Abstract

Oral vaccines have several advantages compared with parenteral administration: they can be relatively cheap to produce in high quantities, easier to administer, and induce intestinal mucosal immunity that can protect against infection. These characteristics have led to successful use of oral vaccines against rotavirus, polio, and cholera. Unfortunately, oral vaccines for all three diseases have demonstrated lower performance in the highest-burden settings where they are most needed. Rotavirus vaccines are estimated to have >85% effectiveness against hospitalization in children <12 months in countries with low child mortality, but only ~65% effectiveness in countries with high child mortality. Similarly, oral polio vaccines have lower immunogenicity in developing country settings compared with high-resource settings. Data are more limited for oral cholera vaccines, but suggest lower titers among children compared with adults, and, for some vaccines, lower efficacy in endemic settings compared with non-endemic settings. These disparities are likely multifactorial, and available evidence suggests a role for maternal factors (e.g. transplacental antibodies, breastmilk), host factors (e.g. genetic polymorphisms—with the best evidence for rotavirus—or previous infection), and environmental factors (e.g. gut microbiome, co-infections). Overall, these data highlight the rather ambiguous and often contradictory nature of evidence on factors affecting oral vaccine response, cautioning against broad extrapolation of outcomes based on one population or one vaccine type. Meaningful impact on performance of oral vaccines will likely only be possible with a suite of interventions, given the complex and multifactorial nature of the problem, and the degree to which contributing factors are intertwined.

Introduction

Orally administered vaccines have been in use since the late 1950s, when the live, attenuated Sabin oral polio vaccine (OPV) was first introduced for mass vaccination campaigns [1]. Since then, oral vaccines have been developed for other diseases transmitted via the fecal-oral route, most notably rotavirus and cholera [2]. Oral vaccines have several advantages compared with parenteral administration: they can be cheaper to produce in high quantities (e.g. OPV is <1/5 the cost of inactivated polio vaccine), easier to administer, and have the ability to induce intestinal mucosal immunity that can protect against infection [2, 3]. Indeed, the drastic reduction in poliomyelitis cases and poliovirus circulation globally since the 1950s is largely attributed to widespread use of OPV in routine immunization programs and outbreak response activities [4]. Rotavirus vaccines have also demonstrated success in reducing global burden of disease, and introduction of rotavirus vaccination into routine immunization programs is estimated to have prevented 139 000 under-5 deaths during 2006–19, before introduction in large countries with high burdens such as India, Pakistan, Nigeria, and DRC [5]. In contrast to rotavirus and polio vaccines, which are given in many countries as standard preventive care for infants and young children, oral cholera vaccines (OCVs) are given only in campaign modality (i.e. via mass administration during discrete periods in high-risk areas). OCVs are used primarily in outbreak settings and in endemic countries, and have the ability to induce herd protection, stopping outbreaks, and averting cases [6, 7]. Despite these successes, oral vaccines for all three diseases have demonstrated lower performance in the highest-burden settings where they are most needed. In the present review, we will introduce these three pathogens and their vaccines, summarize vaccine performance disparities, and present evidence for contributing factors.

Pathogen overview

Rotavirus

Rotavirus is a leading cause of severe acute gastroenteritis in children under the age of 5 years. Prior to the introduction of live-attenuated oral rotavirus vaccines, nearly every child experienced a rotavirus infection by the age of 5 years, leading to over 2 million hospitalizations and an estimated 528 000 deaths each year [8, 9]. Vaccine introduction in over 120 countries since 2006 has resulted in substantial reductions in the burden of rotavirus-associated morbidity and mortality. However, an estimated 108 470 deaths were still attributable to rotavirus in 2021, with nearly all occurring in low and middle-income countries (LMICs) and over 70% occurring in the WHO African region [10].

Rotaviruses are non-enveloped, double-stranded (ds) RNA viruses within the family Sedoreoviridae. The 11 segments of dsRNA code for six structural proteins (viral proteins, VP) and 6 non-structural proteins. Traditionally, rotavirus is classified into G- and P-types based on sequence differences in the outer capsid proteins VP7 and VP4, respectively. Recently, a more comprehensive typing system based on the whole genome has been established. The first human rotavirus was visualized in thin-section electron micrographs of duodenal biopsies from children with gastroenteritis in 1973 [11]. Shortly thereafter, the virus was identified in stool samples of children with diarrhea [12] and subsequently recognized as the major cause of pediatric gastroenteritis worldwide. The spectrum of clinical presentations is broad, ranging from asymptomatic infections and mild diarrhea to severe, life-threatening gastroenteritis that may require hospitalization. Dehydration, electrolyte imbalance, and death may occur if adequate and timely fluid replacement is not provided. Rotavirus pathogenesis is multifactorial and involves both malabsorptive and secretory mechanisms of diarrhea [13–15]. Rotavirus infection also results in the secretion of serotonin that can alter gut motility and activate vomiting centers in the brain [16]. Rotavirus infection and disease are not limited to the gastrointestinal tract, and extraintestinal complications involving the central nervous system can occur [17].

Poliovirus

In the decades before polio vaccine introduction, polioviruses caused hundreds of thousands of children to become paralyzed each year. Due to a coordinated global effort to vaccinate children, cases of paralysis have dropped by over 99%, and two out of three wild poliovirus (WPV) serotypes have been eradicated [1]. Despite this tremendous progress, endemic WPV1 circulation continues in Afghanistan and Pakistan in 2024, while other geographies have experienced outbreaks due to reverted vaccine-derived virus [18]. Several factors contribute to the complexity of polio eradication, including viral and vaccine characteristics in addition to conflict settings and cultural factors that complicate vaccination efforts in certain geographies.

Polioviruses are non-enveloped, single-stranded RNA viruses of the Picornaviridae family, existing in three serotypes (1, 2, 3). The poliovirus mRNA is translated as one polypeptide which undergoes protease cleavage into 10 viral proteins. Poliovirus transmission occurs primarily through the fecal-oral route in lower-sanitation settings, with the oral-oral route gaining primacy in higher-sanitation settings [19]. Viruses replicate initially in the oropharyngeal and intestinal mucosa and are shed in the feces. In the majority of infections, viral replication is generally confined to the gastrointestinal tract and a transient viremia, and symptoms are either non-existent or non-specific (e.g. fever, malaise) [20]. However, in rare cases (~1–2%), a prolonged secondary viremia can provide the opportunity for the virus to infect the central nervous system via retrograde axonal transport or by directly crossing the blood-brain barrier [20, 21]. Infection of motor neurons can lead to limb paralysis or even death, when breathing muscles are affected. No cure is available, and treatment of paralysis is limited to physical therapy or mechanical ventilation where indicated [19].

Cholera

In 2015, annual cholera burden was estimated as 1.3–4 million cases per year and 21 000–143 000 related deaths [22]. With the exception of sporadic cases associated with ingestion of uncooked shellfish in coastal areas in a few high-income countries (HICs), cholera infection and disease are almost exclusively associated with extreme poverty, lack of access to clean water, and inadequate sanitation [23]. Climate change together with other factors (e.g. political conflict, forced migration, and economic and social disruptions induced by the COVID-19 pandemic) have further propagated cholera outbreaks since 2020, and as of 31 January 2024, a new surge in infections had been reported in at least 30 countries [24]. Notably, several reporting countries were either non-endemic countries (e.g. Lebanon and Syria) or had not been affected by cholera for a long period of time (e.g. Haiti and Dominican Republic). In January 2023, WHO raised the global cholera crisis to a grade 3 emergency, the highest level of the grading system [25].

Cholera, a diarrheal disease caused by Vibrio cholerae, is spread from person to person by the fecal-oral route or through contaminated food or water. In its most severe form, affected individuals have voluminous watery stools which can lead to dehydration and death within hours, even in otherwise healthy adults, if prompt rehydration is not initiated [26]. After an infectious dose (105–108 organisms [27]) is ingested, this non-invasive pathogen colonizes the small intestine and causes illness principally through excretion of a cholera toxin, CTX, that leads to a secretory diarrhea [28]. There are two serogroups associated with severe human disease, O1 and O139; 99% of disease globally is attributed to the O1 serogroup, which has two serotypes, Inaba and Ogawa [29]. Although serologically distinguishable, because of a high degree of similarity in their outer surface polysaccharide, the O-specific polysaccharide (OSP), they are highly cross-reactive and indistinguishable in disease caused. Both natural history studies and human challenge studies have confirmed they are also cross-protective (i.e. infection with either confers a period of homologous and heterologous protection) [30]. Serum vibriocidal antibody has long been recognized as a correlate of protection against cholera, with OSP as the most influential antigen [31].

Vaccines and vaccine performance

Rotavirus

Currently, four oral rotavirus vaccines (Rotarix, RotaTeq, Rotavac, and Rotasiil) are prequalified by the World Health Organization (WHO) for global use, meaning that quality, safety, and efficacy data have been reviewed by WHO, and these products have been deemed acceptable for global procurement. Three additional vaccines are nationally licensed in Vietnam and China (Rotavin-M1 in Vietnam and Lanzhou Lamb Rotavirus [LLR] and LLR3 vaccines in China) (Table 1). All rotavirus vaccines are based on live, attenuated strains; RotaTeq and Rotasiil are reassorted bovine-human viruses, Rotarix and Rotavin are based on human strains attenuated via passage in Vero cells, Rotavac is based on a naturally attenuated bovine-human neonatal strain, LLR is based on a lamb strain, and LLR3 is based on a human-lamb reassortant strain [32, 33]. Several additional vaccines are in preclinical and clinical development, including an oral vaccine to be delivered on a neonatal schedule [33]. Despite inclusion in the national immunization programs of over 120 countries, global rotavirus vaccine coverage in 2023 was only 55%, with averages of 59%, 66%, and 58% for high-, lower middle-, and low-income countries, respectively (Fig. 1) [34].

Table 1:

licensed oral rotavirus vaccines and their characteristics, 2024

VaccineManufacturerGenotype(s)FormulationWHO-prequalification (year)Number of dosesSchedule
RotaTeqMerckG1, G2, G3, G4, P [8]LiquidYes (2008)3Dose 1 starting at 6 to 12 weeks of age; subsequent doses at 4- to 10-week intervals; dose 3 should not be given after 32 weeks of age
RotarixGSKG1P [8]LiquidYes (2009)2Dose 1 starting 6 weeks; dose 2 at least 4 weeks after dose 1 and up to 24 weeks of age
RotavacBharat Biotech International LimitedG9P [11]Liquid (frozen and non-frozen)Yes (2018)3Dose 1 starting at 6 weeks of age; subsequent doses at 4 week intervals; dose 3 should not be given after 34 weeks of age
RotasiilSerum Institute of IndiaG1, G2, G3, G4, G9Liquid, LyophilizedYes (2018)3Dose 1 starting at 6 weeks of age; subsequent doses at 4-week intervals; dose 3 should not be given after 34 weeks of age
Rotavin-M1PolyvacG1P [8]Liquid (frozen and non-frozen)No (NA)2Dose 1 starting at 6 weeks of age; second dose after 60 days and up to 6 months of age
Lanzhou Lamb RotavirusLanzhou Institute of Biological ProductsG10P [15]LiquidNo (NA)3Dose 1 starting at 2 months of age; annual dose up to 3 years of age
Lanzhou Lamb Rotavirus3Lanzhou Institute of Biological ProductsG2, G3, G4LiquidNo (NA)3Dose 1 starting at 2 months of age; annual dose up to 3 years of age
VaccineManufacturerGenotype(s)FormulationWHO-prequalification (year)Number of dosesSchedule
RotaTeqMerckG1, G2, G3, G4, P [8]LiquidYes (2008)3Dose 1 starting at 6 to 12 weeks of age; subsequent doses at 4- to 10-week intervals; dose 3 should not be given after 32 weeks of age
RotarixGSKG1P [8]LiquidYes (2009)2Dose 1 starting 6 weeks; dose 2 at least 4 weeks after dose 1 and up to 24 weeks of age
RotavacBharat Biotech International LimitedG9P [11]Liquid (frozen and non-frozen)Yes (2018)3Dose 1 starting at 6 weeks of age; subsequent doses at 4 week intervals; dose 3 should not be given after 34 weeks of age
RotasiilSerum Institute of IndiaG1, G2, G3, G4, G9Liquid, LyophilizedYes (2018)3Dose 1 starting at 6 weeks of age; subsequent doses at 4-week intervals; dose 3 should not be given after 34 weeks of age
Rotavin-M1PolyvacG1P [8]Liquid (frozen and non-frozen)No (NA)2Dose 1 starting at 6 weeks of age; second dose after 60 days and up to 6 months of age
Lanzhou Lamb RotavirusLanzhou Institute of Biological ProductsG10P [15]LiquidNo (NA)3Dose 1 starting at 2 months of age; annual dose up to 3 years of age
Lanzhou Lamb Rotavirus3Lanzhou Institute of Biological ProductsG2, G3, G4LiquidNo (NA)3Dose 1 starting at 2 months of age; annual dose up to 3 years of age
Table 1:

licensed oral rotavirus vaccines and their characteristics, 2024

VaccineManufacturerGenotype(s)FormulationWHO-prequalification (year)Number of dosesSchedule
RotaTeqMerckG1, G2, G3, G4, P [8]LiquidYes (2008)3Dose 1 starting at 6 to 12 weeks of age; subsequent doses at 4- to 10-week intervals; dose 3 should not be given after 32 weeks of age
RotarixGSKG1P [8]LiquidYes (2009)2Dose 1 starting 6 weeks; dose 2 at least 4 weeks after dose 1 and up to 24 weeks of age
RotavacBharat Biotech International LimitedG9P [11]Liquid (frozen and non-frozen)Yes (2018)3Dose 1 starting at 6 weeks of age; subsequent doses at 4 week intervals; dose 3 should not be given after 34 weeks of age
RotasiilSerum Institute of IndiaG1, G2, G3, G4, G9Liquid, LyophilizedYes (2018)3Dose 1 starting at 6 weeks of age; subsequent doses at 4-week intervals; dose 3 should not be given after 34 weeks of age
Rotavin-M1PolyvacG1P [8]Liquid (frozen and non-frozen)No (NA)2Dose 1 starting at 6 weeks of age; second dose after 60 days and up to 6 months of age
Lanzhou Lamb RotavirusLanzhou Institute of Biological ProductsG10P [15]LiquidNo (NA)3Dose 1 starting at 2 months of age; annual dose up to 3 years of age
Lanzhou Lamb Rotavirus3Lanzhou Institute of Biological ProductsG2, G3, G4LiquidNo (NA)3Dose 1 starting at 2 months of age; annual dose up to 3 years of age
VaccineManufacturerGenotype(s)FormulationWHO-prequalification (year)Number of dosesSchedule
RotaTeqMerckG1, G2, G3, G4, P [8]LiquidYes (2008)3Dose 1 starting at 6 to 12 weeks of age; subsequent doses at 4- to 10-week intervals; dose 3 should not be given after 32 weeks of age
RotarixGSKG1P [8]LiquidYes (2009)2Dose 1 starting 6 weeks; dose 2 at least 4 weeks after dose 1 and up to 24 weeks of age
RotavacBharat Biotech International LimitedG9P [11]Liquid (frozen and non-frozen)Yes (2018)3Dose 1 starting at 6 weeks of age; subsequent doses at 4 week intervals; dose 3 should not be given after 34 weeks of age
RotasiilSerum Institute of IndiaG1, G2, G3, G4, G9Liquid, LyophilizedYes (2018)3Dose 1 starting at 6 weeks of age; subsequent doses at 4-week intervals; dose 3 should not be given after 34 weeks of age
Rotavin-M1PolyvacG1P [8]Liquid (frozen and non-frozen)No (NA)2Dose 1 starting at 6 weeks of age; second dose after 60 days and up to 6 months of age
Lanzhou Lamb RotavirusLanzhou Institute of Biological ProductsG10P [15]LiquidNo (NA)3Dose 1 starting at 2 months of age; annual dose up to 3 years of age
Lanzhou Lamb Rotavirus3Lanzhou Institute of Biological ProductsG2, G3, G4LiquidNo (NA)3Dose 1 starting at 2 months of age; annual dose up to 3 years of age
 Two maps of vaccine coverage. The first map shows the percentage of children, by country, who have received a complete series of rotavirus vaccine. The second map shows the percentage of children, by country, who have received at least 3 doses of polio vaccine. Countries are shaded to represent the percent coverage achieved in each country.
Figure 1:

rotavirus vaccine complete-series coverage (A) and polio vaccine 3-dose coverage (B) among children at 1 year of age, by country, 2023

Rotavirus vaccines are estimated to have averted >139 000 deaths [5] since their introduction in 2006, and >500 000 hospitalizations in 2019 alone [35], among children <5. Since 2019, several countries with large birth cohorts (DRC, Nigeria, India, Indonesia) have introduced rotavirus vaccines into their national schedules [34], suggesting that impact might be even greater if measured in 2024. Despite this tremendous success, there are geographic gaps in vaccine performance. In HICs such as the USA and Finland, where initial clinical trials of Rotarix and RotaTeq took place, efficacy of rotavirus vaccines against severe rotavirus gastroenteritis in the first year of life was >90% [36, 37]. However, later clinical trials conducted in sub-Saharan Africa and Asia, including trials of the newer vaccines, Rotasiil and Rotavac, demonstrated markedly lower efficacy of <70% (and even <40%) [38–43].

Performance disparities are also evident in real-world usage. One meta-analysis estimated Rotarix vaccine effectiveness among children <12 months of age to be 86% (95% CI: 81–90%) in countries with low child mortality, 77% (66–85%) in countries with medium child mortality, and 63% (54–70%) in countries with high child mortality; for RotaTeq, vaccine effectiveness was 86% (76–92%) in countries with low child mortality and 66% (51–76%) in countries with high child mortality [44]. Among children in their second year of life, Rotarix vaccine effectiveness was again estimated as lower in countries with high child mortality, compared to countries with low child mortality; sparse data precluded estimates for RotaTeq. Evaluation of vaccine impact (decrease in rotavirus-associated hospitalizations following rotavirus vaccine introduction) also showed differences by child mortality stratum, with the greatest impact in low-mortality settings (78% reduction among children <12 months of age; inter-quartile range [IQR]: 65–85%), and lower impact in higher-mortality settings (55%; 41–64%) [45]. Among all children <5 years, impact estimates were somewhat lower, and disparities were somewhat lessened (66% [IQR 49–76%] reduction in the low-mortality setting vs. 50% [IQR, 41–65%] in the highest-mortality setting). Despite the fact that higher-mortality settings may show a lower estimated impact of rotavirus vaccination when measured via percent reduction, their high baseline burden can result in a greater number of hospitalizations and deaths averted when compared with lower-mortality settings [38].

Due to their more recent introduction, effectiveness and impact data are not readily available for Rotavac and Rotasiil. However, given their similarities to existing vaccines, it is expected that performance differences would be similar as with efficacy estimates. Indeed, preliminary effectiveness data from studies in India suggest performance in line with above estimates for high- and medium-child mortality settings (unpublished data).

Poliovirus

Two different types of vaccines are available to prevent poliomyelitis: the parenterally administered Salk inactivated polio vaccine, and the live, attenuated oral poliovirus vaccine (OPV; developed via cell culture passaging) [19]. While a complete series of either vaccine provides a high level of protection against paralysis, OPV is preferred for routine immunization in high-risk settings and for outbreak response due to its low cost, ease of use, and superior ability to induce intestinal mucosal immunity—required to interrupt transmission. Unfortunately, as the live viruses in first-generation (Sabin-strain) OPV replicate in the gut, their inherent genetic instability can cause them to mutate and recombine with other enteroviruses [46]. In extremely rare cases, vaccine virus that has mutated in the intestines can cause paralysis in a vaccinated child or contact, termed Vaccine-Associated Paralytic Polio (VAPP). Vaccine and vaccine-like viruses that are shed in the feces can also infect unprotected children; in areas of low immunization coverage, long transmission chains can allow mutated viruses to regain neurovirulence. These circulating vaccine-derived polioviruses (cVDPVs) cause outbreaks which must in turn be responded to using OPV, potentially leading to new cVDPVs [46]. In 2021, a new, more genetically stable vaccine was rolled out: novel oral poliovirus vaccine, serotype 2 (nOPV2), constructed with unique attenuating mutations that reduce the likelihood of harmful mutations and recombinations that allow the virus to rapidly regain neurovirulence [47]. Type 2 was prioritized for new vaccine development due to its higher risk of generating cVDPVs, compared with other serotypes; novel OPVs for type 1 and type 3 virus are also under development [48]. Evidence from field use to date has borne out nOPV2’s dramatically lower likelihood of reversion as well as its comparable effectiveness in closing outbreaks, compared with its predecessor Sabin OPV [49].

Since the 1970s, researchers have observed global variation in OPV performance [50, 51]. Patriarca and colleagues contrasted low prevalence of serum neutralizing antibody activity after 3 doses of trivalent OPV in ‘developing’ countries with higher prevalence in industrialized countries (e.g. US, Canada, Europe), observing a weighted average of 73%, 90%, and 70% prevalence for types 1, 2, 3, respectively. By contrast, a study in the US demonstrated >97% seroprevalence for all three types after three doses of tOPV [52] (Fig. 2). The authors identified interference from Sabin 2 and other enterovirus infections as key drivers of impaired OPV immunogenicity in these settings. Another review of serum neutralizing antibody activity after monovalent OPV administration identified similar geographic differences, with type 1 seroprevalence after one dose ranging from 53% in Uganda to 100% in USSR and the Netherlands, type 2 from 77% in Mexico to 100% in USSR, and type 3 from 52% in Brazil to 100% in the Netherlands [54]. The authors note differences between studies in sites in temperate compared to non-temperate latitudes, with lower median seroconversion rates in non-temperate (81%, 89%, 72%) versus temperate (95%, 98%, 94%) settings (Fig. 3). A later study in Mozambique (non-temperate) also found relatively lower seroconversion following one dose mOPV2 (61%) [55]. Supplemental Figure 1 also shows differences by country income strata. More recent studies of nOPV2 have shown similar differences, with a study in the Gambia (classified as low-income) finding significantly lower two-dose seroconversion in infants and children (67% and 74%, respectively) [56], compared with results from earlier studies conducted in high- and middle-income countries (98–100% in Panama, 96% in Dominican Republic, 86–90% in Bangladesh) [57–60].

A three-panel plot showing per-dose seroconversion by vaccine (color) and serotype (panel), plotted with seroconversion on the y axis and under-five mortality on the x axis. An inverse relationship can be seen between seroconversion and under-five mortality particularly for types 1 and 3.
Figure 2:

seroconversion rates in children administered tOPV (from Patriarca et al. [50]), bOPV (from Macklin et al. [53]), or mOPV1, 2, 3 (Caceres et al. [54]) plotted by UNICEF estimated under-5 mortality in country and year of study (year of publication used if year of study not reported). Seroconversion adjusted to single dose conversion as 1 − (1 − S/N)^(1/D), where S is the number seroconverting, N is the number studied, and D is the number of doses administered

Table 3:

comparison of seroconversion rate and GMT following two doses of Shanchol among adults and children under 5 years of age in cholera-endemic countries

Adults (18 years and above)Children (1–5 years)
O1 InabaO1 OgawaO1 InabaO1 Ogawa
nSeroconversion rate [95% CI]GMT [95% CI]nSeroconversion rate [95% CI]GMT [95% CI]nSeroconversion rate [95% CI]GMT [95% CI]nSeroconversion rate [95% CI]GMT [95% CI]
IndiaaBaseline37-186 [93, 370]34-377 [211, 673]12-24 [6, 100]7-20 [1, 377]
Post second dose46% [31, 62]1003 [763, 1319]41% [26, 58]1204 [858, 1690]92% [65, 99]604 [311, 1172]86% [49, 97]525 [116, 2384]
BangladeshbBaseline416-44 [38, 51]416-60 [52, 69]214-19 [15, 23]214-17 [14, 21]
Post second dose83% [79, 86]435 [390, 485]73% [69, 77]375 [354, 440]86% [81, 90]354 [288, 431]85% [79, 89]263 [215, 323]
PhilippinescBaseline376-36 [35,37]376-74 [73, 75]74-4 [4, 5]74-5 [4, 5]
Post second dose76% [72, 81]732 [727 737]74% [69, 78]962 [957, 967]84% [73, 91]159 [147 171]86% [77, 93]283 [271, 295]
NepaldBaseline312-24 [18, 32]312-52 [40, 70]238-4 [3, 5]238-4 [3, 6]
Post second dose79% [75, 83]620 [523, 735]75% [71, 78]1036 [896, 1197]94% [92, 97]398 [319, 496]94% [91, 97]604 [486, 725]
EthiopiaeBaseline53-17 [9, 32]37-24 [10, 55]17-3 [1, 11]16-3 [1, 9]
Post second dose81% [69, 89]254 [160, 403]70% [54, 83]306 [160, 587]53% [31, 74]15 [4, 51]75% [51, 90]73 [25, 217]
Adults (18 years and above)Children (1–5 years)
O1 InabaO1 OgawaO1 InabaO1 Ogawa
nSeroconversion rate [95% CI]GMT [95% CI]nSeroconversion rate [95% CI]GMT [95% CI]nSeroconversion rate [95% CI]GMT [95% CI]nSeroconversion rate [95% CI]GMT [95% CI]
IndiaaBaseline37-186 [93, 370]34-377 [211, 673]12-24 [6, 100]7-20 [1, 377]
Post second dose46% [31, 62]1003 [763, 1319]41% [26, 58]1204 [858, 1690]92% [65, 99]604 [311, 1172]86% [49, 97]525 [116, 2384]
BangladeshbBaseline416-44 [38, 51]416-60 [52, 69]214-19 [15, 23]214-17 [14, 21]
Post second dose83% [79, 86]435 [390, 485]73% [69, 77]375 [354, 440]86% [81, 90]354 [288, 431]85% [79, 89]263 [215, 323]
PhilippinescBaseline376-36 [35,37]376-74 [73, 75]74-4 [4, 5]74-5 [4, 5]
Post second dose76% [72, 81]732 [727 737]74% [69, 78]962 [957, 967]84% [73, 91]159 [147 171]86% [77, 93]283 [271, 295]
NepaldBaseline312-24 [18, 32]312-52 [40, 70]238-4 [3, 5]238-4 [3, 6]
Post second dose79% [75, 83]620 [523, 735]75% [71, 78]1036 [896, 1197]94% [92, 97]398 [319, 496]94% [91, 97]604 [486, 725]
EthiopiaeBaseline53-17 [9, 32]37-24 [10, 55]17-3 [1, 11]16-3 [1, 9]
Post second dose81% [69, 89]254 [160, 403]70% [54, 83]306 [160, 587]53% [31, 74]15 [4, 51]75% [51, 90]73 [25, 217]

Abbreviations: CI: confidence interval; GMT: geometric mean titers; n: number of participants analyzed.

The seroconversion rate is the proportion of participants with at least 4-fold rise vibriocidal titers against Vibrio cholerae O1 Inaba and O1 Ogawa from baseline to post second dose. Vibriocidal assay performed in different laboratories for each trial using similar methods.

aShanchol trial, post dose 2 Shanchol only.

bCholvax trial, post dose 2 Shanchol only.

cEuvichol trial, post dose 2 Shanchol only.

dEuvichol-S trial, post dose 2 Shanchol only.

eShanchol trial, post dose 2 Shanchol only.

Table 3:

comparison of seroconversion rate and GMT following two doses of Shanchol among adults and children under 5 years of age in cholera-endemic countries

Adults (18 years and above)Children (1–5 years)
O1 InabaO1 OgawaO1 InabaO1 Ogawa
nSeroconversion rate [95% CI]GMT [95% CI]nSeroconversion rate [95% CI]GMT [95% CI]nSeroconversion rate [95% CI]GMT [95% CI]nSeroconversion rate [95% CI]GMT [95% CI]
IndiaaBaseline37-186 [93, 370]34-377 [211, 673]12-24 [6, 100]7-20 [1, 377]
Post second dose46% [31, 62]1003 [763, 1319]41% [26, 58]1204 [858, 1690]92% [65, 99]604 [311, 1172]86% [49, 97]525 [116, 2384]
BangladeshbBaseline416-44 [38, 51]416-60 [52, 69]214-19 [15, 23]214-17 [14, 21]
Post second dose83% [79, 86]435 [390, 485]73% [69, 77]375 [354, 440]86% [81, 90]354 [288, 431]85% [79, 89]263 [215, 323]
PhilippinescBaseline376-36 [35,37]376-74 [73, 75]74-4 [4, 5]74-5 [4, 5]
Post second dose76% [72, 81]732 [727 737]74% [69, 78]962 [957, 967]84% [73, 91]159 [147 171]86% [77, 93]283 [271, 295]
NepaldBaseline312-24 [18, 32]312-52 [40, 70]238-4 [3, 5]238-4 [3, 6]
Post second dose79% [75, 83]620 [523, 735]75% [71, 78]1036 [896, 1197]94% [92, 97]398 [319, 496]94% [91, 97]604 [486, 725]
EthiopiaeBaseline53-17 [9, 32]37-24 [10, 55]17-3 [1, 11]16-3 [1, 9]
Post second dose81% [69, 89]254 [160, 403]70% [54, 83]306 [160, 587]53% [31, 74]15 [4, 51]75% [51, 90]73 [25, 217]
Adults (18 years and above)Children (1–5 years)
O1 InabaO1 OgawaO1 InabaO1 Ogawa
nSeroconversion rate [95% CI]GMT [95% CI]nSeroconversion rate [95% CI]GMT [95% CI]nSeroconversion rate [95% CI]GMT [95% CI]nSeroconversion rate [95% CI]GMT [95% CI]
IndiaaBaseline37-186 [93, 370]34-377 [211, 673]12-24 [6, 100]7-20 [1, 377]
Post second dose46% [31, 62]1003 [763, 1319]41% [26, 58]1204 [858, 1690]92% [65, 99]604 [311, 1172]86% [49, 97]525 [116, 2384]
BangladeshbBaseline416-44 [38, 51]416-60 [52, 69]214-19 [15, 23]214-17 [14, 21]
Post second dose83% [79, 86]435 [390, 485]73% [69, 77]375 [354, 440]86% [81, 90]354 [288, 431]85% [79, 89]263 [215, 323]
PhilippinescBaseline376-36 [35,37]376-74 [73, 75]74-4 [4, 5]74-5 [4, 5]
Post second dose76% [72, 81]732 [727 737]74% [69, 78]962 [957, 967]84% [73, 91]159 [147 171]86% [77, 93]283 [271, 295]
NepaldBaseline312-24 [18, 32]312-52 [40, 70]238-4 [3, 5]238-4 [3, 6]
Post second dose79% [75, 83]620 [523, 735]75% [71, 78]1036 [896, 1197]94% [92, 97]398 [319, 496]94% [91, 97]604 [486, 725]
EthiopiaeBaseline53-17 [9, 32]37-24 [10, 55]17-3 [1, 11]16-3 [1, 9]
Post second dose81% [69, 89]254 [160, 403]70% [54, 83]306 [160, 587]53% [31, 74]15 [4, 51]75% [51, 90]73 [25, 217]

Abbreviations: CI: confidence interval; GMT: geometric mean titers; n: number of participants analyzed.

The seroconversion rate is the proportion of participants with at least 4-fold rise vibriocidal titers against Vibrio cholerae O1 Inaba and O1 Ogawa from baseline to post second dose. Vibriocidal assay performed in different laboratories for each trial using similar methods.

aShanchol trial, post dose 2 Shanchol only.

bCholvax trial, post dose 2 Shanchol only.

cEuvichol trial, post dose 2 Shanchol only.

dEuvichol-S trial, post dose 2 Shanchol only.

eShanchol trial, post dose 2 Shanchol only.

A nine-panel boxplot showing per-dose seroconversion plotted by vaccine type, serotype, and latitude. A difference can be seen in per-dose seroconversion to tOPV by temperate versus non-temperate latitude.
Figure 3:

seroconversion rates in children administered (from Patriarca et al. [50]), bOPV (from Macklin et al. [53]), or mOPV1, 2, 3 (Caceres et al. [54]) plotted by non-temperate (between Tropic of Cancer and Capricorn) or temperate latitude. Seroconversion adjusted to single dose conversion as 1 − (1 − S/N)^(1/D), where S is the number seroconverting, N is the number studied, and D is the number of doses administered

Although global comparisons are limited because effectiveness studies are only feasible in settings with active poliovirus transmission, some case–control studies have identified sub-national differences in OPV effectiveness against poliomyelitis, with lower effectiveness in places that have historically struggled to control poliovirus transmission (Table 2). Grassly and colleagues demonstrated significantly lower effectiveness of tOPV and mOPV1 against WPV1 poliomyelitis in Uttar Pradesh than in the rest of India (11% per-dose effectiveness of 1 dose tOPV in Uttar Pradesh compared to 23% in the rest of India) [61]. Mangal and colleagues showed lower effectiveness of OPVs against WPV1 and WPV3 in northern states of Nigeria relative to southern states [62].

Table 2:

matched case–control studies of OPV effectiveness against WPV1 poliomyelitis

PaperYearsLocationVaccineMatched casesEffectiveness a
Balraj 1990b1988Tamil Nadu, IndiaOPV (3 doses relative to 0, type not specified)6962 (27–80)c
Sutter 1991b1988–89OmanOPV (3 doses relative to 0, type not specified)7091 (36–99)
Deming 1992b1986The GambiatOPV (3 or more doses, relative to 0–2)16472 (57–82)
Grassly 20071997–2006Uttar Pradesh, IndiatOPV (1 dose, relative to 0)149911 (7–14)d
Bihar, India20419 (8–29)d
Rest of India39923 (17–29)d
Jenkins 20082001–07North-west NigeriatOPV (1 dose, relative to 0)92513 (6–19)e
North-east Nigeria15823 (7–37)e
North-central Nigeria6823 (1–40)e
Southern Nigeria2354 (4–78)e
O’Reilly 20122001–11Pakistan & AfghanistantOPV (1 dose, relative to 0)88313 (6–19)
Mangal 20142001–12Northern NigeriatOPV (1 dose, relative to 0)130719 (16–23)e
Southern Nigeria6336 (21–57)e
Mahamud 20142013SomaliaOPV (1–3 doses relative to 0, type not specified)9959 (2–83)
Chard 20212010–20AfghanistanOPV (1 dose, any type)24919 (15–22)
PaperYearsLocationVaccineMatched casesEffectiveness a
Balraj 1990b1988Tamil Nadu, IndiaOPV (3 doses relative to 0, type not specified)6962 (27–80)c
Sutter 1991b1988–89OmanOPV (3 doses relative to 0, type not specified)7091 (36–99)
Deming 1992b1986The GambiatOPV (3 or more doses, relative to 0–2)16472 (57–82)
Grassly 20071997–2006Uttar Pradesh, IndiatOPV (1 dose, relative to 0)149911 (7–14)d
Bihar, India20419 (8–29)d
Rest of India39923 (17–29)d
Jenkins 20082001–07North-west NigeriatOPV (1 dose, relative to 0)92513 (6–19)e
North-east Nigeria15823 (7–37)e
North-central Nigeria6823 (1–40)e
Southern Nigeria2354 (4–78)e
O’Reilly 20122001–11Pakistan & AfghanistantOPV (1 dose, relative to 0)88313 (6–19)
Mangal 20142001–12Northern NigeriatOPV (1 dose, relative to 0)130719 (16–23)e
Southern Nigeria6336 (21–57)e
Mahamud 20142013SomaliaOPV (1–3 doses relative to 0, type not specified)9959 (2–83)
Chard 20212010–20AfghanistanOPV (1 dose, any type)24919 (15–22)

aVaccine effectiveness against WPV1 poliomyelitis.

bControls not test-negative acute flaccid paralysis cases.

cUnmatched case–control analysis.

dAssuming 100% routine immunization coverage.

eAssuming 0% routine immunization coverage.

Table 2:

matched case–control studies of OPV effectiveness against WPV1 poliomyelitis

PaperYearsLocationVaccineMatched casesEffectiveness a
Balraj 1990b1988Tamil Nadu, IndiaOPV (3 doses relative to 0, type not specified)6962 (27–80)c
Sutter 1991b1988–89OmanOPV (3 doses relative to 0, type not specified)7091 (36–99)
Deming 1992b1986The GambiatOPV (3 or more doses, relative to 0–2)16472 (57–82)
Grassly 20071997–2006Uttar Pradesh, IndiatOPV (1 dose, relative to 0)149911 (7–14)d
Bihar, India20419 (8–29)d
Rest of India39923 (17–29)d
Jenkins 20082001–07North-west NigeriatOPV (1 dose, relative to 0)92513 (6–19)e
North-east Nigeria15823 (7–37)e
North-central Nigeria6823 (1–40)e
Southern Nigeria2354 (4–78)e
O’Reilly 20122001–11Pakistan & AfghanistantOPV (1 dose, relative to 0)88313 (6–19)
Mangal 20142001–12Northern NigeriatOPV (1 dose, relative to 0)130719 (16–23)e
Southern Nigeria6336 (21–57)e
Mahamud 20142013SomaliaOPV (1–3 doses relative to 0, type not specified)9959 (2–83)
Chard 20212010–20AfghanistanOPV (1 dose, any type)24919 (15–22)
PaperYearsLocationVaccineMatched casesEffectiveness a
Balraj 1990b1988Tamil Nadu, IndiaOPV (3 doses relative to 0, type not specified)6962 (27–80)c
Sutter 1991b1988–89OmanOPV (3 doses relative to 0, type not specified)7091 (36–99)
Deming 1992b1986The GambiatOPV (3 or more doses, relative to 0–2)16472 (57–82)
Grassly 20071997–2006Uttar Pradesh, IndiatOPV (1 dose, relative to 0)149911 (7–14)d
Bihar, India20419 (8–29)d
Rest of India39923 (17–29)d
Jenkins 20082001–07North-west NigeriatOPV (1 dose, relative to 0)92513 (6–19)e
North-east Nigeria15823 (7–37)e
North-central Nigeria6823 (1–40)e
Southern Nigeria2354 (4–78)e
O’Reilly 20122001–11Pakistan & AfghanistantOPV (1 dose, relative to 0)88313 (6–19)
Mangal 20142001–12Northern NigeriatOPV (1 dose, relative to 0)130719 (16–23)e
Southern Nigeria6336 (21–57)e
Mahamud 20142013SomaliaOPV (1–3 doses relative to 0, type not specified)9959 (2–83)
Chard 20212010–20AfghanistanOPV (1 dose, any type)24919 (15–22)

aVaccine effectiveness against WPV1 poliomyelitis.

bControls not test-negative acute flaccid paralysis cases.

cUnmatched case–control analysis.

dAssuming 100% routine immunization coverage.

eAssuming 0% routine immunization coverage.

While serum neutralizing antibody titer of ≥1 in 8 has been established as a correlate of protection for poliomyelitis [63], it is difficult to directly compare immunogenicity and effectiveness across settings because clinical immunogenicity studies tend not to be conducted in the same places as effectiveness studies for reasons of feasibility. While seroprevalence surveys are more feasible to conduct in low-income settings, using these data to assess OPV performance requires accurate ascertainment of vaccination history, which can be difficult.

Cholera

There are three types of OCVs commercially available: inactivated whole-cell bacteria alone (WC), inactivated whole-cell bacteria with recombinant cholera toxin B (WC + rCTB), and live attenuated (LA, developed via genetic modification of a classical strain). The latter two vaccines, WC + rCTB and LA, are approved for use in some HICs for travelers 2 years and older since 1991 and 2016, respectively, but are not used in public health programs in endemic countries. The first WHO prequalified (2011) WC vaccine, Shanchol™ (Shantha Biotechnic, India), is no longer produced. A WC vaccine with an identical composition, Euvichol-Plus® (EuBiologics, Korea) is registered in several endemic countries and has been WHO prequalified since 2015. This vaccine is accessed through the Gavi-supported cholera vaccine stockpile and delivered through mass vaccination campaigns as a two-dose regimen with a two-week interval among those 1 year or older. Several killed and live attenuated OCVs are in preclinical and clinical development [28].

Due to the different indications for these products, they followed different clinical development pathways, either predominantly in HIC or in low-income countries (LICs) where cholera occurs. Consequently, there is very little published data to allow direct comparison of performance of these vaccine types in both settings.

The safety, immunogenicity, and efficacy of the WC vaccines have been almost exclusively evaluated among children and adults in cholera-affected LIC settings. Shanchol™ was registered and prequalified based on a randomized controlled trial (RCT) in Kolkata, India demonstrating efficacy of 67% in all ages combined after two years of follow-up [64, 65]. Differences in efficacy were observed between age groups, with the lowest efficacy (49%) shown in children 1–<5 years old. The trend toward lower efficacy in children < 5 years was confirmed in a meta-analysis demonstrating a pooled efficacy of inactivated OCVs of 30% (95%CI 15–42%) for children < 5 as compared to 64% (58–70%) for those ≥ 5 years [66]. All subsequent similarly composed WC vaccines have been registered and prequalified based on clinical trials conducted in endemic countries and designed to demonstrate immunologic non-inferiority to Shanchol™. One exception is a small phase 1 trial in Korean adults; a seroconversion rate (SCR, >4-fold rise) of 95% was observed for both key antigens, Inaba and Ogawa, following the standard two-dose regimen of Euvichol®, a vaccine with an identical composition [67]. In immunogenicity trials using Shanchol™, across several cholera-endemic countries, the SCRs were consistent and lower than in the Korean adults. However, among endemic populations and particularly older adults, there is high seroprevalence at baseline, and individuals with high baseline titers are less likely to have an additional 4-fold rise after vaccination [68]. As children under 5 years tend to have lower baseline seropositivity and titers, their SCRs tend to be higher than adults (though adults have higher final titers). Table 3 summarizes seroconversion rates and geometric mean titers (GMT) observed following two doses of Shanchol™ in several countries [66, 69–72]. For WC OCVs, the blunted antibody response and lower efficacy seen among young children compared to adults in the same setting may relate to the reduced ability of the immune system of a young child to generate a strong and enduring response to the polysaccharide primary protective antigen OSP [73].

Vaccines based on the combination of inactivated whole-cell bacteria and the non-toxigenic component of the Cholera Toxin dimer, CTB (WC + rCTB) have been evaluated in both HIC and LIC (Table 4). Early prototypes of the vaccine were evaluated in a human challenge among 9 US adults and found to have an efficacy of 64% four weeks after completing a three-dose series [74]. In a large, randomized, placebo-controlled trial in Bangladeshi individuals aged 2–45 years, efficacy at 6 months was estimated at 85% (95% CI: 62–94), consistent across all age strata. However, at the one-year interval, the cumulative efficacy had dropped to 62% among all participants and 38% among those aged 2–5 years [75, 76]. In Peru, two studies demonstrated conflicting results (86% short-term efficacy among male military recruits, but 0% efficacy in the first year of surveillance in a large community-based randomized trial) [77, 78]. Notably, in the community-based trial, 61% efficacy (86% against cholera requiring hospitalization) was seen during the second season following a third, booster dose [78]. Two studies of the two-dose regimen, in Mozambique and Zanzibar, estimated effectiveness of 84% and 79%, respectively [79, 80].

Table 4:

comparison of efficacy and effectiveness estimates of inactivated whole cell bacteria with recombinant cholera toxin B (WC + rCTB) and live-attenuated (LA) vaccines in HICs and LICs

WC + rCTB vaccines
HICLIC
USA (challenge study)
1 month
Matlab, Bangladesh
12 months
Peru, Military
5 months
Pampas, Peru
12 months
Efficacy64%a62%b86%0%c
Biera, Mozambique
5 months
Zanzibar, Tanzania
15 months
Effectiveness84%d79%e
Live attenuated vaccines based on CVD 103
HICLIC
USA (challenge study)
1 month
Jakarta, Indonesia
Orochol (210×108CFU)Vaxchora (4×108 to 2×109CFU)Orochol-E (5×109CFU)
Efficacy79.0%f79.5%g14%h
EffectivenessPohnpei, Micronesia
Orochol-E (5×109CFU)
79.2%i
WC + rCTB vaccines
HICLIC
USA (challenge study)
1 month
Matlab, Bangladesh
12 months
Peru, Military
5 months
Pampas, Peru
12 months
Efficacy64%a62%b86%0%c
Biera, Mozambique
5 months
Zanzibar, Tanzania
15 months
Effectiveness84%d79%e
Live attenuated vaccines based on CVD 103
HICLIC
USA (challenge study)
1 month
Jakarta, Indonesia
Orochol (210×108CFU)Vaxchora (4×108 to 2×109CFU)Orochol-E (5×109CFU)
Efficacy79.0%f79.5%g14%h
EffectivenessPohnpei, Micronesia
Orochol-E (5×109CFU)
79.2%i

a4 weeks post three-dose vaccination.

b1 year post vaccination with three doses.

c1 year post vaccination with two doses.

d6 months post one or two-dose vaccination.

e1 year post vaccination with two doses.

fBased on prevention of > 3L diarrhea three months post vaccination.

gBased on prevention of > 3L diarrhea three months post vaccination.

hNo statistically significant evidence of efficacy throughout 4 years of follow-up.

i3 months post vaccination.

Table 4:

comparison of efficacy and effectiveness estimates of inactivated whole cell bacteria with recombinant cholera toxin B (WC + rCTB) and live-attenuated (LA) vaccines in HICs and LICs

WC + rCTB vaccines
HICLIC
USA (challenge study)
1 month
Matlab, Bangladesh
12 months
Peru, Military
5 months
Pampas, Peru
12 months
Efficacy64%a62%b86%0%c
Biera, Mozambique
5 months
Zanzibar, Tanzania
15 months
Effectiveness84%d79%e
Live attenuated vaccines based on CVD 103
HICLIC
USA (challenge study)
1 month
Jakarta, Indonesia
Orochol (210×108CFU)Vaxchora (4×108 to 2×109CFU)Orochol-E (5×109CFU)
Efficacy79.0%f79.5%g14%h
EffectivenessPohnpei, Micronesia
Orochol-E (5×109CFU)
79.2%i
WC + rCTB vaccines
HICLIC
USA (challenge study)
1 month
Matlab, Bangladesh
12 months
Peru, Military
5 months
Pampas, Peru
12 months
Efficacy64%a62%b86%0%c
Biera, Mozambique
5 months
Zanzibar, Tanzania
15 months
Effectiveness84%d79%e
Live attenuated vaccines based on CVD 103
HICLIC
USA (challenge study)
1 month
Jakarta, Indonesia
Orochol (210×108CFU)Vaxchora (4×108 to 2×109CFU)Orochol-E (5×109CFU)
Efficacy79.0%f79.5%g14%h
EffectivenessPohnpei, Micronesia
Orochol-E (5×109CFU)
79.2%i

a4 weeks post three-dose vaccination.

b1 year post vaccination with three doses.

c1 year post vaccination with two doses.

d6 months post one or two-dose vaccination.

e1 year post vaccination with two doses.

fBased on prevention of > 3L diarrhea three months post vaccination.

gBased on prevention of > 3L diarrhea three months post vaccination.

hNo statistically significant evidence of efficacy throughout 4 years of follow-up.

i3 months post vaccination.

There have been several commercial versions of the LA OCV, all based on the genetically attenuated CVD 103-HgR strain derived from a pathogenic classical Inaba strain 569B [81–84]. Throughout several different challenge trials in US volunteers, high levels of protection ranging from 90 to 95% at 1 month and 79.5–95.4% at 3 months were consistently demonstrated [81–83]. Experience with CVD 103-HgR-based vaccines is more limited in LICs. One retrospective cohort study was conducted following a mass vaccination campaign in response to a cholera introduction on the non-endemic Pohnpei island (Federated States of Micronesia). The campaign targeted all individuals older than 2 years and achieved ~45% vaccination coverage; analysis demonstrated 79% effectiveness among all ages combined [85]. However, a large RCT enrolling participants aged 2–41 years in Jakarta, Indonesia with a 4-year follow-up did not demonstrate efficacy against medically attended cholera following a single dose of CVD 103-HgR; neither analysis of shorter post-vaccination intervals nor analysis by age strata yielded significant results [86]. In a nested immunogenicity study, there was a negative relationship between baseline titer and fold antibody rise, and SCR was 42% among those with elevated baseline titers compared to 86% among individuals with low baseline titer [86].

Evidence for understanding disparities in performance

Evidence suggests that maternal, infant, and environmental factors, in addition to characteristics of the vaccines themselves, all may contribute to the poor performance of oral vaccines in LMICs. The influence of these factors spans a timescale that ranges from before the birth of the infant to well after the vaccines have been administered.

Maternal factors and antibody interference

The most well-studied maternal factors are transplacental and breast milk antibodies [50, 87]. Serum anti-rotavirus IgG that is passively transferred from mother to infant interferes with seroconversion to several live, oral rotavirus vaccines (ORVs) [88–91], and placentally transferred maternal anti-polio IgG has long been known to interfere with seroconversion after OPV, with the magnitude of the effect apparently serotype-specific [50, 92–95]. Given that OCVs are not given below one year of age, transplacental antibodies are not likely to contribute much to observed differences. Indeed, a recent study aimed at determining the half-life of maternal antibodies to multiple antigens in the diphtheria, pertussis, and tetanus vaccine estimated the range to be between 28.7 and 35.1 days, irrespective of demographic or antigen factors [96]. These data provide a rationale for delaying the first dose of vaccination with ORV or OPV as a strategy to potentially reduce the interference from transplacental IgG. However, studies comparing seroconversion rates between children receiving the Rotarix vaccine at 6 and 10 weeks versus 10 and 14 weeks did not consistently show improvements in response [97]; a potential exception is a study in Bangladesh which found higher-than-expected efficacy for Rotarix given on a 10- and 17-week schedule, though there was no early-schedule arm for comparison [98]. Historical analysis did not find significant differences in performance of OPV when the first dose was given at 8 compared to 6 weeks [50], and OPV continues to be given on an early schedule in many countries to achieve early protection for infants and align with the primary series of other antigens. Baseline antibody levels may also be elevated due to natural infection; indeed, some work suggests that failure to fully account for factors affecting susceptibility (including natural infection) could partially explain lower efficacy and effectiveness estimates for ORVs in high-burden settings [99]. Similarly, interference from antibodies resulting from prior exposure to cholera or related antigens is postulated to be a major reason that the LA OCV demonstrated no efficacy in the endemic population of Jakarta, but high efficacy or effectiveness in human challenge studies and in the non-endemic population of Pohnpei, Micronesia [77]. Although it is increasingly recognized that other immune cells (e.g. T lymphocytes, macrophages) can also be transferred across the placenta and may play a role in protecting infants from infection by pathogens [100], their role in infant response to vaccines has not been systematically studied.

Human breast milk is a rich source of bioactive compounds, including secretory IgA as well as other immunologic factors, growth factors, and other non-nutritive elements [101]. Several studies have demonstrated significant differences in the levels of anti-rotavirus IgA and neutralizing antibodies in human milk in samples collected from mothers in LMICs compared to HICs [90, 102–108]. Colostrum (compared with other milk stages) has been demonstrated to be highest in anti-polio IgA [50]. Withholding breast feeding has therefore evaluated as an intervention to improve vaccine response. However, there is little evidence for impact for ORVs [109, 110], with one study in Pakistan even showing higher response in infants who were breastfed immediately post-vaccination [111]. Furthermore, a recent study evaluating maternal anti-rotavirus antibody levels between India, UK, and Malawi showed similar or higher antibody levels in the UK compared to India, yet vaccine virus shedding and seroconversion rates were significantly higher in the UK [112]. Similarly, although some early research suggested an inhibitory effect of frequent breastfeeding on OPV response, particularly in the first days of life [50, 113], later research has found very little effect [50, 114, 115] or even a positive association [116, 117]. Given the older age at which cholera vaccines are typically given, few studies have assessed the impact of withholding breastfeeding. However, one study in Bangladesh found that vibriocidal responses were higher among older infants given buffered Dukoral (WC + rCTB) when breastfeeding was withheld for 3 h prior to vaccination; no differences were seen among younger infants or in IgA or IgG antitoxin responses [118]. Bioactive non-antibody components in human milk such as lactoferrin, lactadherin, and tenascin-C have also been considered for their effects on ORV response. However, the few systematic assessments of these factors in LMIC populations have demonstrated conflicting outcomes [106, 119, 120]. In vitro studies also indicate the potential for human milk oligosaccharides (HMO) to influence rotavirus vaccine replication, and this remains to be validated in population studies [121]. HMOs and other breast milk components have not been studied for their influence on OPV or OCV response. However, one study found that infants receiving formula supplemented with bovine milk oligosaccharides had a higher fecal IgA response to OPV compared with infants receiving conventional formula; the authors speculated that this effect was mediated through differences in the infant gut microbiome [122].

Infant and maternal–infant factors

Genetic differences in the expression of histo-blood group antigens (HBGAs) may play a role in differences in ORV response. Specifically, differences in the expression of functional fucosyltransferase (FUT) genes have been evaluated. FUT2 and FUT3 determine the expression of α(1,2) and α(1,3) fucosylated HBGAs that establish individual secretor and Lewis status, respectively. These glycans serve as cellular attachment factors for several human rotaviruses in a VP4 genotype-dependent manner and thus can influence susceptibility to live ORVs as well as wild-type rotavirus strains. Several studies have reported lower seroconversion rates to Rotarix vaccine in non-secretor infants (lacking a functional FUT2 gene), while the effects on vaccine shedding have been less clear [123–130]. The association of the Lewis A phenotype with reduced seroconversion or vaccine shedding has also been described in some studies. There are fewer studies on the effect of secretor status and Lewis status on other licensed rotavirus vaccines [131–133]. Outside the context of VP4–glycan interactions, differences in HBGA expression are also associated with differences in microbiome and HMO composition, and thus may indirectly influence vaccine response. Two studies have determined the effect of maternal secretor status on infant rotavirus vaccine response but with conflicting results, possibly related to the underlying differences in the study populations given that one study was conducted in the USA and the other in Bangladesh [124, 134]. In contrast, HBGAs have not been found to play a role in OPV immunogenicity [135]. Secretor status has not been found to significantly affect OCV immunogenicity [136]. Although O blood type has been associated with increased risk for severe cholera disease, the evidence for the effect of O blood type on immune response has been mixed [137–139], perhaps related in part due to differences in the vaccines studied and the mechanism by which they stimulate an immune response. Small studies have found genes potentially associated with the development of paralytic polio [140] or susceptibility to cholera [141], but these have yet to be studied in relation to vaccine response.

Nutritional status is another infant factor that may affect vaccine response. A review of rotavirus vaccine performance found that vaccine effectiveness and efficacy point estimates were 37–64% lower in malnourished versus well-nourished children, when categorized by length-for-age (a measure of chronic undernutrition) [142]. Several studies have also noted reduced immunogenicity of OPV in children with chronic malnutrition [117, 143, 144]. Few similar studies have been conducted for cholera vaccines, but no association between chronic undernutrition and response to Dukoral or CTB subunit was found in two small studies in Bangladesh [145, 146]. Micronutrients have also been investigated for their effect on oral vaccine response. Despite the established effects of vitamin A on overall child health, and linkages of vitamin D and immune function, evidence to-date is limited and has not supported a clear relationship between these micronutrients and response to ORV, OPV, or OCV [147–149]. Although zinc has not been found to be associated with response to ORV [149, 150] or OPV [151], some studies have suggested a positive effect of zinc on vibriocidal immune response to killed cholera vaccine [118, 152, 153]. Nonetheless, meta-analysis has not found a significant effect of micronutrient supplementation on oral vaccine response [147]. Nutritional status is also highly intertwined with other social and environmental factors, including co-infections and their impact on gut health, making this a complex area of study.

Environmental factors

For live vaccines that replicate in the infant gut, the intestinal microenvironment is likely to play a critical role in vaccine response. Significant associations have been described between markers of environmental enteric dysfunction (EED) and lack of response to ORVs [116, 154, 155] or impaired response to OPV [116, 156] or live-attenuated (LA) OCV in some studies [157]. EED is thought to be the result of repeated enteric infections in settings of low hygiene and sanitation infrastructure and is characterized by increased inflammation as well as changes in intestinal architecture including villous blunting and crypt hyperplasia. Although a number of biomarkers have been studied, none are widely accepted as diagnostic, except intestinal biopsy. While it is logical to infer that EED would be unfavorable for the replication of live vaccines, increased seroconversion to ORVs in children with markers associated with EED has also been described [158, 159]. A positive association between markers of EED response to WC OCV has also been reported in children, suggesting a complex relationship among EED, inflammation, and immune response to mucosal vaccines [145]. In terms of microbial ecology at the time of vaccination, the co-administration of other vaccines (OPV influence on ORV being the most-studied example), differences in the intestinal microbiome and virome, as well as concurrent infections have all been evaluated as factors influencing oral vaccine response in different studies. While the extent to which OPV interferes with ORV response has varied among studies, a large analysis pooling data from 33 high- and low-childhood mortality regions showed that OPV co-administration is significantly associated with reduced seroconversion to ORVs [160]. In contrast, OPV immunogenicity does not appear to be affected by ORV co-administration [147, 161–169]. In one study of co-administration of WC OCV and OPV in toddlers (aged 1–3 years), immune responses to both vaccines were comparable in the co-administration and single administration groups [170]. The presence of non-polio enteroviruses has also been associated with poor response to ORVs [171, 172] as well as poor responses to OPV in multiple countries [171, 173, 174], but has not been well studied for OCVs. There are also conflicting results from different studies on the association of intestinal microbiome composition and oral vaccine response. Correlations between presence of Enterobacteriaceae and seroconversion to ORVs have been described in Ghana and Pakistan [172, 175]. However, these differences were not seen in a cohort from India, UK, and Malawi [112, 176]. In studies conducted in Bangladesh and China, increased prevalence of Actinobacteria (such as Bifidobacteria) was positively associated with OPV response, but no impact was found in a cohort of infants in South India [174]. The relationship of other co-infections or microbiome diversity to OPV response appears complex, with interpretations complicated by small sample sizes [171, 177–179]. Few comparable data exist for OCVs, and although helminth burden has been negatively associated with response to cholera infection, antihelminthic treatment has not been shown to improve OCV response [147, 180, 181]. Finally, other environmental factors that impact oral vaccine response include differences in access to water, sanitation, and hygiene that may result in a higher force of infection and higher rates of other enteric infections in LMICs.

Vaccine factors

Vaccine-related factors, including dose and strain, may also play a role in overall vaccine performance. Increasing the dose titer has been explored as a way to improve vaccine immunogenicity, but without supportive evidence to date for ORV [182], and with a small effect demonstrated via meta-analysis for OPV and OCV [147]. Administration of a booster dose of ORV has also been associated with increased immune response [183], while for OPV, a birth dose is recommended to secure immune response in high-risk populations [184]. Limited data are available to determine the value of additional doses of OCV. Strain or genotype mismatch has also been suggested as a possible mechanism for differing vaccine performance. Analysis has shown that ORVs are slightly less effective against heterotypic versus vaccine strains of rotavirus [185, 186]. OPV and OCV protect against circulating serotypes and serogroups, and the effects of genetic variations on vaccine performance have not been comprehensively studied.

Conclusions

Despite the tremendous successes of oral vaccines against rotavirus, polio, and cholera, their performance is still limited in LMICs among young children. Although performance disparities have been well described, they are not as well understood. Overall, these data highlight the rather ambiguous and often contradictory nature of evidence on factors affecting oral vaccine response, cautioning against broad extrapolation of outcomes based on one population or one vaccine type. Indeed, a systematic analysis of interventions aimed at improving oral vaccine response found little evidence to support withholding breastfeeding, vaccine buffering, or narrowing the dose window between vaccines [147]. Even interventions well supported by evidence, such as separating oral rotavirus and oral poliovirus vaccine administration, may not be feasible programmatically. In addition, meaningful impact on performance of oral vaccines will likely only be possible with a suite of interventions, given the complex and multifactorial nature of the problem and our lack of complete understanding. Development and deployment of new vaccines and innovative precision population health approaches that incorporate region-specific environment and host factors may be required to successfully improve the performance of oral vaccines in LMICs.

Abbreviations

    Abbreviations
     
  • cVDPV

    circulating vaccine-derived poliovirus

  •  
  • DRC

    Democratic Republic of Congo

  •  
  • dsRNA

    double-stranded RNA

  •  
  • EED

    environmental enteric dysfunction

  •  
  • FUT

    fucosyltransferase

  •  
  • GMT

    geometric mean titers

  •  
  • HBGA

    histo-blood group antigens

  •  
  • HIC

    high-income countries

  •  
  • HMO

    human milk oligosaccharide

  •  
  • LA

    live, attenuated

  •  
  • LMIC

    lower- and middle-income countries

  •  
  • OCV

    Oral Cholera Vaccine

  •  
  • OPV

    Oral Polio Vaccine

  •  
  • ORV

    Oral Rotavirus Vaccine

  •  
  • OSP

    O-specific polysaccharide

  •  
  • RCT

    randomized controlled trial

  •  
  • rCTB

    recombinant cholera toxin B

  •  
  • SCR

    seroconversion rate

  •  
  • VAPP

    vaccine associated - paralytic polio

  •  
  • WC

    whole-cell

  •  
  • WHO

    World Health Organization

  •  
  • WPV

    wild poliovirus

Acknowledgements

Not applicable.

Ethical Approval

Not applicable.

Conflict of interest

The authors report no conflicts of interest.

Funding

This work was conducted as part of the authors’ job responsibilities.

Data Availability

All data reported are publicly available.

Author Contributions

Rachel Burke (Conceptualization, Formal analysis, Visualization, Writing—original draft, Writing—review & editing), Sasirekha Ramani (Writing—original draft, Writing—review & editing), Julia Lynch (Formal analysis, Writing—original draft, Writing—review & editing), Laura Cooper (Formal analysis, Visualization, Writing—original draft, Writing—review & editing), Haeun Cho (Data curation, Formal analysis, Writing—review & editing), Carl Kirkwood (Conceptualization, Writing—review & editing), Ananda Bandyopadhyay (Supervision, Writing—review & editing), Duncan Steele (Conceptualization, Supervision, Writing—review & editing), and Gagandeep (Cherry) Kang (Conceptualization, Supervision, Writing—review & editing)

References

1.

World Health Organization
. History of polio vaccination. https://www.who.int/news-room/spotlight/history-of-vaccination/history-of-polio-vaccination. (
June 14
, date last accessed).

2.

Parker
EP
,
Ramani
S
,
Lopman
BA
,
Church
JA
,
Iturriza-Gómara
M
,
Prendergast
AJ
, et al.
Causes of impaired oral vaccine efficacy in developing countries
.
Future Microbiol
2018
,
13
,
97
118
. doi: https://doi.org/

3.

Holmgren
J
,
Czerkinsky
C.
Mucosal immunity and vaccines
.
Nat Med
2005
,
11
,
S45
S53
. doi: https://doi.org/

4.

Modlin
JF
,
Bandyopadhyay
AS
,
Sutter
R.
Immunization against poliomyelitis and the challenges to worldwide poliomyelitis eradication
.
J Infect Dis
2021
,
224
,
S398
404
. doi: https://doi.org/

5.

Clark
A
,
Mahmud
S
,
Debellut
F
,
Pecenka
C
,
Jit
M
,
Perin
J
, et al.
Estimating the global impact of rotavirus vaccines on child mortality
.
Int J Infect Dis
2023
,
137
,
90
7
. doi: https://doi.org/

6.

Lee
EC
,
Azman
AS
,
Kaminsky
J
,
Moore
SM
,
McKay
HS
,
Lessler
J.
The projected impact of geographic targeting of oral cholera vaccination in sub-Saharan Africa: a modeling study
.
PLoS Med
2019
,
16
,
e1003003
. doi: https://doi.org/

7.

Wierzba
TF.
Oral cholera vaccines and their impact on the global burden of disease
.
Human Vaccin Immunother
2019
,
15
,
1294
301
. doi: https://doi.org/

8.

Parashar
UD
,
Hummelman
EG
,
Bresee
JS
,
Miller
MA
,
Glass
RI.
Global illness and deaths caused by rotavirus disease in children
.
Emerg Infect Dis
2003
,
9
,
565
72
. doi: https://doi.org/

9.

Tate
JE
,
Burton
AH
,
Boschi-Pinto
C
,
Parashar
UD
,
World Health Organization–Coordinated Global Rotavirus Surveillance Network
.
Global, regional, and national estimates of rotavirus mortality in children <5 years of age, 2000-2013
.
Clin Infect Dis
2016
,
62
,
S96
S105
. doi: https://doi.org/

10.

Black
RE
,
Perin
J
,
Yeung
D
,
Rajeev
T
,
Miller
J
,
Elwood
SE
, et al.
Estimated global and regional causes of deaths from diarrhoea in children younger than 5 years during 2000-21: a systematic review and Bayesian multinomial analysis
.
Lancet Glob Health
2024
,
12
,
e919
28
. doi: https://doi.org/

11.

Bishop
RF
,
Davidson
GP
,
Holmes
IH
,
Ruck
BJ.
Virus particles in epithelial cells of duodenal mucosa from children with acute non-bacterial gastroenteritis
.
Lancet
1973
,
2
,
1281
3
. doi: https://doi.org/

12.

Bishop
RF
,
Davidson
GP
,
Holmes
IH
,
Ruck
BJ.
Detection of a new virus by electron microscopy of faecal extracts from children with acute gastroenteritis
.
Lancet
1974
,
1
,
149
51
. doi: https://doi.org/

13.

Estes
M
,
Kapikian
A
,
Knipe
D
,
Howley
P.
Rotaviruses
. In:
Knipe
DM
,
Howley
PM
,
Griffin
DE
,
Lamb
RA
,
Martin
MA
,
Roizman
B
,
Straus
SE
(eds),
Fields Virology
.
Philadelphia
:
Kluwer Health/Lippincott, Williams and Wilkins
,
2007
,
1917
74
.

14.

Ball
JM
,
Tian
P
,
Zeng
CQ
,
Morris
AP
,
Estes
MK.
Age-dependent diarrhea induced by a rotaviral nonstructural glycoprotein
.
Science
1996
,
272
,
101
4
. doi: https://doi.org/

15.

Crawford
SE
,
Ramani
S
,
Tate
JE
,
Parashar
UD
,
Svensson
L
,
Hagbom
M
, et al.
Rotavirus infection
.
Nat Rev Dis Primers
2017
,
3
,
17083
. doi: https://doi.org/

16.

Hagbom
M
,
Istrate
C
,
Engblom
D
,
Karlsson
T
,
Rodriguez-Diaz
J
,
Buesa
J
, et al.
Rotavirus stimulates release of serotonin (5-HT) from human enterochromaffin cells and activates brain structures involved in nausea and vomiting
.
PLoS Pathog
2011
,
7
,
e1002115
. doi: https://doi.org/

17.

Gomez-Rial
J
,
Sanchez-Batan
S
,
Rivero-Calle
I
,
Pardo-Seco
J
,
Martinón-Martínez
JM
,
Salas
A
, et al.
Rotavirus infection beyond the gut
.
Infect Drug Resist
2019
,
12
,
55
64
. doi: https://doi.org/

18.

Geiger
K
,
Stehling-Ariza
T
,
Bigouette
JP
,
Bennett
SD
,
Burns
CC
,
Quddus
A
, et al.
Progress toward poliomyelitis eradication - Worldwide, January 2022-December 2023
.
MMWR Morb Mortal Wkly Rep
2024
,
73
,
441
6
. doi: https://doi.org/

19.

Bandyopadhyay
AS
,
Garon
J
,
Seib
K
,
Orenstein
WA.
Polio vaccination: past, present and future
.
Future Microbiol
2015
,
10
,
791
808
. doi: https://doi.org/

20.

Racaniello
VR.
One hundred years of poliovirus pathogenesis
.
Virology
2006
,
344
,
9
16
. doi: https://doi.org/

21.

Ohka
S
,
Nihei
C
,
Yamazaki
M
,
Nomoto
A.
Poliovirus trafficking toward central nervous system via human poliovirus receptor-dependent and -independent pathway
.
Front Microbiol
2012
,
3
,
147
. doi: https://doi.org/

22.

Ali
M
,
Nelson
AR
,
Lopez
AL
,
Sack
DA.
Updated global burden of cholera in endemic countries
.
PLoS NeglTrop Dis
2015
,
9
,
e0003832
. doi: https://doi.org/

23.

Ilic
I
,
Ilic
M.
Global patterns of trends in cholera mortality
.
Trop Med Infect Dis
2023
,
8
,
169
. doi: https://doi.org/

24.

New analysis confirms world seeing an upsurge of cholera. https://www.who.int/news/item/22-09-2023-new-analysis-confirms-world-seeing-an-upsurge-of-cholera. (

July 5
, date last accessed).

25.

World Health Organization Emergency Response Team
. Multi-country outbreak of cholera, External situation report #13 - 17 April 2024. https://www.who.int/publications/m/item/multi-country-outbreak-of-cholera--external-situation-report--13---17-april-2024. (
July 5
, date last accessed).

26.

Chowdhury
F
,
Ross
AG
,
Islam
MT
,
McMillan
NAJ
,
Qadri
F.
Diagnosis, management, and future control of cholera
.
Clin Microbiol Rev
2022
,
35
,
e0021121
. doi: https://doi.org/

27.

Nelson
EJ
,
Nelson
DS
,
Salam
MA
,
Sack
DA.
Antibiotics for both moderate and severe cholera
.
N Engl J Med
2011
,
364
,
5
7
. doi: https://doi.org/

28.

Montero
DA
,
Vidal
RM
,
Velasco
J
,
George
S
,
Lucero
Y
,
Gómez
LA
, et al.
Vibrio cholerae, classification, pathogenesis, immune response, and trends in vaccine development
.
Front Med (Lausanne)
2023
,
10
,
1155751
. doi: https://doi.org/

29.

Kanungo
S
,
Azman
AS
,
Ramamurthy
T
,
Deen
J
,
Dutta
S.
Cholera
.
Lancet
2022
,
399
,
1429
40
. doi: https://doi.org/

30.

Weil
AA
,
Becker
RL
,
Harris
JB.
Vibrio cholerae at the intersection of immunity and the microbiome
.
mSphere
2019
,
4
,
e00597
19
. doi: https://doi.org/

31.

Iyer
AS
,
Harris
JB.
Correlates of protection for cholera
.
J Infect Dis
2021
,
224
,
S732
S737
. doi: https://doi.org/

32.

International Vaccine Access Center
.
Current and upcoming rotavirus vaccines
.
2020
. https://view-hub.org/sites/default/files/2020-09/ROTA-Brief2-ProductLandscape-SP-1-3.pdf (
17 July 2024
, date last accessed).

33.

Cates
JE
,
Tate
JE
,
Parashar
U.
Rotavirus vaccines: progress and new developments
.
Expert Opin Biol Ther
2022
,
22
,
423
32
. doi: https://doi.org/

34.

World Health Organization
.
Rotavirus vaccines completed dose (RotaC) immunization coverage among 1-year-olds (%)
. https://immunizationdata.who.int/global/wiise-detail-page/rotavirus-vaccination-coverage?CODE=Global&GROUP=Countries&ANTIGEN=ROTAC&YEAR= (
17 July 2024
, date last accessed).

35.

Hallowell
BD
,
Chavers
T
,
Parashar
U
,
Tate
JE.
Global Estimates of Rotavirus Hospitalizations Among Children Below 5 Years in 2019 and Current and Projected Impacts of Rotavirus Vaccination
.
J. Pediatric Infect. Dis. Soc.
2021
,
11
,
149
58
. doi: https://doi.org/

36.

Ruiz-Palacios
GM
,
Pérez-Schael
I
,
Velázquez
FR
,
Abate
H
,
Breuer
T
,
Clemens
SC
, et al.
Safety and efficacy of an attenuated vaccine against severe rotavirus gastroenteritis
.
N Engl J Med
2006
,
354
,
11
22
. doi: https://doi.org/

37.

Vesikari
T
,
Matson
DO
,
Dennehy
P
,
Van Damme
P
,
Santosham
M
,
Rodriguez
Z
, et al.
Safety and efficacy of a pentavalent human-bovine (WC3) reassortant rotavirus vaccine
.
N Engl J Med
2006
,
354
,
23
33
. doi: https://doi.org/

38.

Madhi
SA
,
Cunliffe
NA
,
Steele
D
,
Witte
D
,
Kirsten
M
,
Louw
C
, et al.
Effect of human rotavirus vaccine on severe diarrhea in African infants
.
N Engl J Med
2010
,
362
,
289
98
. doi: https://doi.org/

39.

Armah
GE
,
Sow
SO
,
Breiman
RF
,
Dallas
MJ
,
Tapia
MD
,
Feikin
DR
, 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: https://doi.org/

40.

Zaman
K
,
Dang
DA
,
Victor
JC
,
Shin
S
,
Yunus
M
,
Dallas
MJ
, 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: https://doi.org/

41.

Isanaka
S
,
Guindo
O
,
Langendorf
C
,
Matar Seck
A
,
Plikaytis
BD
,
Sayinzoga-Makombe
N
, et al.
Efficacy of a low-cost, heat-stable oral rotavirus vaccine in Niger
.
N Engl J Med
2017
,
376
,
1121
30
. doi: https://doi.org/

42.

Kulkarni
PS
,
Desai
S
,
Tewari
T
,
Kawade
A
,
Goyal
N
,
Garg
BS
, et al.
A randomized Phase III clinical trial to assess the efficacy of a bovine-human reassortant pentavalent rotavirus vaccine in Indian infants
.
Vaccine
2017
,
35
,
6228
37
. doi: https://doi.org/

43.

Bhandari
N
,
Rongsen-Chandola
T
,
Bavdekar
A
,
John
J
,
Antony
K
,
Taneja
S
, et al.
Efficacy of a monovalent human-bovine (116E) rotavirus vaccine in Indian infants: a randomised, double-blind, placebo-controlled trial
.
Lancet
2014
,
383
,
2136
43
. doi: https://doi.org/

44.

Burnett
E
,
Parashar
UD
,
Tate
JE.
Real-world effectiveness of rotavirus vaccines, 2006-19: a literature review and meta-analysis
.
Lancet Glob Health
2020
,
8
,
e1195
202
. doi: https://doi.org/

45.

Burnett
E
,
Parashar
UD
,
Tate
JE.
Global impact of rotavirus vaccination on diarrhea hospitalizations and deaths among children 5 years old: 2006–2019
.
J Infect Dis
2020
,
222
,
1731
9
. doi: https://doi.org/

46.

Burns
CC
,
Diop
OM
,
Sutter
RW
,
Kew
OM.
Vaccine-derived polioviruses
.
J Infect Dis
2014
,
210
,
S283
93
. doi: https://doi.org/

47.

Kurji
FD
,
Bandyopadhyay
AS
,
Zipursky
S
,
Cooper
LV
,
Gast
C
,
Toher
M
, et al.
Novel oral polio vaccine Type 2 use for polio outbreak response: a global effort for a global health emergency
.
Pathogens
2024
,
13
,
273
. doi: https://doi.org/

48.

Yeh
MT
,
Smith
M
,
Carlyle
S
,
Konopka-Anstadt
JL
,
Burns
CC
,
Konz
J
, et al.
Genetic stabilization of attenuated oral vaccines against poliovirus types 1 and 3
.
Nature
2023
,
619
,
135
42
. doi: https://doi.org/

49.

Peak
CM LH
,
Voorman
A
,
Cooper
LV
,
Hawes
K
,
Bandyopadhyay
AS.
Monitoring the risk of Type-2 circulating vaccine-derived poliovirus emergence during roll-out of Type-2 novel oral polio vaccine
. 9th International Conference on Infectious Disease Dynamics.
Bologna, Italy
:
Epidemics
,
2023
.

50.

Patriarca
PA
,
Wright
PF
,
John
TJ.
Factors affecting the immunogenicity of oral poliovirus vaccine in developing countries: review
.
Rev Infect Dis
1991
,
13
,
926
39
. doi: https://doi.org/

51.

John
TJ.
Geographic Variation in Vaccine Efficacy: The Polio Experience
.
New York, NY
:
Springer New York
,
1989
,
61
6
.

52.

McBean
AM
,
Thoms
ML
,
Albrecht
P
,
Cuthie
JC
,
Bernier
R.
Serologic response to oral polio vaccine and enhanced-potency inactivated polio vaccines
.
Am J Epidemiol
1988
,
128
,
615
28
. doi: https://doi.org/

53.

Macklin
GR
,
Grassly
NC
,
Sutter
RW
,
Mach
O
,
Bandyopadhyay
AS
,
Edmunds
WJ
, et al.
Vaccine schedules and the effect on humoral and intestinal immunity against poliovirus: a systematic review and network meta-analysis
.
Lancet Infect Dis
2019
,
19
,
1121
8
. doi: https://doi.org/

54.

Caceres
VM
,
Sutter
RW.
Sabin monovalent oral polio vaccines: review of past experiences and their potential use after polio eradication
.
Clin Infect Dis
2001
,
33
,
531
41
. doi: https://doi.org/

55.

de Deus
N
,
Capitine
IPU
,
Bauhofer
AFL
,
Marques
S
,
Cassocera
M
,
Chissaque
A
, et al.
Immunogenicity of reduced-dose monovalent Type 2 oral poliovirus vaccine in Mocuba, Mozambique
.
J Infect Dis
2022
,
226
,
292
8
. doi: https://doi.org/

56.

Ochoge
M
,
Futa
AC
,
Umesi
A
,
Affleck
L
,
Kotei
L
,
Daffeh
B
, et al.
Safety of the novel oral poliovirus vaccine type 2 (nOPV2) in infants and young children aged 1 to <5 years and lot-to-lot consistency of the immune response to nOPV2 in infants in The Gambia: a phase 3, double-blind, randomised controlled trial
.
Lancet
2024
,
403
,
1164
75
. doi: https://doi.org/

57.

Saez-Llorens
X
,
Bandyopadhyay
AS
,
Gast
C
,
Leon
TD
,
DeAntonio
R
,
Jimeno
J
, et al.
Safety and immunogenicity of two novel type 2 oral poliovirus vaccine candidates compared with a monovalent type 2 oral poliovirus vaccine in children and infants: two clinical trials
.
Lancet
2021
,
397
,
27
38
. doi: https://doi.org/

58.

Wilkinson
AL
,
Zaman
K
,
Hoque
M
,
Estivariz
CF
,
Burns
CC
,
Konopka-Anstadt
JL
, et al.
Immunogenicity of novel oral poliovirus vaccine type 2 administered concomitantly with bivalent oral poliovirus vaccine: an open-label, non-inferiority, randomised, controlled trial
.
Lancet Infect Dis
2023
,
23
,
1062
71
. doi: https://doi.org/

59.

Zaman
K
,
Bandyopadhyay
AS
,
Hoque
M
,
Gast
C
,
Yunus
M
,
Jamil
KM
, et al.
Evaluation of the safety, immunogenicity, and faecal shedding of novel oral polio vaccine type 2 in healthy newborn infants in Bangladesh: a randomised, controlled, phase 2 clinical trial
.
Lancet
2023
,
401
,
131
9
. doi: https://doi.org/

60.

Rivera Mejia
L
,
Pena Mendez
L
,
Bandyopadhyay
AS
,
Gast
C
,
Mazara
S
,
Rodriguez
K
, et al.
Safety and immunogenicity of shorter interval schedules of the novel oral poliovirus vaccine type 2 in infants: a phase 3, randomised, controlled, non-inferiority study in the Dominican Republic
.
Lancet Infect Dis
2024
,
24
,
275
84
. doi: https://doi.org/

61.

Grassly
NC
,
Wenger
J
,
Durrani
S
,
Bahl
S
,
Deshpande
JM
,
Sutter
RW
, et al.
Protective efficacy of a monovalent oral type 1 poliovirus vaccine: a case-control study
.
Lancet
2007
,
369
,
1356
62
. doi: https://doi.org/

62.

Mangal
TD
,
Aylward
RB
,
Mwanza
M
,
Gasasira
A
,
Abanida
E
,
Pate
MA
, et al.
Key issues in the persistence of poliomyelitis in Nigeria: a case-control study
.
Lancet Glob Health
2014
,
2
,
e90
7
. doi: https://doi.org/

63.

Plotkin
SA.
Immunologic correlates of protection induced by vaccination
.
Pediatr Infect Dis J
2001
,
20
,
63
75
. doi: https://doi.org/

64.

Sur
D
,
Lopez
AL
,
Kanungo
S
,
Paisley
A
,
Manna
B
,
Ali
M
, et al.
Efficacy and safety of a modified killed-whole-cell oral cholera vaccine in India: an interim analysis of a cluster-randomised, double-blind, placebo-controlled trial
.
Lancet
2009
,
374
,
1694
702
. doi: https://doi.org/

65.

Bhattacharya
SK
,
Sur
D
,
Ali
M
,
Kanungo
S
,
You
YA
,
Manna
B
, et al.
5 year efficacy of a bivalent killed whole-cell oral cholera vaccine in Kolkata, India: a cluster-randomised, double-blind, placebo-controlled trial
.
Lancet Infect Dis
2013
,
13
,
1050
6
. doi: https://doi.org/

66.

Bi
Q
,
Ferreras
E
,
Pezzoli
L
,
Legros
D
,
Ivers
LC
,
Date
K
, et al.
Protection against cholera from killed whole-cell oral cholera vaccines: a systematic review and meta-analysis
.
Lancet Infect Dis
2017
,
17
,
1080
8
. doi: https://doi.org/

67.

Baik
YO
,
Choi
SK
,
Kim
JW
,
Yang
JS
,
Kim
IY
,
Kim
CW
, et al.
Safety and immunogenicity assessment of an oral cholera vaccine through phase I clinical trial in Korea
.
J Korean Med Sci
2014
,
29
,
494
501
. doi: https://doi.org/

68.

Kim
YCDR.
Immunogenicity of the Bivalent Killed, Whole-Cell, Oral Cholera Vaccine: A Meta-analysis
.
Baltimore, MD, USA
:
American Society of Tropical Medicine and Hygiene
,
2017
.

69.

Baik
YO
,
Choi
SK
,
Olveda
RM
,
Espos
RA
,
Ligsay
AD
,
Montellano
MB
, et al.
A randomized, non-inferiority trial comparing two bivalent killed, whole cell, oral cholera vaccines (Euvichol vs Shanchol) in the Philippines
.
Vaccine
2015
,
33
,
6360
5
. doi: https://doi.org/

70.

Chowdhury
F
,
Akter
A
,
Bhuiyan
TR
,
Tauheed
I
,
Teshome
S
,
Sil
A
, et al.
A non-inferiority trial comparing two killed, whole cell, oral cholera vaccines (Cholvax vs. Shanchol) in Dhaka, Bangladesh
.
Vaccine
2022
,
40
,
640
9
. doi: https://doi.org/

71.

Song
KR
,
Chapagain
RH
,
Tamrakar
D
,
Shrestha
R
,
Kanodia
P
,
Chaudhary
S
, et al.
Safety and immunogenicity of the Euvichol-S oral cholera vaccine for prevention of Vibrio cholerae O1 infection in Nepal: an observer-blind, active-controlled, randomised, non-inferiority, phase 3 trial
.
Lancet Glob Health
2024
,
12
,
e826
37
. doi: https://doi.org/

72.

Desai
SN
,
Akalu
Z
,
Teferi
M
,
Manna
B
,
Teshome
S
,
Park
JY
, et al.
Comparison of immune responses to a killed bivalent whole cell oral cholera vaccine between endemic and less endemic settings
.
Trop Med Int Health
2016
,
21
,
194
201
. doi: https://doi.org/

73.

Rijkers
GT
,
Sanders
EA
,
Breukels
MA
,
Zegers
BJ.
Infant B cell responses to polysaccharide determinants
.
Vaccine
1998
,
16
,
1396
400
. doi: https://doi.org/

74.

Black
RE
,
Levine
MM
,
Clements
ML
,
Young
CR
,
Svennerholm
AM
,
Holmgren
J.
Protective efficacy in humans of killed whole-vibrio oral cholera vaccine with and without the B subunit of cholera toxin
.
Infect Immun
1987
,
55
,
1116
20
. doi: https://doi.org/

75.

Clemens
JD
,
Harris
JR
,
Sack
DA
,
Chakraborty
J
,
Ahmed
F
,
Stanton
BF
, et al.
Field trial of oral cholera vaccines in Bangladesh: results of one year of follow-up
.
J Infect Dis
1988
,
158
,
60
9
. doi: https://doi.org/

76.

Clemens
JD
,
Sack
DA
,
Harris
JR
,
Chakraborty
J
,
Khan
MR
,
Stanton
BF
, et al.
Field trial of oral cholera vaccines in Bangladesh
.
Lancet
1986
,
2
,
124
7
. doi: https://doi.org/

77.

Sanchez
JL
,
Vasquez
B
,
Begue
RE
,
Meza
R
,
Castellares
G
,
Cabezas
C
, et al.
Protective efficacy of oral whole-cell/recombinant-B-subunit cholera vaccine in Peruvian military recruits
.
Lancet
1994
,
344
,
1273
6
. doi: https://doi.org/

78.

Taylor
DN
,
Cardenas
V
,
Sanchez
JL
,
Bégué
RE
,
Gilman
R
,
Bautista
C
, et al.
Two-year study of the protective efficacy of the oral whole cell plus recombinant B subunit cholera vaccine in Peru
.
J Infect Dis
2000
,
181
,
1667
73
. doi: https://doi.org/

79.

Lucas
ME
,
Deen
JL
,
von Seidlein
L
,
Wang
X-Y
,
Ampuero
J
,
Puri
M
, et al.
Effectiveness of mass oral cholera vaccination in Beira, Mozambique
.
N Engl J Med
2005
,
352
,
757
67
. doi: https://doi.org/

80.

Khatib
AM
,
Ali
M
,
von Seidlein
L
,
Kim
DR
,
Hashim
R
,
Reyburn
R
, et al.
Effectiveness of an oral cholera vaccine in Zanzibar: findings from a mass vaccination campaign and observational cohort study
.
Lancet Infect Dis
2012
,
12
,
837
44
. doi: https://doi.org/

81.

Kaper
JB
,
Levine
MM.
Recombinant attenuated Vibrio cholerae strains used as live oral vaccines
.
Res Microbiol
1990
,
141
,
901
6
. doi: https://doi.org/

82.

Levine
MM
,
Kaper
JB
,
Herrington
D
,
Ketley
J
,
Losonsky
G
,
Tacket
CO
, et al.
Safety, immunogenicity, and efficacy of recombinant live oral cholera vaccines, CVD 103 and CVD 103-HgR
.
Lancet
1988
,
2
,
467
70
. doi: https://doi.org/

83.

Chen
WH
,
Cohen
MB
,
Kirkpatrick
BD
,
Brady
RC
,
Galloway
D
,
Gurwith
M
, et al.
Single-dose live oral cholera vaccine CVD 103-HgR protects against human experimental infection with Vibrio cholerae O1 El Tor
.
Clin Infect Dis
2016
,
62
,
1329
35
. doi: https://doi.org/

84.

Chen
WH
,
Greenberg
RN
,
Pasetti
MF
,
Livio
S
,
Lock
M
,
Gurwith
M
, et al.
Safety and immunogenicity of single-dose live oral cholera vaccine strain CVD 103-HgR, prepared from new master and working cell banks
.
Clin Vaccine Immunol
2014
,
21
,
66
73
. doi: https://doi.org/

85.

Calain
P
,
Chaine
JP
,
Johnson
E
,
Hawley
M-L
,
O’Leary
MJ
,
Oshitani
H
, et al.
Can oral cholera vaccination play a role in controlling a cholera outbreak
?
Vaccine
2004
,
22
,
2444
51
. doi: https://doi.org/

86.

Richie
EE
,
Punjabi
NH
,
Sidharta
YY
,
Peetosutan
KK
,
Sukandar
MM
,
Wasserman
SS
, et al.
Efficacy trial of single-dose live oral cholera vaccine CVD 103-HgR in North Jakarta, Indonesia, a cholera-endemic area
.
Vaccine
2000
,
18
,
2399
410
. doi: https://doi.org/

87.

Otero
CE
,
Langel
SN
,
Blasi
M
,
Permar
SR.
Maternal antibody interference contributes to reduced rotavirus vaccine efficacy in developing countries
.
PLoS Pathog
2020
,
16
,
e1009010
. doi: https://doi.org/

88.

Becker-Dreps
S
,
Vilchez
S
,
Velasquez
D
,
Moon
S-S
,
Hudgens
MG
,
Zambrana
LE
, et al.
Rotavirus-specific IgG antibodies from mothers’ serum may inhibit infant immune responses to the pentavalent rotavirus vaccine
.
Pediatr Infect Dis J
2015
,
34
,
115
6
. doi: https://doi.org/

89.

Appaiahgari
MB
,
Glass
R
,
Singh
S
,
Taneja
S
,
Rongsen-Chandola
T
,
Bhandari
N
, et al.
Transplacental rotavirus IgG interferes with immune response to live oral rotavirus vaccine ORV-116E in Indian infants
.
Vaccine
2014
,
32
,
651
6
. doi: https://doi.org/

90.

Moon
SS
,
Groome
MJ
,
Velasquez
DE
,
Parashar
UD
,
Jones
S
,
Koen
A
, et al.
Prevaccination rotavirus serum IgG and IgA are associated with lower immunogenicity of live, oral human rotavirus vaccine in South African infants
.
Clin Infect Dis
2016
,
62
,
157
65
. doi: https://doi.org/

91.

Chan
J
,
Nirwati
H
,
Triasih
R
,
Bogdanovic-Sakran
N
,
Soenarto
Y
,
Hakimi
M
, et al.
Maternal antibodies to rotavirus: could they interfere with live rotavirus vaccines in developing countries
?
Vaccine
2011
,
29
,
1242
7
. doi: https://doi.org/

92.

Sabin
AB
,
Michaels
RH
,
Ziring
P
,
Krugman
S
,
Warren
J.
Effect of oral poliovirus vaccine in newborn children. II. Intestinal resistance and antibody response at 6 months in children fed type I vaccine at birth
.
Pediatrics
1963
,
31
,
641
50
.

93.

Warren
RJ
,
Lepow
ML
,
Bartsch
GE
,
Robbins
FC.
The relationship of maternal antibody, breast feeding, and age to the susceptibility of newborn infants to infection with attenuated polioviruses
.
Pediatrics
1964
,
34
,
4
13
.

94.

Estivariz
CF
,
Anand
A
,
Gary
HE
Jr
,
Rahman
M
,
Islam
J
,
Bari
TI
, et al.
Immunogenicity of three doses of bivalent, trivalent, or type 1 monovalent oral poliovirus vaccines with a 2 week interval between doses in Bangladesh: an open-label, non-inferiority, randomised, controlled trial
.
Lancet Infect Dis
2015
,
15
,
898
904
. doi: https://doi.org/

95.

Asturias
EJ
,
Dueger
EL
,
Omer
SB
,
Melville
A
,
Nates
SV
,
Laassri
M
, et al.
Randomized trial of inactivated and live polio vaccine schedules in Guatemalan infants
.
J Infect Dis
2007
,
196
,
692
8
. doi: https://doi.org/

96.

Oguti
B
,
Ali
A
,
Andrews
N
,
Barug
D
,
Anh Dang
D
,
Halperin
SA
, et al.
The half-life of maternal transplacental antibodies against diphtheria, tetanus, and pertussis in infants: an individual participant data meta-analysis
.
Vaccine
2022
,
40
,
450
8
. doi: https://doi.org/

97.

Gruber
JF
,
Gruber
LM
,
Weber
RP
,
Becker-Dreps
S
,
Jonsson Funk
M.
Rotavirus vaccine schedules and vaccine response among infants in low- and middle-income countries: a systematic review
.
Open Forum Infect Dis
2017
,
4
,
ofx066
. doi: https://doi.org/

98.

Colgate
ER
,
Haque
R
,
Dickson
DM
,
Carmolli
MP
,
Mychaleckyj
JC
,
Nayak
U
, et al.
Delayed dosing of oral rotavirus vaccine demonstrates decreased risk of rotavirus gastroenteritis associated with serum zinc: a randomized controlled trial
.
Clin Infect Dis
2016
,
63
,
634
41
. doi: https://doi.org/

99.

Lee
B.
Update on rotavirus vaccine underperformance in low- to middle-income countries and next-generation vaccines
.
Human Vaccin Immunother
2021
,
17
,
1787
802
. doi: https://doi.org/

100.

Sereme
Y
,
Toumi
E
,
Saifi
E
,
Faury
H
,
Skurnik
D.
Maternal immune factors involved in the prevention or facilitation of neonatal bacterial infections
.
Cell Immunol
2024
,
395-396
,
104796
. doi: https://doi.org/

101.

Ballard
O
,
Morrow
AL.
Human milk composition: nutrients and bioactive factors
.
Pediatr Clin North Am
2013
,
60
,
49
74
. doi: https://doi.org/

102.

Ray
PG
,
Kelkar
SD.
Prevalence of neutralizing antibodies against different rotavirus serotypes in children with severe rotavirus-induced diarrhea and their mothers
.
Clin Diagn Lab Immunol
2004
,
11
,
186
94
. doi: https://doi.org/

103.

Losonsky
GA
,
D’Alessandra de Rimer
H.
Rotavirus specific breast milk antibody in two populations and possible correlates of protection
.
Adv Exp Med Biol
1991
,
310
,
265
9
. doi: https://doi.org/

104.

Brussow
H
,
Benitez
O
,
Uribe
F
,
Sidoti
J
,
Rosa
K
,
Cravioto
A.
Rotavirus-inhibitory activity in serial milk samples from Mexican women and rotavirus infections in their children during their first year of life
.
J Clin Microbiol
1993
,
31
,
593
7
. doi: https://doi.org/

105.

Moon
SS
,
Wang
Y
,
Shane
AL
,
Nguyen
T
,
Ray
P
,
Dennehy
P
, et al.
Inhibitory effect of breast milk on infectivity of live oral rotavirus vaccines
.
Pediatr Infect Dis J
2010
,
29
,
919
23
. doi: https://doi.org/

106.

Moon
SS
,
Tate
JE
,
Ray
P
,
Dennehy
PH
,
Archary
D
,
Coutsoudis
A
, 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: https://doi.org/

107.

Chilengi
R
,
Simuyandi
M
,
Beach
L
,
Mwila
K
,
Becker-Dreps
S
,
Emperador
DM
, et al.
Association of maternal immunity with rotavirus vaccine immunogenicity in Zambian infants
.
PLoS One
2016
,
11
,
e0150100
. doi: https://doi.org/

108.

Trang
NV
,
Braeckman
T
,
Lernout
T
,
Hau
VTB
,
Anh
Le T K
,
Luan
Le T
, et al.
Prevalence of rotavirus antibodies in breast milk and inhibitory effects to rotavirus vaccines
.
Human Vaccin Immunother
2014
,
10
,
3681
7
. doi: https://doi.org/

109.

Rongsen-Chandola
T
,
Strand
TA
,
Goyal
N
,
Flem
E
,
Rathore
SS
,
Arya
A
, et al.
Effect of withholding breastfeeding on the immune response to a live oral rotavirus vaccine in North Indian infants
.
Vaccine
2014
,
32
,
A134
9
. doi: https://doi.org/

110.

Groome
MJ
,
Moon
SS
,
Velasquez
D
,
Jones
S
,
Koen
A
,
van Niekerk
N
, et al.
Effect of breastfeeding on immunogenicity of oral live-attenuated human rotavirus vaccine: a randomized trial in HIV-uninfected infants in Soweto, South Africa
.
Bull World Health Organ
2014
,
92
,
238
45
. doi: https://doi.org/

111.

Ali
A
,
Kazi
AM
,
Cortese
MM
,
Fleming
JA
,
Moon
SS
,
Parashar
UD
, et al.
Impact of withholding breastfeeding at the time of vaccination on the immunogenicity of oral rotavirus vaccine--a randomized trial
.
PLoS One
2015
,
10
,
e0127622
. doi: https://doi.org/

112.

Parker
EPK
,
Bronowski
C
,
Sindhu
KNC
,
Babji
S
,
Benny
B
,
Carmona-Vicente
N
, et al.
Impact of maternal antibodies and microbiota development on the immunogenicity of oral rotavirus vaccine in African, Indian, and European infants
.
Nat Commun
2021
,
12
,
7288
. doi: https://doi.org/

113.

Plotkin
SA
,
Katz
M
,
Brown
RE
,
Pagano
JS.
Oral poliovirus vaccination in newborn African infants. The inhibitory effect of breast feeding
.
Am J Dis Child
1966
,
111
,
27
30
. doi: https://doi.org/

114.

Domok
I
,
Balayan
MS
,
Fayinka
OA
,
Skrtic
N
,
Soneji
AD
,
Harland
PS.
Factors affecting the efficacy of live poliovirus vaccine in warm climates. Efficacy of type 1 Sabin vaccine administered together with antihuman gamma-globulin horse serum to breast-fed and artificially fed infants in Uganda
.
Bull World Health Organ
1974
,
51
,
333
47
.

115.

Pan
WK
,
Seidman
JC
,
Ali
A
,
Hoest
C
,
Mason
C
,
Mondal
D
, et al.
Oral polio vaccine response in the MAL-ED birth cohort study: Considerations for polio eradication strategies
.
Vaccine
2019
,
37
,
352
65
. doi: https://doi.org/

116.

Naylor
C
,
Lu
M
,
Haque
R
,
Mondal
D
,
Buonomo
E
,
Nayak
U
, et al.
Environmental enteropathy, oral vaccine failure and growth faltering in infants in Bangladesh
.
eBioMedicine
2015
,
2
,
1759
66
. doi: https://doi.org/

117.

Haque
R
,
Snider
C
,
Liu
Y
,
Ma
JZ
,
Liu
L
,
Nayak
U
, et al.
Oral polio vaccine response in breast fed infants with malnutrition and diarrhea
.
Vaccine
2014
,
32
,
478
82
. doi: https://doi.org/

118.

Ahmed
T
,
Svennerholm
AM
,
Al Tarique
A
,
Sultana
GN
,
Qadri
F.
Enhanced immunogenicity of an oral inactivated cholera vaccine in infants in Bangladesh obtained by zinc supplementation and by temporary withholding breast-feeding
.
Vaccine
2009
,
27
,
1433
9
. doi: https://doi.org/

119.

Becker-Dreps
S
,
Choi
WS
,
Stamper
L
,
Vilchez
S
,
Velasquez
DE
,
Moon
S-S
, et al.
Innate immune factors in mothers’ breast milk and their lack of association with rotavirus vaccine immunogenicity in Nicaraguan Infants
.
J. Pediatric Infect. Dis. Soc.
2017
,
6
,
87
90
. doi: https://doi.org/

120.

Mwila-Kazimbaya
K
,
Garcia
MP
,
Bosomprah
S
,
Laban
NM
,
Chisenga
CC
,
Permar
SR
, et al.
Effect of innate antiviral glycoproteins in breast milk on seroconversion to rotavirus vaccine (Rotarix) in children in Lusaka, Zambia
.
PLoS One
2017
,
12
,
e0189351
. doi: https://doi.org/

121.

Ramani
S
,
Stewart
CJ
,
Laucirica
DR
,
Ajami
NJ
,
Robertson
B
,
Autran
CA
, et al.
Human milk oligosaccharides, milk microbiome and infant gut microbiome modulate neonatal rotavirus infection
.
Nat Commun
2018
,
9
,
5010
. doi: https://doi.org/

122.

Estorninos
E
,
Lawenko
RB
,
Palestroque
E
,
Sprenger
N
,
Benyacoub
J
,
Kortman
GAM
, et al.
Term infant formula supplemented with milk-derived oligosaccharides shifts the gut microbiota closer to that of human milk-fed infants and improves intestinal immune defense: a randomized controlled trial
.
Am J Clin Nutr
2022
,
115
,
142
53
. doi: https://doi.org/

123.

Sharma
S
,
Nordgren
J.
Effect of infant and maternal secretor status on rotavirus vaccine take-an overview
.
Viruses
2021
,
13
,
1144
. doi: https://doi.org/

124.

Burke
RM
,
Payne
DC
,
McNeal
M
,
Conrey
SC
,
Burrell
AR
,
Mattison
CP
, et al.
Correlates of rotavirus vaccine shedding and seroconversion in a U.S. cohort of healthy infants
.
J Infect Dis
2024
,
230
,
754
62
. doi: https://doi.org/

125.

Middleton
BF
,
Danchin
M
,
Cunliffe
NA
,
Jones
MA
,
Boniface
K
,
Kirkwood
CD
, et al.
Histo-blood group antigen profile of Australian Aboriginal children and seropositivity following oral rotavirus vaccination
.
Vaccine
2023
,
41
,
3579
83
. doi: https://doi.org/

126.

Magwira
CA
,
Kgosana
LP
,
Esona
MD
,
Seheri
ML.
Low fecal rotavirus vaccine virus shedding is significantly associated with non-secretor histo-blood group antigen phenotype among infants in northern Pretoria, South Africa
.
Vaccine
2020
,
38
,
8260
3
. doi: https://doi.org/

127.

Armah
GE
,
Cortese
MM
,
Dennis
FE
,
Yu
Y
,
Morrow
AL
,
McNeal
MM
, et al.
Rotavirus vaccine take in infants is associated with secretor status
.
J Infect Dis
2019
,
219
,
746
9
. doi: https://doi.org/

128.

Kazi
AM
,
Cortese
MM
,
Yu
Y
,
Lopman
B
,
Morrow
AL
,
Fleming
JA
, et al.
Secretor and salivary ABO blood group antigen status predict rotavirus vaccine take in infants
.
J Infect Dis
2017
,
215
,
786
9
. doi: https://doi.org/

129.

Cantelli
CP
,
Velloso
AJ
,
Assis
RMS
,
Barros
JJ
,
Mello
FCA
,
Cunha
DC
, et al.
Rotavirus A shedding and HBGA host genetic susceptibility in a birth community-cohort, Rio de Janeiro, Brazil, 2014-2018
.
Sci Rep
2020
,
10
,
6965
. doi: https://doi.org/

130.

Pollock
L
,
Bennett
A
,
Jere
KC
,
Dube
Q
,
Mandolo
J
,
Bar-Zeev
N
, et al.
Nonsecretor histo-blood group antigen phenotype is associated with reduced risk of clinical rotavirus vaccine failure in Malawian infants
.
Clin Infect Dis
2019
,
69
,
1313
9
. doi: https://doi.org/

131.

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
2018
,
8
,
1502
. doi: https://doi.org/

132.

Bucardo
F
,
Reyes
Y
,
Ronnelid
Y
,
González
F
,
Sharma
S
,
Svensson
L
, et al.
Histo-blood group antigens and rotavirus vaccine shedding in Nicaraguan infants
.
Sci Rep
2019
,
9
,
10764
. doi: https://doi.org/

133.

El-Heneidy
A
,
Cheung
C
,
Lambert
SB
,
Wang
CYT
,
Whiley
DM
,
Sly
PD
, et al.
Histo-blood group antigens and rotavirus vaccine virus shedding in Australian infants
.
Pathology (Phila)
2022
,
54
,
928
34
. doi: https://doi.org/

134.

Williams
FB
,
Kader
A
,
Colgate
ER
,
Dickson
DM
,
Carmolli
M
,
Uddin
MI
, et al.
Maternal secretor status affects oral rotavirus vaccine response in breastfed infants in Bangladesh
.
J Infect Dis
2021
,
224
,
1147
51
. doi: https://doi.org/

135.

Parker
EPK
,
Whitfield
H
,
Baskar
C
,
Giri
S
,
John
J
,
Grassly
NC
, et al.
FUT2 secretor status is not associated with oral poliovirus vaccine immunogenicity in South Indian infants
.
J Infect Dis
2019
,
219
,
578
81
. doi: https://doi.org/

136.

Chisenga
CC
,
Bosomprah
S
,
Chilyabanyama
ON
,
Alabi
P
,
Simuyandi
M
,
Mwaba
J
, et al.
Assessment of the influence of ABO blood groups on oral cholera vaccine immunogenicity in a cholera endemic area in Zambia
.
BMC Public Health
2023
,
23
,
152
. doi: https://doi.org/

137.

Ramamurthy
T
,
Wagener
D
,
Chowdhury
G
,
Majumder
PP.
A large study on immunological response to a whole-cell killed oral cholera vaccine reveals that there are significant geographical differences in response and that O blood group individuals do not elicit a higher response
.
Clin Vaccine Immunol
2010
,
17
,
1232
7
. doi: https://doi.org/

138.

Lagos
R
,
Avendano
A
,
Prado
V
,
Horwitz
I
,
Wasserman
S
,
Losonsky
G
, et al.
Attenuated live cholera vaccine strain CVD 103-HgR elicits significantly higher serum vibriocidal antibody titers in persons of blood group O
.
Infect Immun
1995
,
63
,
707
9
. doi: https://doi.org/

139.

Clemens
JD
,
Sack
DA
,
Harris
JR
,
Chakraborty
J
,
Khan
MR
,
Huda
S
, et al.
ABO blood groups and cholera: new observations on specificity of risk and modification of vaccine efficacy
.
J Infect Dis
1989
,
159
,
770
3
. doi: https://doi.org/

140.

Andersen
NB
,
Larsen
SM
,
Nissen
SK
,
Jørgensen
SE
,
Mardahl
M
,
Christiansen
M
, et al.
Host genetics, innate immune responses, and cellular death pathways in poliomyelitis patients
.
Front Microbiol
2019
,
10
,
1495
. doi: https://doi.org/

141.

Karlsson
EK
,
Harris
JB
,
Tabrizi
S
,
Rahman
A
,
Shlyakhter
I
,
Patterson
N
, et al.
Natural selection in a Bangladeshi population from the cholera-endemic Ganges river delta
.
Sci Transl Med
2013
,
5
,
192ra86
. doi: https://doi.org/

142.

Burnett
E
,
Parashar
UD
,
Tate
JE.
Rotavirus infection, illness, and vaccine performance in malnourished children: a review of the literature
.
Pediatr Infect Dis J
2021
,
40
,
930
6
. doi: https://doi.org/

143.

Bahl
S
,
Estívariz
CF
,
Sutter
RW
,
Sarkar
BK
,
Verma
H
,
Jain
V
, et al.
Cross-sectional serologic assessment of immunity to poliovirus infection in high-risk areas of northern India
.
J Infect Dis
2014
,
210
,
S243
51
. doi: https://doi.org/

144.

Saleem
AF
,
Mach
O
,
Quadri
F
,
Khan
A
,
Bhatti
Z
,
Rehman
NU
, et al.
Immunogenicity of poliovirus vaccines in chronically malnourished infants: A randomized controlled trial in Pakistan
.
Vaccine
2015
,
33
,
2757
63
. doi: https://doi.org/

145.

Uddin
MI
,
Islam
S
,
Nishat
NS
,
Hossain
M
,
Rafique
TA
,
Rashu
R
, et al.
Biomarkers of environmental enteropathy are positively associated with immune responses to an oral cholera vaccine in Bangladeshi children
.
PLoS NeglTrop Dis
2016
,
10
,
e0005039
. doi: https://doi.org/

146.

Glass
RI
,
Svennerholm
AM
,
Stoll
BJ
,
Khan
MR
,
Huda
S
,
Huq
MI
, et al.
Effects of undernutrition on infection with Vibrio cholerae O1 and on response to oral cholera vaccine
.
Pediatr Infect Dis J
1989
,
8
,
105
9
.

147.

Church
JA
,
Parker
EP
,
Kirkpatrick
BD
,
Grassly
NC
,
Prendergast
AJ.
Interventions to improve oral vaccine performance: a systematic review and meta-analysis
.
Lancet Infect Dis
2019
,
19
,
203
14
. doi: https://doi.org/

148.

Church
JA
,
Rukobo
S
,
Govha
M
,
Carmolli
MP
,
Diehl
SA
,
Chasekwa
B
, et al.
Neonatal vitamin A supplementation and immune responses to oral polio vaccine in Zimbabwean infants
.
Trans R Soc Trop Med Hyg
2019
,
113
,
110
5
. doi: https://doi.org/

149.

Das
R
,
Jobayer Chisti
M
,
Ahshanul Haque
M
,
Ashraful Alam
M
,
Das
S
,
Mahfuz
M
, et al.
Evaluating association of vaccine response to low serum zinc and vitamin D levels in children of a birth cohort study in Dhaka
.
Vaccine
2021
,
39
,
59
67
. doi: https://doi.org/

150.

Lazarus
RP
,
John
J
,
Shanmugasundaram
E
,
Rajan
AK
,
Thiagarajan
S
,
Giri
S
, et al.
The effect of probiotics and zinc supplementation on the immune response to oral rotavirus vaccine: A randomized, factorial design, placebo-controlled study among Indian infants
.
Vaccine
2018
,
36
,
273
9
. doi: https://doi.org/

151.

Habib
MA
,
Soofi
S
,
Sheraz
A
,
Bhatti
ZS
,
Okayasu
H
,
Zaidi
SZ
, et al.
Zinc supplementation fails to increase the immunogenicity of oral poliovirus vaccine: a randomized controlled trial
.
Vaccine
2015
,
33
,
819
25
. doi: https://doi.org/

152.

Albert
MJ
,
Qadri
F
,
Wahed
MA
,
Ahmed
T
,
Rahman
ASMH
,
Ahmed
F
, et al.
Supplementation with zinc, but not vitamin A, improves seroconversion to vibriocidal antibody in children given an oral cholera vaccine
.
J Infect Dis
2003
,
187
,
909
13
. doi: https://doi.org/

153.

Karlsen
TH
,
Sommerfelt
H
,
Klomstad
S
,
Andersen
PK
,
Strand
TA
,
Ulvik
RJ
, et al.
Intestinal and systemic immune responses to an oral cholera toxoid B subunit whole-cell vaccine administered during zinc supplementation
.
Infect Immun
2003
,
71
,
3909
13
. doi: https://doi.org/

154.

Becker-Dreps
S
,
Vilchez
S
,
Bucardo
F
,
Twitchell
E
,
Choi
WS
,
Hudgens
MG
, et al.
The association between fecal biomarkers of environmental enteropathy and rotavirus vaccine response in Nicaraguan infants
.
Pediatr Infect Dis J
2017
,
36
,
412
6
. doi: https://doi.org/

155.

Mwila-Kazimbaya
K
,
Bosomprah
S
,
Chilyabanyama
ON
,
Chisenga
CC
,
Chibuye
M
,
Laban
NM
, et al.
Association of biomarkers of enteric dysfunction, systemic inflammation, and growth hormone resistance with seroconversion to oral rotavirus vaccine: A lasso for inference approach
.
PLoS One
2023
,
18
,
e0293101
. doi: https://doi.org/

156.

Kosek
MN
,
Mduma
E
,
Kosek
PS
,
Lee
GO
,
Svensen
E
,
Pan
WKY
, 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
2016
,
95
,
928
37
. doi: https://doi.org/

157.

Lagos
R
,
Fasano
A
,
Wasserman
SS
,
Prado
V
,
San Martin
O
,
Abrego
P
, et al.
Effect of small bowel bacterial overgrowth on the immunogenicity of single-dose live oral cholera vaccine CVD 103-HgR
.
J Infect Dis
1999
,
180
,
1709
12
. doi: https://doi.org/

158.

Mwape
I
,
Bosomprah
S
,
Mwaba
J
,
Mwila-Kazimbaya
K
,
Laban
NM
,
Chisenga
CC
, et al.
Immunogenicity of rotavirus vaccine (RotarixTM) in infants with environmental enteric dysfunction
.
PLoS One
2017
,
12
,
e0187761
. doi: https://doi.org/

159.

Church
JA
,
Rukobo
S
,
Govha
M
,
Gough
EK
,
Chasekwa
B
,
Lee
B
, et al.
Associations between biomarkers of environmental enteric dysfunction and oral rotavirus vaccine immunogenicity in rural Zimbabwean infants
.
eClinicalMedicine
2021
,
41
,
101173
. doi: https://doi.org/

160.

Baker
JM
,
Tate
JE
,
Leon
J
,
Haber
MJ
,
Lopman
BA.
Antirotavirus IgA seroconversion rates in children who receive concomitant oral poliovirus vaccine: a secondary, pooled analysis of Phase II and III trial data from 33 countries
.
PLoS Med
2019
,
16
,
e1003005
. doi: https://doi.org/

161.

Emperador
DM
,
Velasquez
DE
,
Estivariz
CF
,
Lopman
B
,
Jiang
B
,
Parashar
U
, et al.
Interference of monovalent, bivalent, and trivalent oral poliovirus vaccines on monovalent rotavirus vaccine immunogenicity in rural Bangladesh
.
Clin Infect Dis
2015
,
62
,
150
6
. doi: https://doi.org/

162.

Steele
AD
,
De Vos
B
,
Tumbo
J
,
Reynders
J
,
Scholtz
F
,
Bos
P
, et al.
Co-administration study in South African infants of a live-attenuated oral human rotavirus vaccine (RIX4414) and poliovirus vaccines
.
Vaccine
2010
,
28
,
6542
8
. doi: https://doi.org/

163.

Li
R-c
,
Huang
T
,
Li
Y
, et al.
Immunogenicity and reactogenicity of the human rotavirus vaccine, RIX4414 oral suspension, when co-administered with routine childhood vaccines in Chinese infants
.
Human Vaccin Immunother
2016
,
12
,
785
93
.

164.

Zaman
K
,
Sack
DA
,
Yunus
M
,
Arifeen
SE
,
Podder
G
,
Azim
T
, et al.
Successful co-administration of a human rotavirus and oral poliovirus vaccines in Bangladeshi infants in a 2-dose schedule at 12 and 16 weeks of age
.
Vaccine
2009
,
27
,
1333
9
. doi: https://doi.org/

165.

Patel
M
,
Steele
AD
,
Parashar
UD.
Influence of oral polio vaccines on performance of the monovalent and pentavalent rotavirus vaccines
.
Vaccine
2012
,
30
,
A30
5
. doi: https://doi.org/

166.

Cowley
D
,
Sari
RM
,
Handley
A
,
Watts
E
,
Bachtiar
NS
,
At Thobari
J
, et al.
Immunogenicity of four doses of oral poliovirus vaccine when co-administered with the human neonatal rotavirus vaccine (RV3-BB)
.
Vaccine
2019
,
37
,
7233
9
. doi: https://doi.org/

167.

Mo
Z
,
Ma
X
,
Luo
P
,
Mo
Y
,
Kaplan
SS
,
Shou
Q
, et al.
Immunogenicity of pentavalent rotavirus vaccine in Chinese infants
.
Vaccine
2019
,
37
,
1836
43
. doi: https://doi.org/

168.

Desai
S
,
Rathi
N
,
Kawade
A
,
Venkatramanan
P
,
Kundu
R
,
Lalwani
SK
, et al.
Non-interference of Bovine-Human reassortant pentavalent rotavirus vaccine ROTASIIL® with the immunogenicity of infant vaccines in comparison with a licensed rotavirus vaccine
.
Vaccine
2018
,
36
,
5519
23
. doi: https://doi.org/

169.

Chandola
TR
,
Taneja
S
,
Goyal
N
,
Antony
K
,
Bhatia
K
,
More
D
, et al.
ROTAVAC((R)) does not interfere with the immune response to childhood vaccines in Indian infants: a randomized placebo controlled trial
.
Heliyon
2017
,
3
,
e00302
. doi: https://doi.org/

170.

Islam
MT
,
Date
K
,
Khan
AI
,
Bhuiyan
TR
,
Khan
ZH
,
Ahmed
S
, et al.
Co-administration of oral cholera vaccine with oral polio vaccine among Bangladeshi young children: a randomized controlled open label trial to assess interference
.
Clin Infect Dis
2022
,
76
,
263
70
. doi: https://doi.org/

171.

Taniuchi
M
,
Platts-Mills
JA
,
Begum
S
,
Uddin
MJ
,
Sobuz
SU
,
Liu
J
, et al.
Impact of enterovirus and other enteric pathogens on oral polio and rotavirus vaccine performance in Bangladeshi infants
.
Vaccine
2016
,
34
,
3068
75
. doi: https://doi.org/

172.

Kim
AH
,
Armah
G
,
Dennis
F
,
Wang
L
,
Rodgers
R
,
Droit
L
, et al.
Enteric virome negatively affects seroconversion following oral rotavirus vaccination in a longitudinally sampled cohort of Ghanaian infants
.
Cell Host Microbe
2022
,
30
,
110
23.e5
. doi: https://doi.org/

173.

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
2014
,
210
,
853
64
. doi: https://doi.org/

174.

Praharaj
I
,
Parker
EPK
,
Giri
S
,
Allen
DJ
,
Silas
S
,
Revathi
R
, et al.
Influence of nonpolio enteroviruses and the bacterial gut microbiota on oral poliovirus vaccine response: a study from South India
.
J Infect Dis
2019
,
219
,
1178
86
. doi: https://doi.org/

175.

Harris
V
,
Ali
A
,
Fuentes
S
,
Korpela
K
,
Kazi
M
,
Tate
J
, et al.
Rotavirus vaccine response correlates with the infant gut microbiota composition in Pakistan
.
Gut Microbes
2018
,
9
,
93
101
. doi: https://doi.org/

176.

Parker
EPK
,
Praharaj
I
,
Zekavati
A
,
Lazarus
RP
,
Giri
S
,
Operario
DJ
, et al.
Influence of the intestinal microbiota on the immunogenicity of oral rotavirus vaccine given to infants in south India
.
Vaccine
2018
,
36
,
264
72
. doi: https://doi.org/

177.

Huda
MN
,
Lewis
Z
,
Kalanetra
KM
,
Rashid
M
,
Ahmad
SM
,
Raqib
R
, et al.
Stool microbiota and vaccine responses of infants
.
Pediatrics
2014
,
134
,
e362
72
. doi: https://doi.org/

178.

Zhao
T
,
Li
J
,
Fu
Y
,
Ye
H
,
Liu
X
,
Li
G
, et al.
Influence of gut microbiota on mucosal IgA antibody response to the polio vaccine
.
npj Vaccines
2020
,
5
,
47
. doi: https://doi.org/

179.

Tan
SK
,
Granados
AC
,
Bouquet
J
,
Hoy-Schulz
YE
,
Green
L
,
Federman
S
, et al.
Metagenomic sequencing of stool samples in Bangladeshi infants: virome association with poliovirus shedding after oral poliovirus vaccination
.
Sci Rep
2020
,
10
,
15392
. doi: https://doi.org/

180.

Chac
D
,
Bhuiyan
TR
,
Saha
A
,
Alam
MM
,
Salma
U
,
Jahan
N
, et al.
Gut microbiota and development of Vibrio cholerae-specific long-term memory B cells in adults after whole-cell killed oral cholera vaccine
.
Infect Immun
2021
,
89
,
e0021721
. doi: https://doi.org/

181.

Harris
JB
,
Podolsky
MJ
,
Bhuiyan
TR
,
Chowdhury
F
,
Khan
AI
,
Larocque
RC
, et al.
Immunologic responses to Vibrio cholerae in patients co-infected with intestinal parasites in Bangladesh
.
PLoS NeglTrop Dis
2009
,
3
,
e403
. doi: https://doi.org/

182.

Lee
B
,
Dickson
DM
,
Alam
M
,
Afreen
S
,
Kader
A
,
Afrin
F
, et al.
The effect of increased inoculum on oral rotavirus vaccine take among infants in Dhaka, Bangladesh: a double-blind, parallel group, randomized, controlled trial
.
Vaccine
2020
,
38
,
90
9
. doi: https://doi.org/

183.

Middleton
BF
,
Fathima
P
,
Snelling
TL
,
Morris
P.
Systematic review of the effect of additional doses of oral rotavirus vaccine on immunogenicity and reduction in diarrhoeal disease among young children
.
eClinicalMedicine
2022
,
54
,
101687
. doi: https://doi.org/

184.

World Health Organization
.
Polio vaccines: WHO position paper – March, 2016 = Note de synthèse de l’OMS sur les vaccins antipoliomyélitiques – mars 2016
.
Weekly Epidemiol Record
2016
,
91
,
145
68
.

185.

Amin
AB
,
Tate
JE
,
Waller
LA
,
Lash
TL
,
Lopman
BA.
Monovalent rotavirus vaccine efficacy against different rotavirus genotypes: a pooled analysis of Phase II and III Trial Data
.
Clin Infect Dis
2023
,
76
,
e1150
6
. doi: https://doi.org/

186.

Cates
JE
,
Amin
AB
,
Tate
JE
,
Lopman
B
,
Parashar
U.
Do rotavirus strains affect vaccine effectiveness? A systematic review and meta-analysis
.
Pediatr Infect Dis J
2021
,
40
,
1135
43
. doi: https://doi.org/

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)