer T Smilowitz, Jackelyn Moya, Melissa A Breck, Chelsea Cook, Annette Fineberg, Kathleen Angkustsiri, Mark A Underwood

Abstract

Background: Historically, bifidobacteria were the dominant intestinal bacteria in breastfed infants. Still abundant in infants in developing nations, levels of intestinal bifidobacteria are low among infants in developed nations. Recent studies have described an intimate relationship between human milk and a specific subspecies of Bifidobacterium, B. longum subsp. infantis (B. infantis), yet supplementation of breastfed, healthy, term infants with this organism, has not been reported. The IMPRINT Study, a Phase I clinical trial, was initiated to determine the safety and tolerability of supplementing breastfed infants with B. infantis (EVC001).

Methods: Eighty mother-infant dyads were enrolled in either lactation support plus B. infantis supplementation (BiLS) or lactation support alone (LS). Starting with Day 7 postnatal, BiLS infants were fed 1.8-2.8 × 1010 CFU B. infantis EVC001 daily in breast milk for 21 days. Mothers collected fecal samples, filled out health questionnaires, and kept daily logs about their infants' feeding and gastrointestinal symptoms from birth until Day 61 postnatal. Safety and tolerability were determined from maternal reports.

Results: There were no differences in the mean gestational age at birth, weight 1 and 2 months postnatal, and breast milk intake between groups. The mean Log10 change in fecal Bifidobacterium from Day 6 to Day 28 was higher (p = 0.0002) for BiLS (6.6 ± 2.8 SD) than for LS infants (3.5 ± 3.5 SD). Daily stool number was higher (p < 0.005) for LS and lower (p < 0.05) for BiLS infants during supplementation than at Baseline. During supplementation, watery stools decreased and soft stools increased by 36% over baseline in BiLS infants (p < 0.05) with no significant changes in stool consistency for the LS infants. None of the safety and tolerability endpoints, including flatulence, bloody stool, body temperature, ratings of gastrointestinal symptoms, use of antibiotics or gas-relieving medications, infant colic, jaundice, number of illnesses, sick doctor visits, or diagnoses of eczema were different for the groups at any point.

Conclusions: The B. infantis EVC001 supplement was safely consumed and well-tolerated. Stools were fewer and better formed in infants in the BiLS group compared with LS group. Adverse events were those expected in healthy infants and not different between groups.

Trial registration: ClinicalTrials.gov NCT02457338 . Registered May 27, 2015.

Keywords: Bifidobacterium longum subspecies infantis; Breast milk; Gut microbiome; Human milk oligosaccharides; Infant; Probiotics; Supplementation; Tolerability.

Figures

Fig. 1
Fig. 1
Consort diagram. Consort diagram describing the number of participating mothers who were screened, randomized into the intervention groups, screened-failed post-randomization, and withdrew throughout the study period
Fig. 2
Fig. 2
Number of infant stools per day. Mean ± SD of reported number of daily infant stools for the LS (red dot plot) and BiLS (blue dot plot) groups during the Baseline, Intervention, and Post-intervention periods. n = 34 for each group during the Baseline and Intervention periods, and n = 33 for the LS, and n = 34 for the BiLS groups during the Post-intervention period. Different superscripts represent significant differences within and between interventions. There was a significant time effect (p < 0.01), time*trt interaction (p < 0.0005), and intervention effect (p < 0.0005). Based on multiple comparison post hoc analysis with Bonferroni corrections, compared with baseline the mean number of stools increased (bp < 0.0005) during the intervention period for the LS group and decreased (cp < 0.05) for the BiLS group. During the Post-intervention period, the mean number of stools returned to Baseline levels for the LS group and decreased from the Intervention period for the BiLS group (dp < 0.0005)
Fig. 3
Fig. 3
Infant stool consistency. a Mean ± SD of the proportion in reported infant stool consistency for the LS (red dot plot) and BiLS (blue dot plot) groups during the Baseline, Intervention, and Post-intervention periods. n = 34 for each group during the Baseline and Intervention periods, n = 33 for the LS group, and n = 34 for the BiLS group during the Post-intervention period. b Mean ± SD of the change in the percentage of reported infant stool consistency for the LS (red dot plot) and BiLS (blue dot plot) groups for difference between Intervention and Baseline (Intervention – Baseline), and Post-intervention and Intervention (Post-intervention – Intervention) periods. n = 34 for each group for Intervention – Baseline, and n = 33 for the LS and n = 34 for the BiLS groups for Post-intervention – Intervention. *p < 0.05
Fig. 4
Fig. 4
Infant tolerability scores. Mean ± SD of reported tolerability scores (red dot plot) and BiLS (blue dot plot) groups during the Baseline, Intervention, and Post-intervention periods. n = 34 for each group during the Baseline and Intervention periods, and n = 33 for the LS and n = 34 for the BiLS groups during the Post-intervention period. a Infant irritability, (b) infant upset after spit-ups, and (c) infant discomfort in passing gas or stool

References

    1. Smilowitz JT, Lebrilla CB, Mills DA, German JB, Freeman SL. Breast milk oligosaccharides: structure-function relationships in the neonate. Annu Rev Nutr. 2014;34:143–169. doi: 10.1146/annurev-nutr-071813-105721.
    1. Coppa GV, Gabrielli O, Pierani P, Catassi C, Carlucci A, Giorgi P. Changes in carbohydrate composition in human milk over 4 months of lactation. Pediatrics. 1993;91(5):637–641.
    1. Chaturvedi P, Warren CD, Altaye M, Morrow AL, Ruiz-Palacios G, Pickering LK, Newburg DS. Fucosylated human milk oligosaccharides vary between individuals and over the course of lactation. Glycobiology. 2001;11(5):365–372. doi: 10.1093/glycob/11.5.365.
    1. Zivkovic AM, German JB, Lebrilla CB, Mills DA. Human milk glycobiome and its impact on the infant gastrointestinal microbiota. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4653–4658. doi: 10.1073/pnas.1000083107.
    1. Tissier H. Recherches sur la flore intestinale des nourrissons: état normal et pathologique. Paris: G. Carré et C. Naud; 1900.
    1. Subramanian S, Blanton LV, Frese SA, Charbonneau M, Mills DA, Gordon JI. Cultivating healthy growth and nutrition through the gut microbiota. Cell. 2015;161(1):36–48. doi: 10.1016/j.cell.2015.03.013.
    1. Bäckhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, Li Y, Xia Y, Xie H, Zhong H. Dynamics and stabilization of the human gut Microbiome during the first year of life. Cell Host Microbe. 2015;17(5):690–703. doi: 10.1016/j.chom.2015.04.004.
    1. Sela D, Chapman J, Adeuya A, Kim J, Chen F, Whitehead T, Lapidus A, Rokhsar D, Lebrilla C, German J. The genome sequence of Bifidobacterium longum subsp. infantis reveals adaptations for milk utilization within the infant microbiome. Proc Natl Acad Sci. 2008;105(48):18964. doi: 10.1073/pnas.0809584105.
    1. LoCascio R, Ninonuevo M, Freeman S, Sela D, Grimm R, Lebrilla C, Mills D, German J. Glycoprofiling of bifidobacterial consumption of human milk oligosaccharides demonstrates strain specific, preferential consumption of small chain glycans secreted in early human lactation. J Agric Food Chem. 2007;55(22):8914–8919. doi: 10.1021/jf0710480.
    1. Garrido D, Ruiz-Moyano S, Lemay DG, Sela DA, German JB, Mills DA: Comparative transcriptomics reveals key differences in the response to milk oligosaccharides of infant gut-associated bifidobacteria. Scientific reports. 2015;5:1-7.
    1. Garrido D, Barile D, Mills DA. A molecular basis for bifidobacterial enrichment in the infant gastrointestinal tract. Advances in Nutrition: An International Review Journal. 2012;3(3):415S–421S. doi: 10.3945/an.111.001586.
    1. Kim JH, An HJ, Garrido D, German JB, Lebrilla CB, Mills DA. Proteomic analysis of Bifidobacterium longum subsp. infantis reveals the metabolic insight on consumption of prebiotics and host glycans. PLoS One. 2013;8(2):e57535. doi: 10.1371/journal.pone.0057535.
    1. Garrido D, Ruiz-Moyano S, Mills DA. Release and utilization of N-acetyl-D-glucosamine from human milk oligosaccharides by Bifidobacterium longum subsp. infantis. Anaerobe. 2012;18(4):430–435. doi: 10.1016/j.anaerobe.2012.04.012.
    1. Ward R, Niñonuevo M, Mills D, Lebrilla C, German J. In vitro fermentability of human milk oligosaccharides by several strains of bifidobacteria. Mol Nutr Food Res. 2007;51(11):1398–1405. doi: 10.1002/mnfr.200700150.
    1. Chichlowski M, De Lartigue G, German JB, Raybould HE, Mills DA. Bifidobacteria isolated from infants and cultured on human milk oligosaccharides affect intestinal epithelial function. J Pediatr Gastroenterol Nutr. 2012;55(3):321–327. doi: 10.1097/MPG.0b013e31824fb899.
    1. Underwood MA, German JB, Lebrilla CB, Mills DA: Bifidobacterium longum subspecies infantis: champion colonizer of the infant gut. Pediatr Res. 2014;77(1-2):229-35.
    1. Pokusaeva K, Fitzgerald GF, van Sinderen D. Carbohydrate metabolism in Bifidobacteria. Genes Nutr. 2011;6(3):285–306. doi: 10.1007/s12263-010-0206-6.
    1. Ganguli K, Meng D, Rautava S, Lu L, Walker WA, Nanthakumar N. Probiotics prevent necrotizing enterocolitis by modulating enterocyte genes that regulate innate immune-mediated inflammation. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2013;304(2):G132–G141. doi: 10.1152/ajpgi.00142.2012.
    1. Underwood MA, Arriola J, Gerber CW, Kaveti A, Kalanetra KM, Kananurak A, Bevins CL, Mills DA, Dvorak B. Bifidobacterium longum subsp. infantis in experimental necrotizing enterocolitis: alterations in inflammation, innate immune response, and the microbiota. Pediatr Res. 2014;76(4):326–333. doi: 10.1038/pr.2014.102.
    1. Bergmann KR, Liu SX, Tian R, Kushnir A, Turner JR, Li H-L, Chou PM, Weber CR, De Plaen IG. Bifidobacteria stabilize claudins at tight junctions and prevent intestinal barrier dysfunction in mouse necrotizing enterocolitis. Am J Pathol. 2013;182(5):1595–1606. doi: 10.1016/j.ajpath.2013.01.013.
    1. Huda M, Lewis Z, Kalanetra K, Rashid M, Raqib R, Qadri F, Underwood M, Mills D, Stephensen C. Stool microbiota and vaccine responses of infants. Pediatrics. 2014;134:1–11. doi: 10.1542/peds.2013-3937.
    1. Lewis ZT, Totten SM, Smilowitz JT, Popovic M, Parker E, Lemay DG, Van Tassell ML, Miller MJ, Jin Y-S, German JB. Maternal fucosyltransferase 2 status affects the gut bifidobacterial communities of breastfed infants. Microbiome. 2015;3(1):13. doi: 10.1186/s40168-015-0071-z.
    1. Bager P, Wohlfahrt J, Westergaard T. Caesarean delivery and risk of atopy and allergic disesase: meta-analyses. Clinical & Experimental Allergy. 2008;38(4):634–642. doi: 10.1111/j.1365-2222.2008.02939.x.
    1. Thavagnanam S, Fleming J, Bromley A, Shields M, Cardwell C. A meta-analysis of the association between Caesarean section and childhood asthma. Clinical & Experimental Allergy. 2008;38(4):629–633. doi: 10.1111/j.1365-2222.2007.02780.x.
    1. Negele K, Heinrich J, Borte M, Berg A, Schaaf B, Lehmann I, Wichmann H, Bolte G. Mode of delivery and development of atopic disease during the first 2 years of life. Pediatr Allergy Immunol. 2004;15(1):48–54. doi: 10.1046/j.0905-6157.2003.00101.x.
    1. Renz-Polster H, David M, Buist A, Vollmer W, O'Connor E, Frazier E, Wall M. Caesarean section delivery and the risk of allergic disorders in childhood. Clinical & Experimental Allergy. 2005;35(11):1466–1472. doi: 10.1111/j.1365-2222.2005.02356.x.
    1. Eggesbø M, Botten G, Stigum H, Nafstad P, Magnus P. Is delivery by cesarean section a risk factor for food allergy? J Allergy Clin Immunol. 2003;112(2):420–426. doi: 10.1067/mai.2003.1610.
    1. Vatanen T, Kostic AD, d’Hennezel E, Siljander H, Franzosa EA, Yassour M, Kolde R, Vlamakis H, Arthur TD, Hämäläinen A-M. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell. 2016;165(4):842–853. doi: 10.1016/j.cell.2016.04.007.
    1. Underwood MA, Kalanetra KM, Bokulich NA, Lewis ZT, Mirmiran M, Tancredi DJ, Mills DA: A Comparison of Two Probiotic Strains of Bifidobacteria in Premature Infants. J Pediatr. 2013;163(6):1585-91.
    1. Håkansson S, Axemo P, Bremme K, Bryngelsson A-L, Carlsson Wallin M, Ekström C-M, Granlund M, Jacobsson B, Källén K, Spetz E. Group B streptococcal carriage in Sweden: a national study on risk factors for mother and infant colonisation. Acta Obstet Gynecol Scand. 2008;87(1):50–58. doi: 10.1080/00016340701802888.
    1. Penders J, Vink C, Driessen C, London N, Thijs C, Stobberingh EE. Quantification of Bifidobacterium spp., Escherichia coli and Clostridium Difficile in faecal samples of breast-fed and formula-fed infants by real-time PCR. FEMS Microbiol Lett. 2005;243(1):141–147. doi: 10.1016/j.femsle.2004.11.052.
    1. Bekkali N, Hamers SL, Reitsma JB, Van Toledo L, Benninga MA. Infant stool form scale: development and results. J Pediatr. 2009;154(4):521–526. doi: 10.1016/j.jpeds.2008.10.010.
    1. Wessel MA, Cobb JC, Jackson EB, HARRIS GS, Detwiler AC. Paroxysmal fussing in infancy, sometimes called" colic". Pediatrics. 1954;14(5):421–435.
    1. Hoy-Schulz YE, Jannat K, Roberts T, Zaidi SH, Unicomb L, Luby S, Parsonnet J. Safety and acceptability of lactobacillus reuteri DSM 17938 and Bifidobacterium longum subspecies infantis 35624 in Bangladeshi infants: a phase I randomized clinical trial. BMC Complement Altern Med. 2016;16(1):1.
    1. Kalliomäki M, Salminen S, Arvilommi H, Kero P, Koskinen P, Isolauri E. Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet. 2001;357(9262):1076–1079. doi: 10.1016/S0140-6736(00)04259-8.
    1. Weaver LT, Ewing G, Taylor LC. The bowel habit of milk-fed infants. J Pediatr Gastroenterol Nutr. 1988;7(4):568–571. doi: 10.1097/00005176-198807000-00015.
    1. Shrago LC, Reifsnider E, Insel K. The neonatal bowel output study: indicators of adequate breast milk intake in neonates. Pediatr Nurs. 2006;32(3):195.
    1. Çamurdan AD, Beyazova U, Özkan S, Tunç VT. Defecation patterns of the infants mainly breastfed from birth till the 12th month: prospective cohort study. Turk J Gastroenterol. 2014;25(1):1–5. doi: 10.5152/tjg.2014.5218.
    1. Tham E, Nathan R, Davidson G, Moore D. Bowel habits of healthy Australian children aged 0-2 years. J Paediatr Child Health. 1996;32(6):504–507. doi: 10.1111/j.1440-1754.1996.tb00963.x.
    1. Fontana M, Bianchi C, Cataldo F, Nibali SC, Cucchiara S, Casali L, Iacono G, Sanfilippo M, Torre G. Bowel frequency in healthy children. Acta Paediatr. 1989;78(5):682–684. doi: 10.1111/j.1651-2227.1989.tb11126.x.
    1. Nyhan WL. Stool frequency of normal infants in the first week of life. Pediatrics. 1952;10(4):414–425.
    1. Tunc VT, Camurdan AD, İlhan MN, Sahin F, Beyazova U. Factors associated with defecation patterns in 0–24-month-old children. Eur J Pediatr. 2008;167(12):1357–1362. doi: 10.1007/s00431-008-0669-2.
    1. Vandeputte D, Falony G, Vieira-Silva S, Tito RY, Joossens M, Raes J. Stool consistency is strongly associated with gut microbiota richness and composition, enterotypes and bacterial growth rates. Gut. 2015;65(1):57–62. doi:10.1136/gutjnl-2015-309618.
    1. Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, Tobe T, Clarke JM, Topping DL, Suzuki T. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature. 2011;469(7331):543–547. doi: 10.1038/nature09646.
    1. Dong P, Yang Y. Wang W-p: the role of intestinal bifidobacteria on immune system development in young rats. Early Hum Dev. 2010;86(1):51–58. doi: 10.1016/j.earlhumdev.2010.01.002.
    1. Crockenberg SB, Smith P. Antecedents of mother-infant interaction and infant irritability in the first three months of life. Infant Behavior and Development. 1982;5(2–4):105–119. doi: 10.1016/S0163-6383(82)80021-0.
    1. Russo M, Martinelli M, Sciorio E, Botta C, Miele E, Vallone G, Staiano A. Stool consistency, but not frequency, correlates with total gastrointestinal transit time in children. J Pediatr. 2013;162(6):1188–1192. doi: 10.1016/j.jpeds.2012.11.082.

Source: PubMed

3
Iratkozz fel