Skip to main content
Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America logoLink to Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America
. 2014 Nov 1;59(Suppl 4):S273–S279. doi: 10.1093/cid/ciu611

Evaluating Associations Between Vaccine Response and Malnutrition, Gut Function, and Enteric Infections in the MAL-ED Cohort Study: Methods and Challenges

Christel Hoest 1, Jessica C Seidman 1, William Pan 2, Ramya Ambikapathi 1, Gagandeep Kang 3, Margaret Kosek 4, Stacey Knobler 1, Carl J Mason 5, Mark Miller 1, The MAL-ED Network Investigators
PMCID: PMC4204607  PMID: 25305297

Abstract

Most vaccine assessments have occurred in well-nourished populations of higher socioeconomic status. However, vaccines are often used in populations with high incidences of malnutrition and infections, in whom the effectiveness of some vaccines is inferior for unknown reasons. The degree and extent of vaccine underperformance have not been systematically studied for most vaccines across differing epidemiologic settings. This paper outlines the methods used and challenges associated with measuring immunological responses to oral vaccines against poliovirus and rotavirus, and parenteral vaccines against pertussis, tetanus, and measles in an observational study that monitored daily illness, monthly growth, intestinal inflammation and permeability, pathogen burden, dietary intake, and micronutrient status in children in 8 countries. This evaluation of vaccine response in the context of low- and middle-income countries is intended to address the gaps in knowledge of the heterogeneity in vaccine response in diverse epidemiological settings and the interplay between infections, nutrition, and immune response.

Keywords: vaccines, malnutrition, enteric infections, gut function, MAL-ED


Multiple factors are associated with immune response to vaccines administered during childhood as measured by antibody titers. Among the most important is the timing of antigen exposure(s), including age of first presentation and the interval between vaccine doses. Other factors may include infections (recent and/or frequent diarrhea, respiratory illness, other concurrent infections) [1, 2], malnutrition, particularly micronutrient deficiencies (vitamin A, iron, and zinc deficiency), stunting and wasting [35], intestinal dysfunction (malabsorption, inflammation, overgrowth) [6], maternal exposures (maternal antibody level, breastfeeding) [7], and early exposures to environmental antigens influenced by high population density, sanitation, or siblings [8].

The amount of antigen presented to the immune system is also affected by the route of delivery. The quantity of antigen presented is likely to be less variable for parenteral vaccines, where a fixed dosage of antigen is injected, as compared to orally administered vaccines where the quantity of antigen presented to the immune system may be influenced by breastfeeding, diet, and enteric infection [9, 10]. Orally administered vaccines against polio (OPV), cholera, and rotavirus elicit poorer immune responses in lower socioeconomic settings [1116]; however, the specific reasons for this lack of an adequate immune response are poorly understood. With the continued administration of OPV, and the recent introductions of oral rotavirus vaccines into vaccination programs in low- and middle-income countries, it is critical to understand the extent to which gut function and nutritional, infectious, and environmental exposures impact immune response to both oral and parenteral vaccines in early childhood.

The Etiology, Risk Factors and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health and Development (MAL-ED) cohort study was designed to measure associations between antibody titer levels to vaccines commonly administered during the first 2 years of life (primarily based on the World Health Organization's [WHO] Expanded Programme on Immunization [EPI] schedule) and micro- and macronutritional status, stool pathogen carriage, diarrheal illness, and assessments of intestinal permeability and inflammation [17]. One of the specific goal was to examine a potentially diminished immune response in the context of gut dysfunction. This measure is crucial when evaluating suspected factors influencing the vaccine response in low- and middle-income countries [18]. The study followed children from birth to 2 years of age in 8 diverse sites (>200 children per site): Dhaka, Bangladesh (BGD); Fortaleza, Brazil (BRF); Vellore, India (INV); Bhaktapur, Nepal (NEB); Loreto, Peru (PEL); Naushahro Feroze, Pakistan (PKN); Venda, South Africa (SAV); and Haydom, Tanzania (TZH) [1926]. This paper outlines the rationale, methodology, and definitions used for the assessment of vaccine response in the MAL-ED cohort study.

METHODS

Vaccine Schedules

The MAL-ED cohort study was observational and vaccines were not supplied or administered by study staff at the 8 study sites. Table 1 lists the national vaccine schedules followed for each of the study sites. Bacillus Calmette-Guerin (BCG), diphtheria-pertussis-tetanus (DPT), hepatitis B, oral polio vaccine (OPV), Haemophilus influenzae type B (Hib), and measles vaccine were administered at all sites; whereas, rotavirus, pneumococcal conjugate (PCV), and yellow fever (YF) vaccines were administered at 2 or more sites. Hib was administered at all sites throughout the study period except INV, where it was introduced in December 2011. In South Africa, inactivated polio vaccine (IPV) was administered as part of the EPI schedule along with OPV. In India and Pakistan, national vaccination campaigns using OPV occurred frequently, permitting measurement of response to a variable number of OPV doses. Vitamin A supplementation was coadministered according to the national vaccination schedule in 7 of 8 sites (Table 1).

Table 1.

MAL-ED Site Vaccination Schedules

Vaccine and Dose No. BGD BRF INV NEB PEL PKN SAV TZH
Bacillus Calmette-Guerin (BCG)
 BCG1 Birth Birth Birth Birth Birth Birth Birth Birth
Oral Polio (OPV)
 OPV1 6w 2m Birth 6w 2m Birth Birth Birth
 OPV2 10w 4m 6w 10w 4m 6w 6w 1m
 OPV3 14w 6m 10w 14w 6m 10w 2m
 OPV4 9m 15m 14w 14w 3m
 OPV5 16-24m
Inactivated Polio (IPV)
 IPV1 6w
 IPV2 10w
 IPV3 14w
 IPV4 18m
Diphtheria/Pertussis/Tetanus (DPT)
 DPT1 6w 2m 6w 6w 2m 6w 6w 1m
 DPT2 10w 4m 10w 10w 4m 10w 10w 2m
 DPT3 14w 6m 14w 14w 6m 14w 14w 3m
 DPT4 15m 16-24m 18m 18m
Hepatitis B (HepB)
 HepB1 6w Birth 6w 6w Birth 6w 6w 1m
 HepB2 10w 1m 10w 10w 2m 10w 10w 2m
 HepB3 14w 6m 14w 14w 4m 14w 14w 3m
 HepB4 6m
Heamophilus Influenzae B (HiB)
 HiB1 6w 2m 6w 6w 2m 6w 6w 1m
 HiB2 10w 4m 10w 10w 4m 10w 10w 2m
 HiB3 14w 6m 14w 14w 6m 14w 14w 3m
 HiB4 18m 18m
Measles
 Measles1 9m 12m 9-12m 9m 12m 9m 9m 9m
 Measles2 15-18m 15m 12-15m 15m 15m 18m
Rotavirus
 Rotavirus1 2m 2m 6w
 Rotavirus2 4m 4m 14w
Yellow Fever
 YF1 9m 12m
Pneumococcal Conjugate (PCV)
 PCV1 2m 3m 6w
 PCV2 4m 5m 14w
 PCV3 6m 7m 9m
 PCV4 12m
Vitamin A
 Vitamin A1 9m 6m 9m 6m 6m 6m 9m
 Vitamin A2 12m 18m 12m 12m 12m 15m
 Vitamin A3 18m 24m 18m 18m 18m 21m
 Vitamin A4 24m 24m

Abbreviations: m, month; w, weeks; BGD, Dhaka, Bangladesh; BRF, Fortaleza, Brazil; INV, Vellore, India; NEB, Bhaktapur, Nepal; PEL, Loreto, Peru; PKN, Naushahro Feroze, Pakistan; SAV, Venda, South Africa; TZH, Haydom, Tanzania.

Vaccine Coverage Data Collection

Although national vaccine schedules specified the ages for the administration of each included vaccine, the actual ages of receipt through routine immunization programs varied. Data on the frequency and timing of vaccine administration were actively collected by MAL-ED field-workers during monthly household visits, ideally within a 2-day window of the monthly birth anniversary [27]. During the monthly household visit, MAL-ED field-workers recorded the vaccines administered and dates of administration on the Monthly Form A/B (MOA/MOB). The information was ideally obtained from the vaccine card; however, when vaccine cards were not available, clinical records were utilized where possible. If neither vaccine nor clinical records were available, field-workers asked the mother or caregiver if vaccines were administered since the previous monthly visit, and inquired about the type and the date of vaccine administration. Additionally, a quarterly assessment of vaccines administered and dates of administration using the Vaccine Information Form (VIF) functioned as a validation tool for the data collected on the monthly forms. The source of vaccine data was also collected on the VIF (ie, vaccine record, clinical record, mother or caregiver's report) for all vaccines administered.

Natural Exposure

Natural exposure to a vaccine-preventable disease (VPD) could influence antibody titers. The MAL-ED study design included a twice-weekly visit and monitored for any reported or referred illness [17]. Although no environmental sampling was conducted, this active surveillance allowed for the possibility to detect whether mild or severe VPD was present in individuals or the occurrence of a VPD outbreak. Wild polioviruses were not known to be circulating in any of our study sites, with the possible exception of the Pakistan site. We were not able to ascertain whether wild viruses were circulating in Pakistan but had not received any reports of poliomyelitis in our study population.

Blood Sample Collection

Blood collection was scheduled for each child at 7 and 15 months of age. On the day of blood collection, the child had to be free of symptoms of illness (ie, fever, acute lower respiratory infection, diarrhea, or dehydration). To accommodate scheduling and illness episodes, the blood collection window extended from 2 days before to 12 days after the target collection date. A maximum of 5 mL of blood was drawn from each child by MAL-ED study staff trained in phlebotomy at the child's home or in a health clinic. Samples were centrifuged at 2000g for 10–15 minutes, and aliquoted and frozen at −70°C prior to shipping and testing.

Vaccine Response Determination

Enzyme-linked immunosorbent assays (ELISA) were used to quantitatively measure the immunoglobulin G (IgG) antibody levels to poliovirus, measles virus, tetanus toxoid, and pertussis toxoid, and the immunoglobulin A (IgA) and IgG antibody levels to rotavirus. Paired samples (at 7 and 15 months) were run on the same plate for all ELISAs. For 6 study sites (BGD, BRF, NEB, PEL, SAV, and TZH), assays for all vaccines other than OPV were performed centrally at the Armed Forces Research Institute for Medical Studies (AFRIMS) in Bangkok, Thailand; INV and PKN study sites performed the assays in-country. For the poliovirus (types 1, 2, and 3) antibody neutralization assay, samples from all 8 sites were evaluated at WHO reference laboratories (Centers for Disease Control and Prevention [CDC], Atlanta, Georgia, for 7 sites and the Enterovirus Research Centre, Mumbai, India, for INV).

Measles, Tetanus, Pertussis, and Poliovirus IgG Assays

Quantitative antimeasles, anti–tetanus toxoid, and anti–pertussis toxin IgG ELISAs (Euroimmun, Lubeck, Germany) and antipoliovirus IgG ELISAs (Genway, San Diego) were centrally procured and run at AFRIMS for 6 sites; kits and supplies were provided and the assays performed on-site in INV and PKN. Assays were conducted according to the manufacturer's instructions.

Rotavirus Assays

Quantitative antirotavirus serum IgG and IgA ELISAs were conducted following previously published methods [2830]. These assays were performed centrally for 6 of the sites, and on site in INV and PKN. In brief, microplates were coated with rabbit antirotavirus IgG, and after washing, either cell lysate or virus preparation was added to alternating rows. Eight 2-fold dilutions starting with 1:80 dilutions of the serum IgA and IgG standards were prepared. Four 2-fold dilutions of 1:20 dilutions of known reference IgA and IgG and unknown serum or plasma samples were prepared. After washing, the serum standard dilutions and serum sample dilutions were added to the microplates. After washing again, biotinylated rabbit antihuman IgA (for the IgA plates) or IgG (for the IgG plates) was added and followed by washing and addition of avidin-biotin-peroxidase complex. After the final wash, O-phenylenediamine dihydrochloride substrate was added to each well and the reaction stopped with sulfuric acid. The plates were read at 492 nm, and the titers were computed from a 4-parameter fit of the transformed optical density values.

Poliovirus Neutralizing Antibody Assays

Randomized and blinded serum samples from BGD, BRF, NEB, PEL, PKN, SAV, and TZH were shipped frozen to the CDC, where neutralization titers were determined. The serum or plasma samples were tested to detect neutralizing antibody to poliovirus types 1, 2, and 3 [31]. In brief, to conduct the microneutralization assay, a 4-fold dilution of each sample ranging from 1:4 to 1:512 was prepared and 25 µL was distributed in each of 6 wells of 96-well microtiter plates. Approximately 100 median tissue culture infective doses (TCID50) of Sabin poliovirus strains were added to replicate wells at each dilution and the mixtures incubated at 37°C for 1 hour before the addition of 50 000 cells/mL of Vero cells. Following further incubation at 37°C for 3 days, the cells were fixed, stained, and examined for cytopathic effect. Positive controls were set up in parallel for each poliovirus as “back titrations” to ensure that an adequate amount of virus was added, with back titrations required in the range of 30–300 TCID50 of input virus. The endpoint was defined as the highest dilution of serum that showed 50% or greater reduction in cytopathic effect. A neutralizing antibody titer of ≥1:8 was considered protective.

Quality Control

The quality control systems established at study sites and laboratories for data collection and sample evaluation were designed to facilitate cross-site comparison. The protocols used for data and sample collection and processing were uniform across all sites. Quality control performed on the vaccine coverage data focused on minimizing discrepancies and inconsistencies between recorded vaccines and vaccination dates. Discrepancies in vaccine names or vaccination dates between the 2 vaccine data collection forms (the monthly surveillance form MOA/MOB and the quarterly VIF) were identified and reported back to the sites for correction. Data from the assays for antibodies were generated centrally at AFRIMS for 6 of the 8 sites. As international shipment of biological samples is not permitted in India and Pakistan, the same kits, controls, and protocols were used to decrease the risk of variability. The poliovirus neutralizations assays were performed at WHO reference laboratories.

CHALLENGES

Data Collection

The vaccine information was ideally obtained from vaccination cards that were legible and fully completed by a health worker at the time of vaccine administration. However, if vaccination cards were not available, study staff relied on other clinical records or the memory of the caregiver for information on the specific vaccines received and dates. A common problem encountered was the inconsistent naming of vaccines on the vaccination cards (eg, brand names and antigens were often used interchangeably). This was a particular concern with the use of combination vaccines. However, as vaccines were mainly provided by the public sector at the MAL-ED study sites, verification of a particular brand distributed from local clinics helped to clarify which vaccines were administered.

Sample Collection

Obtaining 5-mL blood volume collection from infants with limited phlebotomy attempts was a significant challenge. Although adequate blood samples were important to complete all assays prescribed by protocol, field-workers sought to maintain a balance between needing multiple attempts to collect the full volume of blood with minimizing the burden on the children and inducing study dropouts. Participants were allowed to refuse blood collection without sacrificing their participation in other areas of the MAL-ED study.

Data Analysis

This is the first study that seeks to evaluate immune responses to routine childhood vaccines in lower socioeconomic settings across 3 continents using unified assessments. The scope of MAL-ED posed significant analytic challenges. The analysis of vaccine response has 2 primary objectives: (1) to identify and quantify factors associated with vaccine titers, and (2) to examine factors that influence the trajectory of vaccine titers within the first 2 years of life. Both objectives pose challenges that require particular statistical care to achieve proper inference due to the variability that exists between and within sites. Perhaps most challenging is modeling the distributional variability of titers that exists due to the variation in vaccine schedules, diarrheal (and other) disease burden, and childhood nutrition. Such factors are often controlled for in a mixed-effects regression framework [32, 33] that includes a site-specific random effect; however, some vaccine titers exhibit distributions that vary across the longitudinal sample, possibly requiring a mixture modeling approach. For example, pertussis titers have a zero-inflated distribution during the first sampling round (mostly due to lack of vaccination, but can also be a result of a limit to detection in the ELISA), while the 15-month sample distribution appears more Gaussian.

Another important challenge is related to the estimation and adjustment of correlation. We have alluded to the site-level random effects that adjust for correlation within sites, but we also must account for correlation due to repeated measures. Although this is relatively straightforward from a statistical point of view, the small number of sites may require specifying them as fixed effects or using Bayesian methods to improve inference. It is also important to note that parameterization of the model is difficult given the variability in data such as age at sample collection and age at vaccination. We have explored several creative approaches to parameterize variables that describe the impact of hypothesized effects on vaccine response.

This was the first study examining these relations; thus, reasonable effect sizes to detect factors related to vaccine response were not readily available. In addition, because the goal of this project was observational, we estimated that 1600 children pooled across 8 sites were sufficient.

DISCUSSION

Factors that modulate the response to vaccines are diverse and complex. MAL-ED, as a longitudinal birth cohort study harmoniously performed at 8 diverse sites with detailed study of enteropathogen exposure, gut inflammation and permeability, and nutritional indices, will offer important insights into the prevalence and extent of hyporesponsiveness to the principal oral and parenteral vaccines. Collection of the vaccine data, as well as data regarding the determinants (nutrition, breastfeeding practices, socioeconomic factors) were obtained using common protocols and thereby were fundamentally comparable across sites and will greatly add to the strength of the analysis of factors underlying differences in vaccine responsiveness.

The vaccination exposures of the MAL-ED cohort varied significantly in terms of vaccination schedules, vaccines used, and the number and the timing of doses. This heterogeneity was unavoidable given the observational nature of the study and the different geographic locations of the study sites. For example, in some countries, OPV was given at birth as a part of EPI, whereas in others, administration was started at 6 weeks or 2 months of age. Additionally, OPV was administered as part of national and subnational immunization campaigns (eg, India and Pakistan and others). The increasing number of OPV doses may lead to higher seroconversion rates, as has been noted in Jordan [34]. Nonetheless, given the rigor of data collection and the quality of the testing, the MAL-ED study should help elucidate biologically relevant links between nutrition, infection, and the immune response.

The innate and adaptive immune responses to vaccines have shown significant variation between populations [35]; therefore, responses ideally should be further evaluated based on genetic background [36]. In addition, analytic methods must take into account the effect of microbial exposure, nutrition, gut function, and the microbiome on the developing immune system [37, 38] and on its response to specific antigens. In terms of microbial exposure, distinguishing responses induced by natural infection rather than vaccination posed an additional challenge in the evaluation of some vaccines, such as rotavirus or pertussis. Although possible immune modulatory effects of the microbiome have been proposed [9], the role of a functional gut in the quality of immune response has not been evaluated for orally delivered vaccines prior to the initiation of this study.

It is challenging to draw conclusions from single-site studies using a variety of different assays as shown in earlier studies. A study addressing anti–measles antibody production showed a significant delay in anti–measles antibody production in malnourished children compared with well-nourished children [39]; however, the malnourished children caught up over time. Additionally, an observational study in Ecuador found low mean IgG and IgM antibody titers to tetanus toxoid in stunted children, but there was no difference in antibodies to diphtheria toxin [3], whereas a small study in Egypt described low diphtheria toxoid antitoxin levels in severely malnourished children assessed 21 days after vaccination [40]. The use of multiple assay formats complicates the comparison of these studies in different settings. The MAL-ED study design created a scenario where it is possible to compare antibody responses across sites in a reliable way. Other factors adding to the level of complexity of the MAL-ED data included prior exposure to infectious agents and the influence of maternal antibodies. Previous studies have shown that prior exposure to influenza and measles vaccines negatively influences vaccine response [41, 42]. Furthermore, the presence or absence of maternal antibodies in young children may affect immune response to both parenteral and oral vaccines [43, 44]. However, because the data reflect the complexity of the real-world setting of intense exposure to pathogens, the conclusions reached are highly relevant to policy and delivery program decisions.

CONCLUSIONS

The complex data sets of the MAL-ED study, which included repeated measures of anthropometry, enteric infections, intestinal function, and antibody estimations to multiple vaccines at 2 time points, create the potential for data that can, for the first time, conduct cross-country comparisons of immune response to oral and parenteral vaccines. These data could potentially clarify associations between host and environmental factors that influence the development and duration of a protective vaccine response, especially relevant in the setting of a global program to eradicate poliomyelitis. The MAL-ED study is critical to developing mechanistic insights, which may further the understanding of the biology of vaccination and lead to the design of better vaccines and/or vaccination strategies.

Supplementary Data

Supplementary materials are available at Clinical Infectious Diseases online (http://cid.oxfordjournals.org). Supplementary materials consist of data provided by the author that are published to benefit the reader. The posted materials are not copyedited. The contents of all supplementary data are the sole responsibility of the authors. Questions or messages regarding errors should be addressed to the author.

Supplementary Data

Notes

Acknowledgments. The authors thank the staff and participants of the MAL-ED Network for their important contributions.

Financial support. The Etiology, Risk Factors and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health and Development Project (MAL-ED) is carried out as a collaborative project supported by the Bill & Melinda Gates Foundation, the Foundation for the National Institutes of Health, and the National Institutes of Health, Fogarty International Center.

Supplement sponsorship. This article appeared as part of the supplement “The Malnutrition and Enteric Disease Study (MAL-ED): Understanding the Consequences for Child Health and Development,” sponsored by the National Institutes of Health and the Foundation for the National Institutes of Health.

Potential conflicts of interest. All authors: No potential conflicts of interest.

All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

References

  • 1.Myaux J, Unicomb L, Besser R, et al. Effect of diarrhea on the humoral response to oral polio vaccination. Pediatr Infect Dis J. 1996;15:204–9. doi: 10.1097/00006454-199603000-00006. [DOI] [PubMed] [Google Scholar]
  • 2.Migasena S, Simasathien S, Samakoses R, et al. Adverse impact of infections on antibody responses to measles vaccination. Vaccine. 1998;16:647–52. doi: 10.1016/s0264-410x(97)00229-6. [DOI] [PubMed] [Google Scholar]
  • 3.Brüssow H, Sidoti J, Dirren H, Freire WB. Effect of malnutrition in Ecuadorian children on titers of serum antibodies to various microbial antigens. Clin Diagn Lab Immunol. 1995;2:62–8. doi: 10.1128/cdli.2.1.62-68.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Benn CS, Balde A, George E, et al. Effect of vitamin A supplementation on measles-specific antibody levels in Guinea-Bissau. Lancet. 2002;359:1313–4. doi: 10.1016/S0140-6736(02)08274-0. [DOI] [PubMed] [Google Scholar]
  • 5.Savy M, Edmond K, Fine PE, et al. Landscape analysis of interactions between nutrition and vaccine responses in children. J Nutr. 2009;139:2154S–218S. doi: 10.3945/jn.109.105312. [DOI] [PubMed] [Google Scholar]
  • 6.Poonam P. The biology of oral tolerance and issues related to oral vaccine design. Curr Pharm Des. 2007;13:2001–7. doi: 10.2174/138161207781039814. [DOI] [PubMed] [Google Scholar]
  • 7.Moon SS, Wang Y, Shane AL, et al. Inhibitory effect of breast milk on infectivity of live oral rotavirus vaccines. Pediatr Infect Dis J. 2010;29:919–23. doi: 10.1097/INF.0b013e3181e232ea. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lopman BA, Pitzer VE, Sarkar R, et al. Understanding reduced rotavirus vaccine efficacy in low socio-economic settings. PLoS One. 2012;7:e41720. doi: 10.1371/journal.pone.0041720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Qadri F, Bhuiyan TR, Sack DA, Svennerholm AM. Immune responses and protection in children in developing countries induced by oral vaccines. Vaccine. 2013;31:452–60. doi: 10.1016/j.vaccine.2012.11.012. [DOI] [PubMed] [Google Scholar]
  • 10.Bubnoff AV. A gut response to vaccines. Available at: http://www.iavireport.org/Back-Issues/Pages/IAVI-Report-15%286%29-AGutResponsetoVaccines.aspx. Accessed 15 April 2014.
  • 11.Grassly N, Fraser C, Wenger J, et al. New strategies for the elimination of polio from India. Science. 2006;314:1150–3. doi: 10.1126/science.1130388. [DOI] [PubMed] [Google Scholar]
  • 12.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. 2009;200(suppl 1):S39–48. doi: 10.1086/605035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.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. 2010;376:615–23. doi: 10.1016/S0140-6736(10)60755-6. [DOI] [PubMed] [Google Scholar]
  • 14.Sow SO, Tapia M, Haidara FC, et al. Efficacy of the oral pentavalent rotavirus vaccine in Mali. Vaccine. 2012;30(suppl 1):A71–8. doi: 10.1016/j.vaccine.2011.11.094. [DOI] [PubMed] [Google Scholar]
  • 15.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. 2010;376:606–14. doi: 10.1016/S0140-6736(10)60889-6. [DOI] [PubMed] [Google Scholar]
  • 16.John TJ, Jayabal P. Oral polio vaccination of children in the tropics. I. The poor seroconversion rates and the absence of viral interference. Am J Epidemiol. 1972;96:263–9. doi: 10.1093/oxfordjournals.aje.a121457. [DOI] [PubMed] [Google Scholar]
  • 17.The MAL-ED Network Investigators. The MAL-ED study: a multinational and multidisciplinary approach to understand the relationship between enteric pathogens, malnutrition, gut physiology, physical growth, cognitive development, and immune responses in infants and children up to 2 years of age in resource-poor environments. Clin Infect Dis. 2014;59(suppl 4):S193–206. doi: 10.1093/cid/ciu653. [DOI] [PubMed] [Google Scholar]
  • 18.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. 2014;59(suppl 4):S239–47. doi: 10.1093/cid/ciu457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Yori PP, Lee G, Olórtegui MP, et al. Santa clara de nanay: the MAL-ED cohort in Peru. Clin Infect Dis. 2014;59(suppl 4):S310–6. doi: 10.1093/cid/ciu460. [DOI] [PubMed] [Google Scholar]
  • 20.Shrestha PS, Shrestha SK, Bodhidatta L, et al. Bhaktapur, Nepal: the MAL-ED birth cohort study in Nepal. Clin Infect Dis. 2014;59(suppl 4):S300–3. doi: 10.1093/cid/ciu459. [DOI] [PubMed] [Google Scholar]
  • 21.Turab A, Soofi SB, Ahmed I, Bhatti Z, Zaidi AK, Bhutta ZA. Demographic, socioeconomic and health characteristics of the MAL-ED network study site in rural Pakistan. Clin Infect Dis. 2014;59(suppl 4):S304–9. doi: 10.1093/cid/ciu391. [DOI] [PubMed] [Google Scholar]
  • 22.Bessong PO, Nyathi E, Mahopo C, et al. Development of the Dzimauli community in Vhembe district, Limpopo province of South Africa for the MAL-ED cohort study. Clin Infect Dis. 2014;59(suppl 4):S317–24. doi: 10.1093/cid/ciu418. [DOI] [PubMed] [Google Scholar]
  • 23.John SM, Thomas RJ, Kaki S, et al. Establishment of the MAL-ED birth cohort study site in Vellore, southern India. Clin Infect Dis. 2014;59(suppl 4):S295–9. doi: 10.1093/cid/ciu390. [DOI] [PubMed] [Google Scholar]
  • 24.Mduma ER, Gratz J, Patil C, et al. The etiology, risk factors and interactions of enteric infections and malnutrition and the consequences for child health and development study (MAL-ED): description of the Tanzanian site. Clin Infect Dis. 2014;59(suppl 4):S325–30. doi: 10.1093/cid/ciu439. [DOI] [PubMed] [Google Scholar]
  • 25.Lima AA, Oriá RB, Soares AM, et al. Geography, population, demography, socioeconomic, anthropometry and environmental status the MAL-ED cohort and case-control study sites in Fortaleza, Ceará, Brazil. Clin Infect Dis. 2014;59(suppl 4):S287–94. doi: 10.1093/cid/ciu438. [DOI] [PubMed] [Google Scholar]
  • 26.Ahmed T, Mahfuz M, Islam M, et al. The MAL-ED cohort study in Mirpur, Bangladesh. Clin Infect Dis. 2014;59(suppl 4):S280–6. doi: 10.1093/cid/ciu458. [DOI] [PubMed] [Google Scholar]
  • 27.Richard SA, Barrett L, Guerrant RL, Checkley W, Miller M. Disease surveillance methods used in the 8 site MAL-ED cohort study. Clin Infect Dis. 2014;59(suppl 4):S220–4. doi: 10.1093/cid/ciu435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Gladstone BP, Ramani S, Mukhopadhya I, et al. Protective effect of natural rotavirus infection in an Indian birth cohort. N Engl J Med. 2011;365:337–46. doi: 10.1056/NEJMoa1006261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ward RL, Bernstein DI, Smith VE, et al. Rotavirus immunoglobulin a responses stimulated by each of 3 doses of a quadrivalent human/bovine reassortant rotavirus vaccine. J Infect Dis. 2004;189:2290–3. doi: 10.1086/421248. [DOI] [PubMed] [Google Scholar]
  • 30.Ward RL, Bernstein DI, Shukla R, et al. Protection of adults rechallenged with a human rotavirus. J Infect Dis. 1990;161:440–5. doi: 10.1093/infdis/161.3.440. [DOI] [PubMed] [Google Scholar]
  • 31.Albrecht P, Enterline JC, Boone EJ, Klutch MJ. Poliovirus and polio antibody assay in HEp-2 and Vero cell cultures. J Biol Stand. 1983;11:91–7. doi: 10.1016/s0092-1157(83)80031-6. [DOI] [PubMed] [Google Scholar]
  • 32.Laird NM, Ware JH. Random-effects models for longitudinal data. Biometrics. 1982;38:963–74. [PubMed] [Google Scholar]
  • 33.Clayton D. Generalized linear mixed models. In: Gilks WR, Richardson S, Spiegelhalter DJ, editors. Markov Chain Monte Carlo in Practice. New York City, NY: Chapman and Hall; 1996. pp. 275–301. [Google Scholar]
  • 34.Reichler MR, Kharabsheh S, Rhodes P, et al. Increased immunogenicity of oral poliovirus vaccine administered in mass vaccination campaigns compared with the routine vaccination program in Jordan. J Infect Dis. 1997;175(suppl 1):S198–204. doi: 10.1093/infdis/175.supplement_1.s198. [DOI] [PubMed] [Google Scholar]
  • 35.Kollmann TR. Variation between populations in the innate immune response to vaccine adjuvants. Front Immunol. 2013;4:81. doi: 10.3389/fimmu.2013.00081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Haralambieva IH, Ovsyannikova IG, Pankratz VS, Kennedy RB, Jacobson RM, Poland GA. The genetic basis for interindividual immune response variation to measles vaccine: new understanding and new vaccine approaches. Expert Rev Vaccines. 2013;12:57–70. doi: 10.1586/erv.12.134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kasturi SP, Skountzou I, Albrecht RA, et al. Programming the magnitude and persistence of antibody responses with innate immunity. Nature. 2011;470:543–7. doi: 10.1038/nature09737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, the gut microbiome and the immune system. Nature. 2011;474:327–36. doi: 10.1038/nature10213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Powell GM. Response to live attenuated measles vaccine in children with severe kwashiorkor. Ann Trop Paediatr. 1982;2:143–5. doi: 10.1080/02724936.1982.11748247. [DOI] [PubMed] [Google Scholar]
  • 40.Hafez M, Aref GH, Mehareb SW, et al. Antibody production and complement system in protein energy malnutrition. J Trop Med Hyg. 1977;80:36–9. [PubMed] [Google Scholar]
  • 41.Gans H, DeHovitz R, Forghani B, Beeler J, Maldonado Y, Arvin AM. Measles and mumps vaccination as a model to investigate the developing immune system: passive and active immunity during the first year of life. Vaccine. 2003;21:3398–405. doi: 10.1016/s0264-410x(03)00341-4. [DOI] [PubMed] [Google Scholar]
  • 42.Pulendran B, Oh JZ, Nakaya HI, Ravindran R, Kazmin DA. Immunity to viruses: learning from successful human vaccines. Immunol Rev. 2013;255:243–55. doi: 10.1111/imr.12099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hahn-Zoric M, Carlsson B, Jalil F, Mellander L, Germanier R, Hanson LA. The influence on the secretory IgA antibody levels in lactating women of oral typhoid and parenteral cholera vaccines given alone or in combination. Scand J Infect Dis. 1989;21:421–6. doi: 10.3109/00365548909167447. [DOI] [PubMed] [Google Scholar]
  • 44.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. 2013;32:863–70. doi: 10.1097/INF.0b013e318290646d. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Data

Articles from Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America are provided here courtesy of Oxford University Press

RESOURCES