We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Exploring the relationship between environmental enteric dysfunction and oral vaccine responses

    James A Church

    *Author for correspondence:

    E-mail Address: j.church@qmul.ac.uk

    Zvitambo Institute for Maternal & Child Health Research, Harare, Zimbabwe

    Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, UK

    ,
    Edward PK Parker

    Department of Infectious Disease Epidemiology, St Mary's Campus, Imperial College London, London, UK

    ,
    Margaret N Kosek

    Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA

    ,
    Gagandeep Kang

    Department of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India

    ,
    Nicholas C Grassly

    Department of Infectious Disease Epidemiology, St Mary's Campus, Imperial College London, London, UK

    ,
    Paul Kelly

    Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, UK

    Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Zambia

    &
    Andrew J Prendergast

    Zvitambo Institute for Maternal & Child Health Research, Harare, Zimbabwe

    Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, UK

    Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA

    Published Online:https://doi.org/10.2217/fmb-2018-0016

    Oral vaccines significantly underperform in low-income countries. One possible contributory factor is environmental enteric dysfunction (EED), a subclinical disorder of small intestinal structure and function among children living in poverty. Here, we review studies describing oral vaccine responses and EED. We identified eight studies evaluating EED and oral vaccine responses. There was substantial heterogeneity in study design and few consistent trends emerged. Four studies reported a negative association between EED and oral vaccine responses; two showed no significant association; and two described a positive correlation. Current evidence is therefore insufficient to determine whether EED contributes to oral vaccine underperformance. We identify roadblocks in the field and future research needs, including carefully designed studies those can investigate this hypothesis further.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1 GBD Diarrhoeal Diseases Collaborators. Estimates of global, regional, and national morbidity, mortality, and aetiologies of diarrhoeal diseases: a systematic analysis for the Global Burden of Disease Study 2015. Lancet Infect. Dis. 17(9), 909–948 (2017).
    • 2 Liu L, Oza S, Hogan D et al. Global, regional, and national causes of child mortality in 2000–13, with projections to inform post-2015 priorities: an updated systematic analysis. Lancet 385(9966), 430–440 (2015).
    • 3 GBD 2016 Disease and Injury Incidence and Prevalence Collaborators Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet 390(10100), 1211–1259 (2017).
    • 4 VIEW-hub. Interactive mapping tool. www.view-hub.org/viz/.
    • 5 Patel M, Shane AL, Parashar UD, Jiang B, Gentsch JR, Glass RI. Oral rotavirus vaccines: how well will they work where they are needed most? J. Infect. Dis. 200(Suppl. 1), S39–S48 (2009).
    • 6 Plotkin SA, Lebrun A, Koprowski H. Vaccination with the CHAT strain of type 1 attenuated poliomyelitis virus in Leopoldville. Belgian Congo. 2. Studies of the safety and efficacy of vaccination. Bull. World Health Organ. 22, 215–234 (1960).
    • 7 Patriarca PA, Wright PF, John TJ. Factors affecting the immunogenicity of oral poliovirus vaccine in developing countries: review. Rev. Infect. Dis. 13(5), 926–939 (1991).
    • 8 Hallander HO, Paniagua M, Espinoza F et al. Calibrated serological techniques demonstrate significant different serum response rates to an oral killed cholera vaccine between Swedish and Nicaraguan children. Vaccine 21(1–2), 138–145 (2002).
    • 9 Armah GE, Sow SO, Breiman RF et al. Efficacy of pentavalent rotavirus vaccine against severe rotavirus gastroenteritis in infants in developing countries in sub-Saharan Africa: a randomised, double-blind, placebo-controlled trial. Lancet 376(9741), 606–614 (2010). •• Efficacy trial demonstrating the underperformance of oral rotavirus vaccine across three low income countries (Ghana, Kenya and Mali).
    • 10 Zaman K, Dang DA, Victor JC et al. Efficacy of pentavalent rotavirus vaccine against severe rotavirus gastroenteritis in infants in developing countries in Asia: a randomised, double-blind, placebo-controlled trial. Lancet 376(9741), 615–623 (2010).
    • 11 Ruiz-Palacios GM, Perez-Schael I, Velazquez FR et al. Safety and efficacy of an attenuated vaccine against severe rotavirus gastroenteritis. N. Engl. J. Med. 354(1), 11–22 (2006).
    • 12 Vesikari T, Matson DO, Dennehy P et al. Safety and efficacy of a pentavalent human–bovine (WC3) reassortant rotavirus vaccine. N. Engl. J. Med. 354(1), 23–33 (2006).
    • 13 Parker EP, Ramani S, Lopman BA et al. Causes of impaired oral vaccine efficacy in developing countries. Future Microbiol. 13, 97–118 (2017). •• Review summarising the risk factors for oral vaccine failure and the heterogeneous nature of the evidence available.
    • 14 Moon SS, Tate JE, Ray P et al. Differential profiles and inhibitory effect on rotavirus vaccines of nonantibody components in breast milk from mothers in developing and developed countries. Pediatr. Infect. Dis. J. 32(8), 863–870 (2013).
    • 15 Platts-Mills JA, Babji S, Bodhidatta L et al. Pathogen-specific burdens of community diarrhoea in developing countries: a multisite birth cohort study (MAL-ED). Lancet Glob. Health 3(9), e564–e575 (2015).
    • 16 Bucardo F, Nordgren J, Reyes Y, Gonzalez F, Sharma S, Svensson L. The Lewis A phenotype is a restriction factor for Rotateq and Rotarix vaccine-take in Nicaraguan children. Sci. Rep. 8(1), 1502 (2018).
    • 17 Black RE, Victora CG, Walker SP et al. Maternal and child undernutrition and overweight in low-income and middle-income countries. Lancet 382(9890), 427–451 (2013).
    • 18 Smith MI, Yatsunenko T, Manary MJ et al. Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science 339(6119), 548–554 (2013).
    • 19 Smith LE, Stoltzfus RJ, Prendergast A. Food chain mycotoxin exposure, gut health, and impaired growth: a conceptual framework. Adv. Nutr. 3(4), 526–531 (2012).
    • 20 Prendergast A, Kelly P. Enteropathies in the developing world: neglected effects on global health. Am. J. Trop. Med. Hyg. 86(5), 756–763 (2012).
    • 21 Campbell DI, Murch SH, Elia M et al. Chronic T cell-mediated enteropathy in rural west African children: relationship with nutritional status and small bowel function. Pediatr. Res. 54(3), 306–311 (2003). • Highlights the potential clinical importance of impaired gut barrier function in mediating stunting in infants in developing countries.
    • 22 Veitch AM, Kelly P, Zulu IS, Segal I, Farthing MJ. Tropical enteropathy: a T-cell-mediated crypt hyperplastic enteropathy. Eur. J. Gastroenterol. Hepatol. 13(10), 1175–1181 (2001).
    • 23 Brewster DR, Manary MJ, Menzies IS, O'loughlin EV, Henry RL. Intestinal permeability in kwashiorkor. Arch. Dis. Child. 76(3), 236–241 (1997).
    • 24 Campbell DI, Elia M, Lunn PG. Growth faltering in rural Gambian infants is associated with impaired small intestinal barrier function, leading to endotoxemia and systemic inflammation. J. Nutr. 133(5), 1332–1338 (2003).
    • 25 Cook GC. Aetiology and pathogenesis of postinfective tropical malabsorption (tropical sprue). Lancet 1(8379), 721–723 (1984).
    • 26 Lindenbaum J, Alam AK, Kent TH. Subclinical small-intestinal disease in East Pakistan. Br. Med. J. 2(5530), 1616–1619 (1966).
    • 27 Chacko CJ, Paulson KA, Mathan VI, Baker SJ. The villus architecture of the small intestine in the tropics: a necropsy study. J. Pathol. 98(2), 146–151 (1969).
    • 28 Gerson CD, Kent TH, Saha JR, Siddiqi N, Lindenbaum J. Recovery of small-intestinal structure and function after residence in the tropics. II. Studies in Indians and Pakistanis living in New York City. Ann. Intern. Med. 75(1), 41–48 (1971).
    • 29 Lindenbaum J, Gerson CD, Kent TH. Recovery of small-intestinal structure and function after residence in the tropics. I. Studies in Peace Corps volunteers. Ann. Intern. Med. 74(2), 218–222 (1971).
    • 30 Lin A, Arnold BF, Afreen S et al. Household environmental conditions are associated with enteropathy and impaired growth in rural Bangladesh. Am. J. Trop. Med. Hyg. 89(1), 130–137 (2013).
    • 31 Brown EM, Wlodarska M, Willing BP et al. Diet and specific microbial exposure trigger features of environmental enteropathy in a novel murine model. Nat. Commun. 6, 7806 (2015). • The first animal model of environmental enteric dysfunction (EED) which showed the importance of undernutrition and specific microbial exposure in recapitulating the intestinal pathology found in humans.
    • 32 Steiner TS, Lima AA, Nataro JP, Guerrant RL. Enteroaggregative Escherichia coli produce intestinal inflammation and growth impairment and cause interleukin-8 release from intestinal epithelial cells. J. Infect. Dis. 177(1), 88–96 (1998).
    • 33 Opintan JA, Newman MJ, Ayeh-Kumi PF et al. Pediatric diarrhea in southern Ghana: etiology and association with intestinal inflammation and malnutrition. Am. J. Trop. Med. Hyg. 83(4), 936–943 (2010).
    • 34 Roy SK, Behrens RH, Haider R et al. Impact of zinc supplementation on intestinal permeability in Bangladeshi children with acute diarrhoea and persistent diarrhoea syndrome. J. Pediatr. Gastroenterol. Nutr. 15(3), 289–296 (1992).
    • 35 Goto R, Panter-Brick C, Northrop-Clewes CA, Manahdhar R, Tuladhar NR. Poor intestinal permeability in mildly stunted Nepali children: associations with weaning practices and Giardia lamblia infection. Br. J. Nutr. 88(2), 141–149 (2002).
    • 36 Donowitz JR, Haque R, Kirkpatrick BD et al. Small intestine bacterial overgrowth and environmental enteropathy in Bangladeshi children. mBio 7(1), e02102–e02115 (2016).
    • 37 Fagundes Neto U, Martins MC, Lima FL, Patricio FR, Toledo MR. Asymptomatic environmental enteropathy among slum-dwelling infants. J. Am. Coll. Nutr. 13(1), 51–56 (1994).
    • 38 Ordiz MI, Stephenson K, Agapova S et al. Environmental enteric dysfunction and the fecal microbiota in Malawian children. Am. J. Trop. Med. Hyg. 96(2), 473–476 (2017).
    • 39 Keusch GT, Denno DM, Black RE et al. Environmental enteric dysfunction: pathogenesis, diagnosis, and clinical consequences. Clin. Infect. Dis. 59(Suppl. 4), S207–S212 (2014). • Historical review of EED including description of four categories of aberrant gut structure and function.
    • 40 Kelly P, Menzies I, Crane R et al. Responses of small intestinal architecture and function over time to environmental factors in a tropical population. Am. J. Trop. Med. Hyg. 70(4), 412–419 (2004). • Evaluates mucosal architecture and function longitudinally and described associations with stool microbiology and environmental factors.
    • 41 Papadia C, Kelly P, Caini S et al. Plasma citrulline as a quantitative biomarker of HIV-associated villous atrophy in a tropical enteropathy population. Clin. Nutr. 29(6), 795–800 (2010).
    • 42 Korpe PS, Petri WA Jr. Environmental enteropathy: critical implications of a poorly understood condition. Trends Mol. Med. 18(6), 328–336 (2012).
    • 43 Serazin AC, Shackelton LA, Wilson C, Bhan MK. Improving the performance of enteric vaccines in the developing world. Nat. Immunol. 11(9), 769–773 (2010).
    • 44 Patel M, Glass RI, Jiang B, Santosham M, Lopman B, Parashar U. A systematic review of anti-rotavirus serum IgA antibody titer as a potential correlate of rotavirus vaccine efficacy. J. Infect. Dis. 208(2), 284–294 (2013).
    • 45 Deming MS, Linkins RW, Jaiteh KO, Hull HF. The clinical efficacy of trivalent oral polio vaccine in The Gambia by season of vaccine administration. J. Infect. Dis. 175(Suppl. 1), S254–S257 (1997).
    • 46 Keusch GT, Rosenberg IH, Denno DM et al. Implications of acquired environmental enteric dysfunction for growth and stunting in infants and children living in low- and middle-income countries. Food Nutr. Bull. 34(3), 357–364 (2013).
    • 47 Petri WA Jr, Naylor C, Haque R. Environmental enteropathy and malnutrition: do we know enough to intervene? BMC Med. 12, 187 (2014).
    • 48 Denno DM, Tarr PI, Nataro JP. Environmental enteric dysfunction: a case definition for intervention trials. Am. J. Trop. Med. Hyg. 97(6), 1643–1646 (2017).
    • 49 Levine MM. Immunogenicity and efficacy of oral vaccines in developing countries: lessons from a live cholera vaccine. BMC Biol. 8, 129 (2010).
    • 50 Gilmartin AA, Petri WA Jr. Exploring the role of environmental enteropathy in malnutrition, infant development and oral vaccine response. Philos. Trans. R. Soc. Lond. B Biol. Sci. 370(1671), pii:20140143 (2015).
    • 51 Becker-Dreps S, Vilchez S, Bucardo F et al. The association between fecal biomarkers of environmental enteropathy and rotavirus vaccine response in Nicaraguan infants. Pediatr. Infect. Dis. J. 36(4), 412–416 (2017). •• Evaluates immune responses to oral rotavirus vaccine in Nicaraguan infants with EED.
    • 52 Grassly NC, Praharaj I, Babji S et al. The effect of azithromycin on the immunogenicity of oral poliovirus vaccine: a double-blind randomised placebo-controlled trial in seronegative Indian infants. Lancet Infect. Dis. 16(8), 905–914 (2016). •• In this randomized placebo controlled trial, azithromycin had no impact on oral polio vaccine immunogenicity but did reduce biomarkers of EED.
    • 53 Kosek MN, Mduma E, Kosek PS et al. Plasma tryptophan and the kynurenine–tryptophan ratio are associated with the acquisition of statural growth deficits and oral vaccine underperformance in populations with environmental enteropathy. Am. J. Trop. Med. Hyg. 95(4), 928–937 (2016). •• Evaluates immune responses to oral polio vaccine in Peruvian infants with EED, employing two novel biomarkers of EED.
    • 54 Lagos R, Fasano A, Wasserman SS et al. Effect of small bowel bacterial overgrowth on the immunogenicity of single-dose live oral cholera vaccine CVD 103-HgR. J. Infect. Dis. 180(5), 1709–1712 (1999).
    • 55 Mwape I, Bosomprah S, Mwaba J et al. Immunogenicity of rotavirus vaccine (RotarixTM) in infants with environmental enteric dysfunction. PLoS ONE 12(12), e0187761 (2017). • Evaluates immune responses to oral rotavirus vaccine in Zambian children with EED.
    • 56 Naylor C, Lu M, Haque R et al. Environmental enteropathy, oral vaccine failure and growth faltering in infants in Bangladesh. EBioMedicine 2(11), 1759–1766 (2015). •• Evaluates immune responses to oral polio and oral rotavirus vaccines in Bangladeshi infants with EED.
    • 57 Uddin MI, Islam S, Nishat NS et al. Biomarkers of environmental enteropathy are positively associated with immune responses to an oral cholera vaccine in Bangladeshi children. PLoS Negl. Trop. Dis. 10(11), e0005039 (2016). •• Evaluates immune responses to oral cholera vaccine in Bangladeshi children with EED.
    • 58 Prendergast AJ, Humphrey JH, Mutasa K et al. Assessment of environmental enteric dysfunction in the SHINE trial: methods and challenges. Clin. Infect. Dis. 61(Suppl. 7), S726–S732 (2015).
    • 59 Peterson KM, Guo X, Elkahloun AG et al. The expression of REG 1A and REG 1B is increased during acute amebic colitis. Parasitol. Int. 60(3), 296–300 (2011).
    • 60 Granlund A, Beisvag V, Torp SH et al. Activation of REG family proteins in colitis. Scand. J. Gastroenterol. 46(11), 1316–1323 (2011).
    • 61 Crenn P, Messing B, Cynober L. Citrulline as a biomarker of intestinal failure due to enterocyte mass reduction. Clin. Nutr. 27(3), 328–339 (2008).
    • 62 Prata MM, Havt A, Bolick DT, Pinkerton R, Lima A, Guerrant RL. Comparisons between myeloperoxidase, lactoferrin, calprotectin and lipocalin-2, as fecal biomarkers of intestinal inflammation in malnourished children. J. Transl. Sci. 2(2), 134–139 (2016).
    • 63 Kosek M, Haque R, Lima A et al. Fecal markers of intestinal inflammation and permeability associated with the subsequent acquisition of linear growth deficits in infants. Am. J. Trop. Med. Hyg. 88(2), 390–396 (2013).
    • 64 Guerrant RL, Leite AM, Pinkerton R et al. Biomarkers of environmental enteropathy, inflammation, stunting, and impaired growth in children in northeast Brazil. PLoS ONE 11(9), e0158772 (2016).
    • 65 Kosek M, Guerrant RL, Kang G et al. Assessment of environmental enteropathy in the MAL-ED cohort study: theoretical and analytic framework. Clin. Infect. Dis. 59(Suppl. 4), S239–S247 (2014).
    • 66 Richard DM, Dawes MA, Mathias CW, Acheson A, Hill-Kapturczak N, Dougherty DM. L-tryptophan: basic metabolic functions, behavioral research and therapeutic indications. Int. J. Tryptophan. Res. 2, 45–60 (2009).
    • 67 Ciorba MA. Indoleamine 2,3 dioxygenase in intestinal disease. Curr. Opin. Gastroenterol. 29(2), 146–152 (2013).
    • 68 Gupta NK, Thaker AI, Kanuri N et al. Serum analysis of tryptophan catabolism pathway: correlation with Crohn's disease activity. Inflamm. Bowel Dis. 18(7), 1214–1220 (2012).
    • 69 Lee S, Byakwaga H, Boum Y et al. Immunologic pathways that predict mortality in HIV-infected Ugandans initiating ART. J. Infect. Dis. 215(8), 1270–1274 (2017).
    • 70 Nikolaus S, Schulte B, Al-Massad N et al. Increased tryptophan metabolism is associated with activity of inflammatory bowel diseases. Gastroenterology 153(6), 1504.e2–1516.e2 (2017).
    • 71 Riordan SM, Mciver CJ, Walker BM, Duncombe VM, Bolin TD, Thomas MC. The lactulose breath hydrogen test and small intestinal bacterial overgrowth. Am. J. Gastroenterol. 91(9), 1795–1803 (1996).
    • 72 Naylor C, Lu M, Haque R et al. Environmental enteropathy, oral vaccine failure and growth faltering in infants in Bangladesh. EBioMedicine 2(11), 1759–1766 (2015).
    • 73 Church JPE, Kirkpatrick B, Grassly N, Prendergast AJ. Interventions to improve oral vaccine performance in developing countries: protocol for a systematic review and meta-analysis. (2017). www.crd.york.ac.uk/PROSPEROFILES/60608_PROTOCOL_20180108.pdf
    • 74 Louis-Auguste J, Greenwald S, Simuyandi M, Soko R, Banda R, Kelly P. High dose multiple micronutrient supplementation improves villous morphology in environmental enteropathy without HIV enteropathy: results from a double-blind randomised placebo controlled trial in Zambian adults. BMC Gastroenterol. 14, 15 (2014).
    • 75 Smith HE, Ryan KN, Stephenson KB et al. Multiple micronutrient supplementation transiently ameliorates environmental enteropathy in Malawian children aged 12–35 months in a randomized controlled clinical trial. J. Nutr. 144(12), 2059–2065 (2014).
    • 76 Wang AZ, Shulman RJ, Crocker AH et al. A combined intervention of zinc, multiple micronutrients, and albendazole does not ameliorate environmental enteric dysfunction or stunting in rural malawian children in a double-blind randomized controlled trial. J. Nutr. 147(1), 97–103 (2017).
    • 77 Trehan I, Benzoni NS, Wang AZ et al. Common beans and cowpeas as complementary foods to reduce environmental enteric dysfunction and stunting in Malawian children: study protocol for two randomized controlled trials. Trials 16, 520 (2015).
    • 78 Jones KD, Hunten-Kirsch B, Laving AM et al. Mesalazine in the initial management of severely acutely malnourished children with environmental enteric dysfunction: a pilot randomized controlled trial. BMC Med. 12, 133 (2014).
    • 79 Trehan I, Shulman RJ, Ou CN, Maleta K, Manary MJ. A randomized, double-blind, placebo-controlled trial of rifaximin, a nonabsorbable antibiotic, in the treatment of tropical enteropathy. Am. J. Gastroenterol. 104(9), 2326–2333 (2009).
    • 80 Galpin L, Manary MJ, Fleming K, Ou CN, Ashorn P, Shulman RJ. Effect of lactobacillus GG on intestinal integrity in Malawian children at risk of tropical enteropathy. Am. J. Clin. Nutr. 82(5), 1040–1045 (2005).
    • 81 Chattha KS, Vlasova AN, Kandasamy S, Rajashekara G, Saif LJ. Divergent immunomodulating effects of probiotics on T cell responses to oral attenuated human rotavirus vaccine and virulent human rotavirus infection in a neonatal gnotobiotic piglet disease model. J. Immunol. 191(5), 2446–2456 (2013).
    • 82 Maier EA, Weage KJ, Guedes MM et al. Protein-energy malnutrition alters IgA responses to rotavirus vaccination and infection but does not impair vaccine efficacy in mice. Vaccine 32(1), 48–53 (2013).
    • 83 Twitchell EL, Tin C, Wen K et al. Modeling human enteric dysbiosis and rotavirus immunity in gnotobiotic pigs. Gut Pathog. 8, 51 (2016).
    • 84 Mckinley M, Leibowitz S, Bronzo R, Zanzi I, Weissman G, Schiffman G. Appropriate response to pneumococcal vaccine in celiac sprue. J. Clin. Gastroenterol. 20(2), 113–116 (1995).
    • 85 Sari S, Dalgic B, Basturk B, Gonen S, Soylemezoglu O. Immunogenicity of hepatitis A vaccine in children with celiac disease. J. Pediatr. Gastroenterol. Nutr. 53(5), 532–535 (2011).
    • 86 Schappi MG, Meier S, Bel M, Siegrist CA, Posfay-Barbe KM, Group HNS. Protective antibody responses to influenza A/H1N1/09 vaccination in children with celiac disease. J. Pediatr. Gastroenterol. Nutr. 54(6), 817–819 (2012).
    • 87 Opri R, Veneri D, Mengoli C, Zanoni G. Immune response to Hepatitis B vaccine in patients with celiac disease: a systematic review and meta-analysis. Hum. Vaccin. Immunother. 11(12), 2800–2805 (2015).
    • 88 Beale AJ, Douglas AP, Parish WE, Hobbs JR. Impaired IgA responses in coeliac disease. Lancet 1(7711), 1198–1200 (1971).
    • 89 Mawhinney H, Love AH. The immunoglobulin class responses to oral poliovaccine in coeliac disease. Clin. Exp. Immunol. 21(3), 399–406 (1975).
    • 90 Burnham WR, Lennard-Jones JE, Hecketsweiler P, Colin R, Geffroy Y. Oral BCG vaccine in Crohn's disease. Gut 20(3), 229–233 (1979).
    • 91 Nguyen DL, Nguyen ET, Bechtold ML. Effect of immunosuppressive therapies for the treatment of inflammatory bowel disease on response to routine vaccinations: a meta-analysis. Dig. Dis. Sci. 60(8), 2446–2453 (2015).
    • 92 Mcgrath EJ, Thomas R, Duggan C, Asmar BI. Pentavalent rotavirus vaccine in infants with surgical gastrointestinal disease. J. Pediatr. Gastroenterol. Nutr. 59(1), 44–48 (2014).
    • 93 Kilhamn J, Brevinge H, Svennerholm AM, Jertborn M. Immune responses in ileostomy fluid and serum after oral cholera vaccination of patients colectomized because of ulcerative colitis. Infect. Immun. 66(8), 3995–3999 (1998).
    • 94 Factors affecting the immunogenicity of oral poliovirus vaccine: a prospective evaluation in Brazil and the Gambia. World Health Organization Collaborative Study Group on Oral Poliovirus Vaccine. J. Infect. Dis. 171(5), 1097–1106 (1995).
    • 95 Cardemil CV, Estivariz C, Shrestha L et al. The effect of diarrheal disease on bivalent oral polio vaccine (bOPV) immune response in infants in Nepal. Vaccine 34(22), 2519–2526 (2016).
    • 96 Parker EP, Kampmann B, Kang G, Grassly NC. Influence of enteric infections on response to oral poliovirus vaccine: a systematic review and meta-analysis. J. Infect. Dis. 210(6), 853–864 (2014).
    • 97 Taniuchi M, Platts-Mills JA, Begum S et al. Impact of enterovirus and other enteric pathogens on oral polio and rotavirus vaccine performance in Bangladeshi infants. Vaccine 34(27), 3068–3075 (2016).
    • 98 Harris VC, Armah G, Fuentes S et al. Significant correlation between the infant gut microbiome and rotavirus vaccine response in rural Ghana. J. Infect. Dis. 215(1), 34–41 (2017).
    • 99 Parker EPK, Praharaj I, Zekavati A et al. Influence of the intestinal microbiota on the immunogenicity of oral rotavirus vaccine given to infants in south India. Vaccine 36(2), 264–272 (2017).
    • 100 Mccormick BJ, Lee GO, Seidman JC et al. Dynamics and trends in fecal biomarkers of gut function in children from 1–24 months in the MAL-ED study. Am. J. Trop. Med. Hyg. 96(2), 465–472 (2017).
    • 101 Angel J, Steele AD, Franco MA. Correlates of protection for rotavirus vaccines: possible alternative trial endpoints, opportunities, and challenges. Hum. Vaccin. Immunother. 10(12), 3659–3671 (2014).
    • 102 Prendergast AJ, Rukobo S, Chasekwa B et al. Stunting is characterized by chronic inflammation in Zimbabwean infants. PLoS ONE 9(2), e86928 (2014).
    • 103 Owino V, Ahmed T, Freemark M et al. Environmental enteric dysfunction and growth failure/stunting in global child health. Pediatrics 138(6), pii:e20160641 (2016).
    • 104 Lu M, Zhou J, Naylor C et al. Application of penalized linear regression methods to the selection of environmental enteropathy biomarkers. Biomark. Res. 5, 9 (2017).