4,764
Views
22
CrossRef citations to date
0
Altmetric
Review

Vaccines against gastroenteritis, current progress and challenges

, &
Pages 1486-1517 | Received 23 Dec 2019, Accepted 12 May 2020, Published online: 18 Jun 2020

ABSTRACT

Enteric viral and bacterial infections continue to be a leading cause of mortality and morbidity in young children in low-income and middle-income countries, the elderly, and immunocompromised individuals. Vaccines are considered an effective and practical preventive approach against the predominantly fecal-to-oral transmitted gastroenteritis particularly in the resource-limited countries or regions where implementation of sanitation systems and supply of safe drinking water are not quickly achievable. While vaccines are available for a few enteric pathogens including rotavirus and cholera, there are no vaccines licensed for many other enteric viral and bacterial pathogens. Challenges in enteric vaccine development include immunological heterogeneity among pathogen strains or isolates, a lack of animal challenge models to evaluate vaccine candidacy, undefined host immune correlates to protection, and a low protective efficacy among young children in endemic regions. In this article, we briefly updated the progress and challenges in vaccines and vaccine development for the leading enteric viral and bacterial pathogens including rotavirus, human calicivirus, Shigella, enterotoxigenic Escherichia coli (ETEC), cholera, nontyphoidal Salmonella, and Campylobacter, and introduced a novel epitope- and structure-based vaccinology platform known as MEFA (multiepitope fusion antigen) and the application of MEFA for developing broadly protective multivalent vaccines against heterogenous pathogens.

Introduction

Enteric diseases remain a leading cause of mortality in young children in developing countries and morbidity in children worldwide.Citation1Citation5 Parasitic pathogens mainly Cryptosporidium spp., viral pathogens including rotavirus and norovirus, and bacterial pathogens Shigella, enterotoxigenic E. coli, Campylobacter, Vibrio cholerae, and Salmonella are the top causes of enteric infections.Citation2,Citation6 These enteric pathogens are mostly fecal-to-oral transmitted. Infections are typically initiated with ingestion of contaminated food and water, leading to diseases including nausea, abdominal cramps, fever, and more commonly diarrhea and dehydration. Enteric infections are self-eliminated in healthy adults but without medical intervention can progress to life-threatening conditions and often death in the young children and immune compromised patients. Implementation of community-wide sanitation systems and supply of safe drinking water would effectively prevent enteric infections. However, for many resource-limited countries, water, sanitation, and hygiene (WASH) is not a goal quickly achievable. Handwashing with soap especially antibacterial soap reduces enteric infections in the short term, but it increases the risk of antimicrobial resistance built up in enteric pathogens. Vaccination, on the other hand, is considered currently more effective and more practical to prevent enteric diseases. Unfortunately, there are no vaccines licensed for many pathogens associated with enteric infections.

Challenges in developing effective vaccines against enteric infections include heterogeneity among strains (genotypes, serotypes, or pathotypes) of individual enteric pathogens, a lack of suitable animal models to assess vaccine efficacy in prior to human subject studies, a poor understanding at details of pathogen pathogenesis or disease mechanism (nontyphoidal Salmonella or Campylobacter), undefined host immune correlates to protection, short-lived protective immunity, absence of suitable or cost-effective cell culture systems to attenuate viral pathogens (human Calicivirus), and inability to achieve satisfied efficacy among young children in endemic regions or countries. Genetic and antigenic heterogeneity is perhaps the primary roadblock in developing broadly effective vaccines for many enteric pathogens. Additionally, multiple pathogens are often present at individual patients with enteric infections. This further complicates diagnostics and disease treatment and demands development of combination vaccines against two or more than two enteric pathogens for effective protection. Combination (combo) vaccines become attractive since the current WHO EPI (expanded program for immunization) is getting increasingly crowded with continuous introduction of new vaccines. In this review, we outlined the current status and challenges of vaccines or vaccine candidates for rotavirus, calicivirus, Shigella, enterotoxigenic E. coli (ETEC), cholera, nontyphoidal Salmonella, and Campylobacter, the top causes of enteric infections based on molecular diagnostics,Citation2,Citation6 and described a new vaccine technology or vaccinology platform for the development of cross-protective multivalent vaccines.

I. Vaccines and vaccine development for viral enteric pathogens

Rotavirus (Rv)

A non-enveloped multi-layered virus is the top viral cause of enteric infections in children especially in countries or regions where rotavirus vaccines have not been introduced.Citation2,Citation7 Nine or ten rotavirus species (A to I or J) have been identified. Four species (A, B, C, H), but predominantly species A (referred as rotavirus hereby), cause human infections or gastroenteritis. Rotavirus genome is composed of 11 double-stranded RNA (dsRNA) segments which encode 11 to 12 proteins, six viral structural proteins (VP1-VP4, VP6, VP7) and five or six non-structural proteins (NSP1-NSP5, or NSP6). Structural proteins VP7 glycoprotein (G antigen; G genotypes) and VP4 protease-sensitive protein (P antigen; P genotypes) located at the outmost layer of rotavirus virion are the host cell-attachment proteins and induce neutralizing antibodies. Six VP7 G genotypes (G1, G2, G3, G4, G9, G12) and three VP4 P genotypes (P4, P6, P8) are the most important associated with human disease, with six combinations or serotypes (G1P[8], G2P[4], G3P[8], G4P[8], G9P[8] and G12P[8]) responsible for over 90% of the severe rotavirus infections.Citation8Citation14 Rotavirus is transmitted primarily by ingestion of contaminated water and food, as well as physical contact of virus carriers including patients and contaminated surfaces, often leading to clinical outcomes ranging from mild watery diarrhea to acute gastroenteritis, dehydration, hypovolemic shock, and death. Oral rehydration (oral rehydration solutions) is the treatment commonly recommended before the introduction of rotavirus vaccines.Citation15 Since the late 1990 s, several rotavirus vaccines have been introduced. Four vaccines have been prequalified or recommended by WHO for global licensing, and two are licensed by individual nations ().

Table 1. Licensed vaccines currently available for enteric pathogens.

Rotashield (human-rhesus RRV)

Rotashield is a quadrivalent vaccine product licensed in 1998. Rotashield consists of rhesus monkey backbone strain G3P[3] and three reassorted strains, with VP7 protein (reassorted) of G1, G2, and G4 to protect against VP7 serotypes G1 to G4. A three-dose regimen provided 70–90% efficacy against moderate-to-severe disease in infants.Citation9Citation11 However, this product was withdrawn from the market in 1999 due to a low risk of association with intestinal intussusception (IS).Citation16,Citation17 Later studies showed that association of Rotashield vaccination with IS was age-relatedCitation18 and that a regimen of two doses administered at the first and the second months yielded 64% protection without IS adverse effect.Citation19

RotaTeq (RV5)

RotaTeq is a pentavalent bovine-human reassorted vaccine developed by Merck. RotaTeq has bovine rotavirus strain G6P[5] as the backbone and is composed of five reassorted strains (G1P[5], G2P[5], G3P[5], G4P[5] and G6P[8]). A regimen of three doses provided 74% efficacy against any rotavirus disease and 98% efficacy against severe disease in Europe and USA.Citation20Citation23 However, efficacy in developing countries ranged from only 51% to 64% for the first year and 20% to 46% in the second year.Citation24Citation26

Rotarix (RV1, RIX4414)

Rotarix is a monovalent vaccine developed by GSK. Rotarix carries tissue culture passage-attenuated human rotavirus strain G1P[8]Citation27 which represents the most common VP4 and VP7 antigens of human rotavirus. At a regimen of two doses, Rotarix yielded similar efficacy as RotaTeq, ranging from 38% to 97% against moderate-to-severe gastroenteritis (a score at ≥7 of Ruuska and Vesikari 20-point scoring system) in different countries or regions.Citation28Citation43

ROTAVAC

ROTAVAC is a monovalent vaccine developed by Bharat Biotech in India. It carries live naturally attenuated strain 116E (G9P[11]) isolated from children without symptoms. ROTAVAC is currently licensed in India and is shown 56% and 49% protection against hospitalization in the first and second year of life, and 35% efficacy against any rotavirus infection at a regimen of three doses.Citation44Citation48

ROTASIIL

Currently licensed in India, ROTASIIL is a bovine-human reassorted pentavalent vaccine (G1[P5], G2[P5], G3P[5], G4P[5], G9P[5]).Citation49Citation51 Developed by Serum Institute of India, ROTASIIL showed 33% efficacy against severe rotavirus gastroenteritis in the first year.Citation52

Additionally, two vaccine products are currently licensed by individual countries. LLR-85 (Lanzhou Lamb Rotavirus Vaccine) based on a lamb strain (G10P[12]) is licensed in China and showed 35–78% efficacy.Citation53Citation56 Rotavin-M1, an attenuated human strain (G1P[8]) is licensed in Vietnam.Citation57,Citation58 Subunit rotavirus vaccines are also under development, including monovalent P2-VP8-P[8]Citation59 and trivalent VAC041 (P2-VP8-P[4]P[6]P[8]),Citation60,Citation61 and efficacy will need to be evaluated in field trials.

Human calicivirus (HuCV)

Unlike rotavirus for which a few vaccines have been prequalified globally or regionally by WHO or individual nations, there is no vaccine licensed for the single-stranded RNA norovirus or other human calicivirus. Norovirus indeed exceeds rotavirus for global disease burden and becomes the most common viral cause of gastroenteritis in developed countries where rotavirus vaccines have been introduced.Citation6,Citation62-Citation65 Norovirus RNA genome consists of three open reading frames (ORFs). One ORF encodes a polyprotein cleaved into seven non-structural proteins (NP1 to NP7), whereas the other two ORFs encode minor capsid protein VP2 and major capsid protein VP1. VP1 is composed of a conservative shell domain (S) and a variable protruding domain (P). Seven norovirus genogroups and 40 genotypes have been identified, among them three genogroups (GI, GII, and GIV) and 29 genotypes are associated with human gastroenteritis.Citation66,Citation67 However, genotype 4 in GII genogroup, GII.4, is by far the most prevalent and is responsible for a majority of acute gastroenteritis cases.Citation68 Therefore, GII.4 along with the initially identified Norwalk virus GI.1 are primarily targeted in norovirus vaccine development.

In contrast to rotavirus which exclusively infects children, norovirus causes gastroenteritis in people of all ages though mostly among young children and the elderly over 65 years. Like rotavirus, norovirus is mainly transmitted via ingestion of contaminated food and water, with outbreaks occur more commonly in close-contact settings including pre-school centers and schools, retirement care institutes, hospitals, as well as cruise ships. The typical clinical outcome is acute gastroenteritis but can be severe dehydration and death if infection progresses without intervention treatments.

There are several norovirus vaccine candidates under clinical or preclinical studies. Because norovirus is unculturable under current cell culture system, developing attenuated whole-cell vaccines becomes prohibitable. Consequently, calicivirus vaccine candidates under investigation are largely based on viral proteins. Norovirus major capsid protein VP1, when expressed in eukaryotic cells, forms virus-like particles (VLP) to exhibit antigenicity similarly to native viral particles. Additionally, the P domain of VP1 after being linked to a polypeptide and expressed in cell culture also aggregates into particles (P particle). Therefore, VP1 virus-like particles and P particles of global pandemic genotype GII.4 and the regional endemic GI genotypes are the primary antigens for norovirus vaccine development.

Bivalent GI.1/GII.4 VLP

GI.1/GII.4 VLP vaccine candidate under development by Takeda is a continuation of monovalent GI.1 VLP (Ligocyte) and is currently the one progressed furthest. GI.1 VLP was initially designed against Norwalk virus. When orally administered without adjuvant, GI.1 VLP showed 83% IgG seroconversion in volunteers.Citation69 Immunogenicity was enhanced when delivered intranasally with adjuvant monophosphoryl lipid A (MPL) and Al(OH)3 at a two-dose regimen.Citation70,Citation71 With GII.4 emerged as the most prevalent genotype, monovalent GI.1 VLP was improved to be a bivalent VLP candidate, by adding consensus GII.4 VLP and administered intramuscularly.Citation72 Intramuscularly immunized adult volunteers developed GI.1- and GII.4-specific antibodies. Interestingly, GI.1/GII.4 VLP-induced antibodies showed an increasing neutralizing activity – antibody blocking histo-blood group antigens (HBGA).Citation73Citation75 When challenged with a GII.4 NoV strain, the vaccinated group showed reduction of disease severity but unfortunately without significant protection against overall gastroenteritis.Citation76 The lack of protection appeared to be caused mainly by a lower attack rate from the challenge strain (37.5% infection rate), presumably caused by preexisting anti-NoV antibodies in volunteer adults resulted from natural exposure to NoV infections. Efficacy of bivalent GI.1/GII.4 VLP against recurrent infection or infection from heterogenous genotypes needs to be further evaluated.

Other vaccine candidates including trivalent GI.3/GII.4/rotavirus rVP6,Citation77,Citation78 P particle derived from a modified P domain of VP1,Citation79,Citation80 and adenovirus- or alphavirus-vector expressing VP1 proteinsCitation81,Citation82 are also under development.

Challenges in developing norovirus vaccines are nicely summarized in two articles.Citation67,Citation83 These challenges include virus heterogeneity and virus constant evolving, an absence of a conventional cell culture system, a lack of a small animal model, markers needed to separate host immune responses to infection versus vaccination, short-lived protective immunity, and the need to define immune correlates of protection to evaluate the efficacy of norovirus vaccine candidates. Currently, there are already three genogroups and 29 genotypes of noroviruses causing gastroenteritis. These viruses are continuously evolving through mutations as well as recombination from the circulating strains. That results in a constant emergence of new variants or strainsCitation84 and potential escape of host immunity derived from vaccination or natural infection. On the other hand, the most vulnerable populations to norovirus gastroenteritis, infants, the elderly and immunocompromised patients have impaired immune function and often respond poorly to norovirus candidate vaccines, leading to a vaccine (otherwise a good candidate) with poor efficacy. The duration of host immunity against norovirus also needs to be better characterized. The short-lived protective immunity has largely discouraged efforts in developing norovirus vaccines since vaccines with short-term protection are only suit for travelers but not for the populations at the highest risk (young children, the elderly, and immunocompromised individuals) who need vaccination the most. Early studies suggested that host immunity last for only months after norovirus natural infection or administration,Citation85Citation88 though another study indicated that host immunity could last for at most several years.Citation89 Perhaps, the lack of an in vitro virus culture system and the absence of small animal models for immunogenicity and efficacy studies are key obstacles that prevent us from understanding pathogenesis of norovirus infection and making progress in vaccine development particularly of live attenuated vaccines. The lack of defined immune correlates to protection against norovirus infection is another key barrier for norovirus vaccine preclinical and clinical evaluation. Total serum antibodies specific to norovirus may not be necessarily correlated to protection against the disease. Only serum antibodies blocking the binding of virus-like particles to HBGA (histo-blood group antigen) were found to be associated with protection against norovirus.Citation73Citation76,Citation90,Citation91 However, volunteers without or with low levels of HBGA due to an absence or a low expression of fucosyltransferase 2 gene (FUT2), despite they developed low serum antibodies after vaccination, were resistant to GI.1 or GII.4 norovirus infection,Citation91,Citation92 misguiding or further complicating vaccine candidacy assessment.

Progress has been also made in vaccine development for the other enteric viruses including astroviruses (Astroviridae), adenoviruses (Adenoviridae), and sapoviruses (Caliciviridae). Other Astroviridae members such as VA-Like and MLB-like astroviruses, Picornaviridae (silivirus, cosavirus), and Parvoviridae families (bocaviruses, bufaviruses) are also isolated from patients (usually in infants and children) with gastroenteritis. Several subunit vaccines have been investigated for prevention against astrovirus infections. In particular, a trivalent subunit vaccine for hepatitis E virus, norovirus, and astrovirus was generated by fusing together the dimeric P domains of the three viruses to form a tetramer.Citation93 When intranasally administered to mice, this trivalent product induced significant neutralizing antibody responses to the P domains of all three viruses. Another subunit astrovirus vaccine candidate used the capsid protein (CP) of mink astrovirus elicited high levels of serum anti-CP antib odies and lymphoproliferation responses and also stimulated IFN-γ levels in mice.Citation94 Importantly, it was observed that virus shedding was suppressed and clinical signs including severe diarrhea were reduced in the litters born to the immunized mink mothers when challenged with a heterogeneous astrovirus strain.Citation94 Future human volunteer studies and clinical trials are needed to assess the efficacy of these vaccine candidates against viral gastroenteritis.

II. Vaccines and vaccine development for bacterial enteric pathogens

Shigella

Gram-negative non-motile, rod-shaped Shigella bacteria are the second and the first leading causes of diarrhea, respectively, in children aged 12–23 and 24–59 months in developing countries,Citation2 though a community-based study suggested the attribution of Shigella bacteria to children’s diarrhea may be less significant.Citation6 Shigella is a genus of four species or groups (S. flexneri, S. sonnei, S. boydii, and S. dysenteriae) with more than 50 serotypes differentiated by lipopolysaccharide (LPS) O antigen specificity. After ingestion and passing through stomach and small intestines, Shigella bacteria invade host colon and rectum epithelial cells, multiply inside host cells, cause inflammation, induce cell death, spread to adjacent epithelial cells and destruct colon and rectum mucosal tissue,Citation95 resulting in shigellosisCitation96 or hemolytic uremic syndrome (HUS) with attribution from Shiga toxin (S. dysenteriae type 1).Citation97 Since only over ten (S. dysenteriae) to less than a couple of hundreds (S. sonnei & S. flexneri) of bacteria are needed to initiate shigellosis, person-to-person contact and even contact by flies can be important routes of transmission.Citation98Citation100

Identified virulence factors of Shigella include type III secretion system (T3SS) essential invasion plasmid antigens (Ipa A, B, C, D), intracellular spreading (Ics) proteins, LPS, and Shiga toxin. Ipa proteins kill macrophage and mediate bacterial adherence and invasion to colon and rectum epithelial cells, and Ics proteins (IcsA also called VirG, IcsB) assist Shigella bacteria for spreading to adjacent cells. Shigella LPS attributes to colon and rectum tissue damage and inflammatory responses, whereas Shiga toxin inactivates protein syntheses in eukaryotic cells. Clinical outcomes include fever, headache, vomiting, dehydration, and watery diarrhea but can progress to bloody and mucus-laden diarrhea, severe abdominal cramping, life-threaten dysentery, HUS, and death. Treatment of shigellosis is limited to supportive care to maintain hydration and to balance electrolyte levels. Antibiotic drugs are still prescribed to treat shigellosis for travelers and dysentery for children and immunocompromised individuals. However, antibiotics become less effective because Shigella spp. increasingly acquire antimicrobial resistance (AMR)Citation101Citation106or even worsen disease progression as some antibiotics promote Shiga toxin production.Citation107,Citation108

There is no vaccine licensed for Shigella. The feasibility of vaccine protection against shigellosis, however, is suggested by field observationCitation109,Citation110 and has been demonstrated in non-human primateCitation111 as well as controlled human infection studies,Citation112 evidenced by that pre-exposure to Shigella infection resulted in immunity and/or protection against subsequent infection or challenge. Moreover, serum antibodies to lipopolysaccharide (LPS) were revealed the correlates to protection.Citation113,Citation114 Unfortunately, LPS O antigens are serotype specific. Heterogeneity of Shigella serotypes is indeed the main challenge in vaccine development. Fortunately, only couples of species and a handful of serotypes are highly prevalent in shigellosis. Of four Shigella spp., S. flexneri (mostly in low-income and lower middle-income countries; responsible for about two-thirds of all diarrheal episodes) and S. sonnei (middle-income and developed countries; responsible for about one-fourth of all episodes) attribute to 90% of shigellosis cases,Citation115 with serotypes S. sonnei, S. flexneri 2a, 3a, and 6 for the vast majority of illnesses.Citation116,Citation117 Current LPS O antigen-based Shigella vaccine candidates are primarily targeting on these serotypes. In contrast to the serotype-specific LPS O antigens, virulence factors Ipa proteins and VirG/IcsA are found conservative among Shigella species and serotypes.Citation116,Citation118 Humans develop antibody responses specific to Ipa and VirG proteins after natural Shigella infection,Citation119Citation122 and antibodies derived from Ipa proteins were found to protect mice against pulmonary lethal challenge with different Shigella species or serotypesCitation123 or monkeys against severe diarrhea,Citation124 indicating Ipa and VirG alternative antigens for cross-protective Shigella vaccines.

Several Shigella vaccine candidates are under investigation. As briefly reviewed by Mani et al. recently,Citation117 these include live attenuated or killed whole-cell candidates, glycoconjugate or bioconjugate candidates, as well as subunit vaccine candidates. While many are at the pre-clinical stage, a few candidates have proceeded to phase I, II, or even phase III studies.

WRSS (WRSS1, WRSs2, WRSs3)

WRSS1, a live attenuated S. sonnei strain (Mosley) with a part of virG gene deleted, was developed by Walter Reed Army Institute of Research (WRAIR). WRSS1 was demonstrated to protect against homologous challenge in guinea pig keratoconjunctivitis model.Citation125 WRSS1 was well tolerated and immunogenic in healthy adults from developed countries,Citation126,Citation127 as well as adults and children in Bangladesh.Citation128 Unfortunately, WRSS1 at the regimen of a single dose of 1 × 10Citation4 inocula showed no evident efficacy against homologous challenge in an endemic setting.Citation129 Subsequently, WRSs2 and WRSs3 were developed, by deleting enterotoxins (ShET1 and ShET2) or further reducing LPS endotoxicity of WSSR1.Citation130 WRSs2 and WRSs3 were well tolerated and induced immune responses in non-human primatesCitation131 and healthy adults,Citation132 but they still showed reactogenicity though at a reduced rate.Citation132 The efficacy of WRSs2 or WRSs3 against homologous (or heterologous) challenge is yet to be demonstrated.

CVD 1208S

CVD 1208S is derived from reconstruction of CVD 1208, a live attenuated S. flexneri 2a mutant with deletion of guaBA locus regulating synthesis of inosine 5ʹ-monophosphate dehydrogenase (encoded by guaB) and guanosine 5ʹ-monophosphate synthetase (encoded by guaA),Citation133 enterotoxin gene set (ShET1) and invasiveness plasmid gene sen (ShET2),Citation134 in animal-free medium.Citation133 CVD 1208 and 1208S were generally well tolerated in healthy adults, and induced robust anti-LPS IgA and IgG antibody-secreting cells (ASC) responses, moderate serum IgG or fecal IgA, but mild serum IgA responses. Currently, there is no published efficacy data available from either candidate strain.

Other live attenuated Shigella vaccine candidates currently under pre-clinical studies include typhoid vaccine Ty21a expressing Shigella LPS O-antigens (also ETEC adhesin and toxoid antigens)Citation135Citation137 and ShigETEC. ShigETEC applied a modified noninvasive Shigella strain Vadizen (Istrati-32) to express ETEC toxoid fusion antigen LTB-STaN12S.Citation138

Inactivated Shigella whole-cell vaccine candidates

Inactivated whole-cell candidates were also investigated for protection against shigellosis. SsWC, formalin-inactivated S. sonnei, which showed to be immunogenic and protective against keratoconjunctivitis in guinea pigs when challenged with S. sonnei, was well tolerated in healthy adults.Citation139 Realized that SsWC is unlikely to provide cross-protection against other Shigella spp. or different serotypes, researchers added two more formalin-inactivated Shigella serotypes, S. flexneri 2a and 3a, to SsWC to produce a trivalent Shigella whole-cell vaccine candidate. This trivalent product induced anti-Ipa (B, C, D) and serotype-specific anti-LPS antibody responses and protected guinea pigs from three homologous serotypes in the keratoconjunctivitis model.Citation140 Additionally, two other inactivated whole-cell products were investigated. Inactivated ∆wzy Shigella flexneri 2a mutant which had O-antigen polysaccharide polymerase gene disrupted induced immunity cross-protective against serotypes 2a, 3a, and 6 in a mouse pulmonary model.Citation141 Heat killed multi-serotype Shigella (HKMS), which combined six Shigella serotypes (S. dysenteriae type 1, S. flexneri 2a, 3a, 6, S. sonnei, S. boydii), protected guinea pigs in a recto-colitis model,Citation142 neonatal mice passively,Citation143 and rabbitsCitation144 against challenge of all six homologous strains.

Polysaccharide-based subunit vaccine candidates

Shigella LPS O-specific polysaccharide moiety (O-antigen) chemically or biologically conjugated to a carrier protein was attempted to improve O-antigen safety and immunogenicity in developing Shigella polysaccharide conjugate vaccines. Indeed, some conjugate vaccine candidates have been investigated more extensively or are advanced the most in clinical studies. S. sonnei-rEPA, LPS O-antigen specific to S. sonnei chemically conjugated to recombinant exoprotein A of Pseudomonas aeruginosa (rEPA), was found safe and immunogenicCitation145,Citation146 and protective against S. sonnei shigellosis in healthy adultsCitation147 as well as children above three years of age, but little protection was observed from the children aged less than three years.Citation148 Other carrier proteins including CRM9 of Corynebacterium diphtheriae and a succinylated nontoxic peptide of Clostridium difficile toxin A were examined for enhancement at Shigella O-antigen immunogenicity.Citation149,Citation150

Different from chemical conjugation, bioconjugation uses E. coli N-linked protein glycosylation machinery to conjugate in vivo Shigella LPS O-antigens to a carrier protein.Citation151,Citation152 To bioconjugate Shigella O-antigens to carrier protein exoprotein A of P. aeruginosa (EPA), a modified EPA gene (harbor N-glycosylation consensus sequences) and a Shigella O-antigen gene were expressed in E. coli. The O-antigen repeating units preassembled in cytoplasm flip to periplasm and polymerize into O-antigen chain. These O-antigen chains then link to EPA protein to form bioconjugates. Bioconjugates are finally released to outer membrane after cell lysis.Citation153 Bioconjugate vaccine candidates for S. dysenteriae (SD133) and S. flexneri 2a (Flexyn2a) were demonstrated safe and immunogenic in phase I trials.Citation154,Citation155

Well-defined synthetic oligosaccharides were investigated as Shigella O-antigen surrogates to improve conjugate vaccine candidacy. It was reported that synthetic saccharides of S. dysenteriae induced anti-LPS antibodies after being conjugated to human serum albuminCitation156 and a synthetic pentadecasaccharide mimicking S. flexneri 2a O-antigen induced protective antibodies when was conjugated to tetanus toxoids.Citation157 However, the feasibility of using synthetic O-antigen surrogates in developing Shigella vaccines is yet to be fully established.Citation153 Additionally, S. sonnei 1790GAHB GMMA (generalized modules for membrane antigens) vaccine, blebs or native outer membrane vesicles shed by a genetically modified S. sonnei strain for a high level of production of LPS blebs,Citation158 was generally safe and induced robust antibody response to S. sonnei LPS in adults from an Africa endemic country.Citation159

Since Shigella O-antigen moieties of LPS are diverse due to variations of repeating units (two to six monosaccharides) thus are serotype specific, anti-LPS antibodies derived from an O-antigen of one Shigella serotype protect against homologous infections but cannot provide cross-protection against heterogeneous serotypes. In addition, conjugation of O-antigens with a carrier protein requires preparations and characterizations of the antigen and the carrier protein separately. Difficulties in conjugate purification and variations in bioconjugation process often lead to a lower conjugation efficiency, resulting in a lower production yield of bioconjugates. Moreover, the low efficiency of bioconjugation leaves a high proportion of carrier protein unglycosylated. This potentially mistakes host immune systems to respond to the carrier protein rather than the O-antigen, further hampering efforts in developing low-cost Shigella bioconjugate vaccines to be licensed in developing countries.

Protein-based subunit vaccine candidates

Protein-based and protein-LPS-based Shigella vaccine candidates under preclinical studies are mainly based on invasion plasmid antigens (Ipa proteins) or a complex of invasins composed of Ipa proteins and LPS. Like LPS, Ipa proteins (IpaB, C, D) induced protective antibodies against Shigella infections.Citation118,Citation160,Citation161 However, differed from the serotype-specific LPS, Ipa proteins are highly conserved among Shigella species and serotypes and even in other gram-negative bacteria,Citation162 thus become leading antigens for the development of cross-protective Shigella vaccines. DBF, a genetic fusion of proteins IpaD and IpaB was demonstrated to induce antibodies not only broadly protective against lethal challenge of S. flexneri, S. sonnei, and S. dysenteriae in mice (pulmonary infection model)Citation123 but also against severe diarrhea in non-human primates (JD Clements, personal communication). Invaplex, a complex of invasins consisting IpaB, C, D proteins and LPS water-phase extracted from Shigella flexneri culture growth, was found to induce protective antibodies to the invasin complex and LPS in mice and guinea pigs.Citation118 After initial examination of safety and immunogenicity, this protein-LPS-based subunit vaccine candidate was further modified to improve immunogenicity and more importantly efficacy.Citation161,Citation163-Citation165 However, the composition of the complex of invasins was never well defined and reproducibility remained as a challenge. A recent study revealed that InvaplexAR carrying recombinant IpaB and IpaC proteins and LPS purified from S. flexneri 2a and S. sonnei had immunogenicity improved and protected mice better against body weight loss.Citation166 Data from future safety and immunogenicity studies from human subjects and eventual field studies will provide better assessment of InvaplexAR or DBF subunit vaccine candidacy for Shigella.

Enterotoxigenic Escherichia coli (ETEC)

ETEC producing heat-stable toxin (STa) and/or heat-labile toxin (LT) is among the top five causes of children’s diarrhea from hospital-setting and community-setting surveillence.Citation2,Citation6 ETEC is also the leading cause of diarrhea in international travelers including deployed civilian and military personnel (travelers’ diarrhea).Citation167Citation169 Bacterial adhesins (fimbrial and non-fimbrial) and enterotoxins are the two types of key virulence factors associated with ETEC infection.Citation170,Citation171 ETEC bacteria produce over 25 adhesins to attach to different host receptors and two very distinctive enterotoxins (LT, STa) to disrupt homeostasis in host small intestinal epithelial cells to cause watery diarrhea. Ingested in contaminated food or water, ETEC bacteria pass host stomach, reach to small intestines, attach to host receptors, and colonize small intestines. Colonization brings ETEC in a proximity to effectively deliver enterotoxins into small intestinal epithelial cells. Enterotoxins elevate epithelial cells intracellular cyclic adenosine monophosphate (cAMP; by LT) and cyclic guanosine monophosphate (cGMP; by STa), leading to fluid hypersecretion and watery diarrhea or severe diarrhea like cholera. Without medical intervention, disease progresses to severe dehydration and causes acute death in young children.

There are no vaccines licensed for ETEC. Challenges in developing effective ETEC vaccines historically include immunological heterogeneity of ETEC pathotypes, identifying proper antigens to target enterotoxins especially the potent and poorly immunogenic STa toxin (until recently), and a lack of suitable animal models to assess vaccine efficacy.Citation171Citation175 An ideal ETEC vaccine would prevent all virulent ETEC pathotypes from colonizing host small intestines and neutralize enterotoxicity of both ETEC toxins. However, under current technologies, developing such a vaccine for ETEC seems unfeasible. A more practical strategy is to develop a vaccine cross-protective against the most prevalent and virulent ETEC pathotypes which cause moderate-to-severe infection.Citation173 ETEC pathotypes producing seven colonization factor antigen (CFA) or coli surface (CS) antigen adhesins [CFA/I, CFA/II (CS1-CS3), or CFA/IV (CS4-CS6)] and enterotoxin STa and/or LT are the most important pathotypes for children’s diarrhea and travelers’ diarrhea.Citation176Citation178 These pathotypes become the primary targets in current ETEC whole-cell (killed or live attenuated) and subunit vaccine development.

ETVAX

A killed whole-cell cocktail vaccine candidate is derived from rCTB-CF, an early killed whole-cell product.Citation179 Consisted of three ETEC strains expressing four adhesins (CFA/I, CS1, CS2, CS3) and an addition of recombinant B subunit of cholera toxin (rCTB) initially, rCTB-CF was afterward modified to carry five killed strains expressing six adhesins (CFA/I, CS1-CS5) (plus recombinant rCTB protein). Cholera toxin B subunit (CTB) was the immunogen in cholera vaccine Dukoral which induced cross-protective immunity against LT-producing ETEC,Citation179 due to the homology between LT B subunit (LTB) and CTB (85% homology of amino acids).Citation180 Orally administered rCTB-CF was tolerated and induced antigen-specific antibody responses in healthy adultsCitation179,Citation181-Citation183 and children aged above 18 months to two years in ETEC endemic countries.Citation184,Citation185 When given to Egyptian children less than two years of age, rCTB-CF-induced responses in only 30% of the children.Citation184 Subsequent efficacy studies, however, showed that rCTB-CF provided little protection to Egyptian infants against ETEC diarrheaCitation186 or only protection against the severity of diarrhea but not the overall diarrhea rate in the US healthy adults traveling to Mexico and Guatemala.Citation187 To further improve anti-CF and anti-LT immunogenicity and to be suitable to vaccinate young children, ETVAX was developed as the next generation vaccine to replace rCTB-CF. ETVAX consists of four inactivated recombinant E. coli strains that hyper-express CFA/I, CS3, CS5, and CS6 adhesins and recombinant subunit protein LCTBA (CTB subunit with seven amino acids replaced by the counterpart residues of LTB subunit).Citation180,Citation188 ETVAX was shown safe and broad immunogenic in healthy adults from developed countries and ETEC endemic countries.Citation189,Citation190 Interestingly, ETVAX-induced antibodies were found to cross-react to CS1, CS14, CS17, and CS7 adhesins.Citation191 A recently completed phase I study showed that ETVAX induced broad antigen-specific responses in Bangladesh children older than 12 months and moderate or mild responses in children less than 12 months of age.Citation192 However, protective efficacy of ETVAX against ETEC diarrhea in the endemic setting for children especially those aged less than 12 months is yet to be evaluated. Because it does not induce protective immunity against the key STa toxin which plays a more important role in children’s diarrhea, ETVAX likely will encounter challenge in providing broad protection against ETEC diarrhea particularly diarrhea caused by STa+ ETEC strains.Citation193

ACE527

A live attenuated vaccine candidate consists of three live strains, ACAM 2025 (CFA/I, LTB), ACAM 2022 (CS5/CS6, LTB) and ACAM 2027 (CS1/CS2/CS3, LTB).Citation194,Citation195 When given two doses of about 1011 CFUs (~3x1010 CFUs per strain), ACE527 induced vigorous immune responses but unfortunately also side effects, mainly vomiting in some adult subjects.Citation196 In a following double-blind placebo-controlled Phase IIb study, this live vaccine candidate (at a dose of 2 × 1011 CFUs) reduced the severity of diarrhea and bacteria shedding in volunteers when challenged with ETEC strain H10407 producing STa and LT.Citation197 Because of the side effects and unsatisfied efficacy, ACE527 was withheld temporarily from further evaluation. A later study using a three-dose regimen at a lower dosage (1010 CFUs, ~3 × 109 CFUs per strain) and the addition of adjuvant dmLT, however, showed 65.9% efficacy against severe diarrhea and 58.5% against diarrhea of any severity in the healthy adults challenged with H10407.Citation198 Interestingly, the same study found that adults vaccinated with ACE527 without adjuvant dmLT were not protected (23.1% efficacy against severe diarrhea). This brings up a question regarding the protective efficacy from the antibodies derived from adjuvant dmLT (which is also an antigen for an ETEC vaccine) against H10407 challenge and the definitive role dmLT played in this vaccine candidate (as an adjuvant, an antigen, or both). It seemed to us that dmLT played a more important role as an adjuvant for ACE527 because the additional anti-LT antibodies from dmLT adjuvant play little role against STa toxin from challenge strain H10407. Surely, ACE527 live attenuated ETEC vaccine candidate needs to be further evaluated for efficacy in adults and more importantly young children from ETEC endemic countries. However, ACE527, like the killed whole-cell candidate ETVAX, does not carry STa antigens to induce protective anti-STa immunity thus becomes limited in protecting against STa-producing ETEC infection.

ETEC subunit vaccine candidates

There are a few protein-based subunit vaccine candidates under development. CFA/I minor subunit tip adhesin CfaE protein was used for the development of a tip adhesin vaccine against ETEC producing CFA/I adhesin or adhesins antigenically homologous to CFA/I. Non-human primates intranasally immunized with CfaE tip adhesin protein developed antibodies that inhibit CFA/I agglutination to bovine erythrocytesCitation199 and further protect against ETEC H10407 challenge.Citation200 Additionally, adults passively acquired hyperimmune bovine IgG raised against CfaE tip adhesin protein were significantly protected against H10407 challenge (63% efficacy).Citation201 These data suggested that tip adhesin CfaE-induced immunity protects the homologous challenge strain (H10407) and possibly ETEC strains expressing adhesins homologous to CFA/I, Class 5 ETEC fimbrial adhesins.Citation202 Unfortunately, CfaE tip adhesin vaccine is unlikely to protect against ETEC strains that express immunologically different adhesins.

MecVax, a multivalent ETEC vaccine candidate is composed of LT-STa toxoid fusion 3xSTaN12S-mnLTR192G/L211A protein and CFA/I/II/IV multiepitope fusion antigen (MEFA) protein.Citation203 Toxoid fusion protein 3xSTaN12S-mnLTR192G/L211A, which carries three copies of STa toxoid STaN12S (the 12th residue asparagine was replaced with serine) and a monomeric peptide composed of one mutant LT A subunit and one LT B subunit,Citation204 induced antibodies that neutralize STa and LT enterotoxicityCitation204-Citation206 and protect against STa+ and LT+ ETEC diarrhea in a pig challenge model.Citation207,Citation208 CFA/I/II/IV MEFA, an epitope- and structure-based recombinant protein carries neutralizing epitope of the seven most important ETEC adhesins (CFA/I, CS1-CS6), induced antibodies that inhibit adherence of ETEC expressing any of these seven adhesins.Citation209Citation211 Because they are thermostabile and importantly antigenic compatible, 3xSTaN12S-mnLTR192G/L211A and CFA/I/II/IV MEFA can be combined and co-administered,Citation212 leading to the production of a multivalent ETEC subunit vaccine, MecVax. When parenterally (intramuscularly, intradermally, subcutaneously) administered, MecVax induced antibodies that neutralized STa and LT enterotoxicity and inhibited adherence of all seven adhesins (CFA/I, CS1-CS6). Most importantly, MecVax protected rabbits from ETEC colonization in small intestines and pigs from ETEC diarrhea.Citation203 MecVax will be prepared under good manufacturing practice (GMP) production and evaluated in future efficacy studies to determine its ETEC vaccine candidacy.

Other ETEC subunit vaccine candidates currently under preclinical studies include newly identified conservative antigens (EtpA, EatA, EaeH, YghJ)Citation213,Citation214 and STa toxoid conjugates.Citation215,Citation216

Diarrheagenic E. coli other than ETEC, including enteropathogenic E. coli (EPEC) and enterohemorrhagic E. coli (EHEC), are also causes of enteric infections. EPEC strains are listed among the top ten causes of diarrhea in children under the age of two years,Citation2,Citation6 but there are no vaccines developed for EPEC either. Ineffective diagnostics of EPEC disease outcomes and a lack of sufficient understanding at EPEC disease mechanisms and host immune responses are among the obstacles in EPEC vaccine development.Citation217 Unlike EPEC or ETEC, EHEC causes only sporadic outbreaks mainly due to consumption of undercooked beef products or raw vegetables. Though efforts are continuously made,Citation218 EHEC vaccine development for humans is discouraged because of a low incidence rate.

Cholera

An acute watery diarrhea caused by Vibrio cholerae remains a significant public health burden globally but more particularly in South Asia and sub-Sahara Africa.Citation219 Over 200 serogroups have been identified based on the variations of bacterial surface LPS. Historically, classical serogroup O1 caused pandemic cholera. Serogroup O139 emerged as a dominant cause of epidemic outbreaks in Asia in the early 1990s but became quiescent afterward, whereas O1 serogroup has reemerged as the primary agent of cholera. Serogroup O1 has two biotypes, classical and El Tor, and each biotype includes two serotypes (Inaba and Ogawa; rarely third serotype Hikojima). Transmitted through ingestion of contaminated water primarily and food as well, Vibrio cholera bacteria pass through stomach, colonize small intestine, deliver cholera toxin (CT; structurally and functionally homologous to heat-labile toxin of enterotoxigenic E. coli – ETEC) into intestinal epithelial cells to disrupt homeostasis in epithelial cells and to cause water hypersecretion and secretory diarrhea, often leading to dehydration and acute death if not treated.

Unlike ETEC or Shigella of which there are no vaccines licensed, cholera has three killed whole-cell oral vaccines (OCV) prequalified by WHO and licensed by many national regulatory authorities: Dukoral, Shanchol, and Euvichol ().

Dukoral

Also known as BS-WC, WC-BS, WC-rBS or CTB-WC, is a monovalent vaccine product initially prepared by Swedish National Bacteriological Laboratory (SBL). This whole-cell vaccine consists of 1 mg CT B subunit recombinant protein (rBS) and killed V. cholerae O1 (1011 CFUs, four strains, two classical biotypes and two El Tor biotypes, 2.5 × 1010 CFUs of each biotype). Dukoral is orally administered in sodium bicarbonate buffer (against stomach acid degradation of CTB). The initial field trial with a regimen of three doses at a six-week interval indicated that Dukoral provided 85% efficacy against cholera for six months in all children (2–5 years and above 5 years) and women above 15 years.Citation220 During the first year following Dukoral vaccination, hospital visits for treatment of diarrhea were reduced by 26% and severe dehydrating and fatal diarrhea were decreased by 49%.Citation221 However, at 36 months following immunization, the efficacy of Dukoral decreased to 26% for children aged 2 to 5 years.Citation222

Shanchol

From the field study of Dukoral, Clemens et al. observed that children aged 2–15 years and women above 15 years administered with the whole-cell vaccine without CTB protein were also protected short term (six months) against cholera, though at a lower efficacy (58%).Citation220 Without CTB recombinant protein, bicarbonate buffer (which was used mainly to prevent CTB protein from stomach acid degradation) was not needed for vaccine administration. This significantly eased the vaccine preparation and administration burden but also reduced vaccine manufacture complexity and cost. To produce an inexpensive oral whole-cell cholera vaccine against O1 and also O139 which emerged in the early 1990 s, VaBiotch (Vietnam) worked with International Vaccine Institute (IVI; South Korea) and developed a bivalent whole-cell cholera vaccine (ORC-Vax, mORCVAX), by adding the serogroup O139 component. ORC-Vax composed of four O1 serogroup strains (heat-killed classical Inaba and Ogawa, formalin-inactivated El Tor Inaba and classical Inaba) and one O139 serogroup strain, without CTB recombinant protein, was included in EPI in Vietnam.Citation223 Reformulated to remove toxin content,Citation224 ORC-Vax was transferred to Shantha Biotechnics in India for good manufacture production and WHO pre-qualification,Citation225 and was marketed as Shanchol afterward.Citation226 Shanchol was demonstrated safe and immunogenic in adults and children aged over 12 months.Citation226 At a regimen of two doses (14 days apart), Shanchol conferred 67%, 66%, and 65% cumulative protective efficacy, respectively, in two, three, and five years postvaccination among Kolkata residents aged one year and older.Citation227Citation229 The field study carried out in Dhaka, however, reported Shanchol of only 37% overall protective efficacy.Citation230 When a single dose regimen was used, however, this vaccine failed in providing adequate protection to young Dhaka children (16% efficacy).Citation231

Euvichol (Euvichol-Plus)

To increase vaccine production for stockpile, IVI also transferred the killed oral cholera vaccine technology to EuBiologics in Korean for fermentation manufacture of a bivalent cholera vaccine based on the same formulation of Shanchol. The product was marketed as Euvichol. Euvichol was demonstrated to be well tolerated and immunogenic.Citation232,Citation233 At a two-dose regimen, Euvichol induced vibriocidal responses at a level comparable to Shanchol.Citation234 Modified by removing thimerosal preservative and packing in plastic tubes instead of glass vials, Euvichol becomes more efficient for vaccine stockpile, transportation, and administer. This modified Euvichol was subsequently labeled as Euvichol-Plus. With an easy delivery and at a lower cost, Euvichol-Plus gains attraction, though efficacy data from field trials are yet to be demonstrated. Euvichol-Plus is in the process to be transferred to Shantan Biotechnics and will be marketed as Vibochol.

Live attenuated cholera vaccines

A few live attenuated cholera strains were developed. CVD 103-HgR (Vaxchora), which was derived from an O1 classical strain with the deletion of cholera toxin A subunit (CTA) and the inclusion of a mercury resistance gene, was shown initially not only tolerable and immunogenic in North American volunteersCitation235 but also protective against cholera challenge.Citation236,Citation237 However, CVD 103-HgR was found not protective against cholera in a subsequent large, randomized, double-blind, and placebo-controlled field study in Indonesia, possibly due to a low incidence of cholera in the country.Citation238 Peru-15 (CholeraGarde) is another well-studied live attenuated oral cholera vaccine. Peru-15 is derived from O1 El Tor Inaba strainCitation239 with a series of gene deletion.Citation240 Peru-15 at a single dose (5x10Citation8 – 1 × 109 CFUs) was found to be well tolerated and immunogenic in adults, children, and infants from Bangladesh,Citation241Citation243 and showed an efficacy of over 90% against diarrhea from a challenge study in North American healthy adults.Citation241 However, Peru-15 needs to be studied in the cholera epidemic setting to assess better its candidacy against cholera.

Campylobacter

Campylobacter infections were reported about 14.3 cases per 100,000 population in the US annually.Citation244 FoodNet confirmed that Campylobacter was the leading cause of foodborne illness in the US, accounting for the incidence rate of 19.5 per 100,000 population in 2018.Citation245 Campylobacter infection rates vary, ranging from 5% to 18% in laboratory-based surveillance in developing countries.Citation246 Major sources of Campylobacter infections are contaminated water and food including incorrectly prepared poultry meat and unpasteurized milk. Patients typically experience mild to severe diarrhea, bloody diarrhea, stomach pain, nausea, and/or vomiting lasting four to seven days. A dozen of Campylobacter species were isolated, but C. jejuni and C. coli are responsible for the most gastroenteritis cases.Citation247 Infections are initiated with Campylobacter bacteria penetrating host gastrointestinal mucus and invading intestinal epithelial cells. That leads to epithelial tissue damages and diarrhea.

The identified virulence factors of C. jejuni include cell adhesion and invasion proteins, capsular polysaccharide (CPS), cytolethal distending toxin (CDT A, B, C), lipooligosaccharide (LOS), and flagella.Citation248 Fibronectin-binding protein CadF,Citation249 aspartate/glutamate-binding protein PEB1Citation250 and surface-exposed lipoprotein JlpACitation251 were also indicated to be involved in adhesion of Campylobacter bacteria to host cells. C. jejuni CPS, a unique antigenic determinant in Penner serotyping scheme for Campylobacter, evades host immune response thus is potentially a key vaccine component. CDT is a multi-subunit protein and induces DNA damage in host epithelial cells to cause apoptotic cell death.Citation252

Preventing Campylobacter infections has been focused largely to decrease contamination at meat products during processing. Since poultry meat is the common source of infections, many preventive measures have been taken to inhibit Campylobacter colonization in the intestine tract of broiler chickens. Several vaccine candidates were tested in animal models, with a few advanced to phase 1 clinical trial.

Inactivated whole-cell vaccine candidates

Killed C. jejuni whole-cell vaccine (CWC) was developed and first evaluated in the early 1990s. CWC was prepared by formalin inactivation of C. jejuni strain 81–176 originally isolated from a child with acute diarrhea after consumption of contaminated raw milk.Citation253,Citation254 A pre-clinical study showed that mice orally immunized with CWC adjuvanted with E. coli heat-labile enterotoxin (LT) (CWC-LT) developed high levels of intestinal sIgA and IgG responses to Campylobacter and LT.Citation254 Moreover, inhibition of C. jejuni colonization was observed in the immunized mice when challenged with the homologous strain four weeks post CWC immunization.Citation254 Rhesus monkeys immunized with CWC-LT developed systemic IgA and IgG to Campylobacter antigens. These promising findings led to safety and immunogenicity phase 1 clinical study.Citation255 However, the followed controlled human infection model (CHIM) study suggested that this vaccine candidate was not protective against infection even from homologous strain C. jejuni 81–176.Citation256

Protein-based subunit vaccine candidates

C. jejuni surface antigens were used for subunit vaccine development. The rFlaA-MBP, a flagellin-based vaccine candidate was prepared by fusing the conserved regions of C. coli VC167 FlaA flagellin with E. coli maltose-binding protein (MBP).Citation257 Mice intranasally immunized with rFlaA-MBP developed antigen-specific serum IgG and intestinal secretory IgA antibodies. When challenged intranasally 26 days after immunization, mice were protected from Campylobacter colonization. However, a followed clinical trial failed to provide protection,Citation256,Citation257 resulting in rFlaA-MBP abandoned from further development.

Protein-polysaccharide conjugate vaccine candidates

Protein-polysaccharide conjugate vaccines were prepared by conjugating Campylobacter polysaccharides to a protein carrier. CPS81-176-CRM197 conjugate vaccine candidate (CJCV1) had the capsule polysaccharide (CPS) of C. jejuni 81–176 (HS23/HS36 serotype complex) crosslinked to well-characterized diphtheria toxin mutant CRM197.Citation258 When administered subcutaneously in mice, CJCV1 (CPS81-176-CRM197) induced robust serum IgG response to CPS81-176. This vaccine candidate was found to significantly decrease illness in mice and to protect the immunized New World monkey (Aotus nancymaae) from diarrhea after intranasal or orogastric challenge with homologous strains C. jejuni.Citation258 These results encouraged a phase 1 trial, in which 48 volunteers received two intramuscular vaccinations, 4 weeks apart, with or without adjuvant Alhydrogel.Citation259 Unfortunately, results showed that CJCV1 did not elicit robust serum IgA and IgG against the CPS moiety,Citation259 likely due to the lack of immunodominant nonstoichiometric O-methyl phosphoramidate (MeOPN) epitopes in CJCV1. Subsequently, new candidate CJCV2 was prepared from C. jejuni 81–176 strain by stably expressing phase-variable MeOPN transferases. CJCV2 is under manufacture for clinical evaluation.Citation259

Nontyphoidal Salmonella

Nontyphoidal salmonellosis (NTS) refers to the disease caused by any serotypes of Salmonella except for typhoidal Salmonella such as S. Typhi and Paratyphi A, B, and C. Two main serovars associated with enteric infections are S. Typhimurium and Enteritidis, causing an estimated 153 million cases of gastroenteritis and an annual death rate of 57,000 deaths globally.Citation260 In the United State, NTS is a leading cause of hospitalization and death due to foodborne illness, with 33% of the 1.2 million NTS cases are due to consumption of contaminated beef, pork, and poultry meat.Citation261 NTS infection results in self-limiting diarrhea in immunocompetent hosts; however, bacteremia occurs in approximately 5% of immunologically compromised individuals or patients with gastrointestinal illness.Citation262 Invasive NTS (iNTS) bacteremia often kills children who suffer from malnourishment or AIDS in the developing world, especially in sub-Saharan Africa, where multilocus sequence type ST313 has been dominant in association with bacteremia.Citation263,Citation264 iNTS isolates within ST313 and S. Typhi might have similar virulence properties and process of adaption to the human host.Citation265 iNTS isolates adhere to the mucosa, invade host small intestinal epithelial cells and then penetrate the mucosal barrier. Once iNTS bacteria are internalized through the M cells in the Peyer’s patches, they are translocated to the intestinal lymphoid follicles, spread to the liver, spleen, or bone-marrow, and disseminated throughout the body, leading to fatal illness by bacteremia.Citation265

Whole-cell vaccine candidates

While licensed vaccines against typhoid Salmonella have been licensed and used globally, there are no vaccines available for NTS. Several vaccine candidates against S. Typhimurium and Enteritidis are under development, but only one NTS vaccine, a live attenuated WT05, has been completed for a Phase 1 study.Citation266 Microscience Limited reported double mutant S. Typhimurium strain WT05 constructed by deleting aroC (encoding enzymes of the prechorismate biosynthetic pathway)Citation267 and ssaV (encoding a protein of the SPI-2 that involves in invasion of host cells)Citation266 genes were attenuated and immunogenic in humans. Adult volunteers receiving 10Citation7 to 109 CFU of WT05 showed no serious adverse effects and mounted S. Typhimurium LPS-specific antibody-secreting cell responses with variable serum IgG and IgA antibodies. However, prolonged stool shedding of WT05 was detected in healthy volunteers for up to 23 days post inoculation.Citation266 Other live attenuated NTS vaccine strains include CVD 1921 and CVD 1923.Citation268 CVD 1921 and CVD 1923 were derived from the invasive wild-type strains S. Typhimurium I77 by deleting genes guaBA (encoding guanine biosynthesis) and clpP (encoding a master protease regulator), or additionally fliD (encoding a capping protein). Oral LD50 analyses confirmed that CVD 1921 (∆guaBAclpP) or CVD 1923 (∆guaBAclpPfliD) were highly attenuated in mice; moreover, 80% to 86% of orally immunized mice were protected against lethal challenge from wild-type S. Typhimurium I77. Additionally, strain CVD 1941 with deletions of guaBA and clpP genes from wild-type S. Enteritidis R11 was also shown attenuated in mice and protected 76% of the immunized mice against S. Enteritidis infection.Citation268

Protein-polysaccharide conjugate vaccine candidates

Glycoconjugate vaccines are also under development for NTS.Citation269 O polysaccharides linked to core polysaccharides (COPS) of S. Enteritidis purified from the genetically attenuated strain CVD 1941 was conjugated to flagellin protein at various polysaccharide/flagellin ratios and examined for enhancement of polysaccharide immunogenicity.Citation269 Mice intramuscularly immunized with COPS-flagellin conjugates developed high anti-LPS and anti-flagellin IgG, and the derived antibodies mediated opsonophagocytosis, an effective antibacterial mechanism against both complement-resistant and complement-sensitive NTS strains. Moreover, a followed challenge study showed mice immunized with COPS-flagellin conjugates, at different ratios of polysaccharide/flagellin, were protected against lethal challenge from wild-type S. Enteritidis R11 at the efficacy ranged from 83% to 100%.Citation269

Protein-based subunit vaccine candidates

Genetic fusions of Salmonella pathogenicity island 1 (SPI-1) or 2 (SPI-2) tip and translocator proteins were applied to develop broadly protective subunit vaccines (S1 and S2) for NTS.270 S1 is the fusion of SipD and SipB, the tip and the translocator of the SPI-1 type three secretion system (T3SS), whereas S2 is the fusion of SseB and SseC, the tip and the translocator of the SPI-2 T3SS. When intramuscularly immunized and adjuvanted with Alhydrogel and monophosphoryl lipid A, S1 or S2 induced immunity to protect 60% of the immunized mice against S. enterica challenges. The immunized mice also had a reduction of cecal inflammation when challenged with homologous strain S. Typhimurium wild-type SL1344 or heterologous strain S. Enteritidis wild-type 125109. Both S1 and S2 candidates elicited high levels of IgG responses to SipB and SipD, or SseB and SseC, along with IgG-secreting cells in mouse spleens and bone marrow. However, no fecal IgA or antigen-specific IgA secreting cells in spleen or bone marrow were detected from the immunized mice.Citation270

Vaccinology for multivalent vaccine development

Heterogeneity among viral genotypes or genogroups and bacterial pathotypes or serogroups is among the key challenges in developing vaccines against gastroenteritis. An effective vaccine needs to induce broad immunity cross-protective against heterogeneous viral or bacterial strains. To overcome this heterogeneity challenge, different vaccinology strategies have been attempted. This includes whole-cell cocktail vaccines which mix together several strains or isolates (inactivated or live attenuated) to expand vaccine coverage, vaccines which carry antigens conserved among different strains for generic immunity, and vaccines combining neutralizing epitopes or antigen domains representing various heterogeneous groups to induce an array of immunity for broad protection. Conventional whole-cell vaccine approach remains much practical for the pathogens of which disease mechanism is not fully characterized and virulence determinants are yet to be determined or safely included. With a serial of passages of pathogenic viral strains in vitro or in vivo, or systematic deletions of virulence genes, attenuate strains or mutant strains can be produced. Attenuated strains can also be obtained from animal originated strains (Jennerian vaccinology) or avirulent strains isolated from asymptomatic children or adults. These strains mimic the virulent strains in inducing host immune responses but do not cause infections, becoming whole-cell vaccine candidates. More specifically, systematically comparative examination of virulence significance among deletion mutants helps to identify pathogen virulence factors, leading to vaccines to target on specific virulence determinants. Whole-genome sequencing without prior knowledge of virulence determinants has reversed the conventional vaccine development process. Reverse vaccinology has a great potential to substantially accelerate vaccine development. By analyzing genome-wide sequence differences of virulent strains and avirulent strains, we can rapidly identify the putative virulence genes associated with pathogenicity and initiate vaccine development.Citation271,Citation272 More recently, advance in structural biology and computational biology including protein modeling further allows us to in silico identify immuno-dominant B-cell and T-cell epitopes or peptide domains and to develop safe and precision vaccines ().

Combining a few live attenuated or inactivated strains certainly broadens immunogenicity spectrum but can introduce unwanted effects. Excessive somatic antigens carried by a cocktail whole-cell vaccine product increase the risk of side effects and potentially deviate host immune responses away from targeting to specific virulence antigens. That leads to adverse effects and likely lower host immune responses, thus vaccine safety concern and unsatisfied protective efficacy. Additionally, a vaccine carrying multiple viral or bacterial strains introduces complexity and difficulties to formulation and manufacture, risking vaccine manufacturability and product affordability particularly to the most needed populations in the low-income countries. Using antigens conserved inclusively among virulent strains, particularly in the format of recombinant proteins, on the other hand, largely eliminates the negative impacts associated with excessive somatic antigens but also less likely alters host mucosal microbiome. However, identifying conservative antigens that induce protective immunity specific to enteric pathogens can be difficult potentially. Additionally, protein-based vaccines often need adjuvants (if the target recombinant proteins possess no adjuvanticity) and booster immunizations to improve induction of host immune responses, may also encounter challenges in inducing robust local mucosal immunity against enteric infections as well as concerns of vaccine manufacturability and affordability if multiple conservative antigens are needed.

An epitope-based and structure-based vaccinology named multiepitope-fusion-antigen (MEFA) is described recently for developing a broadly protective vaccine for ETEC.Citation209,Citation273-Citation277 Assisted with structural biology and computational biology, MEFA vaccinology uses a platform to combine structural vaccinology and epitope vaccinology and applies a backbone protein to present neutralizing epitopes of multiple heterogeneous strains to produce a single multivalent immunogen (). Ideally, this backbone protein is a key virulence factor protein which is nontoxic and strongly immunogenic (better also exhibits adjuvanticity), possesses multiple well-separated epitopes and a stable secondary structure, and is easily expressed and purified. With substitutions of the backbone epitopes with foreign neutralizing epitopes, guided by protein modeling and molecular dynamic simulation, a MEFA protein mimics foreign epitope native antigenicity and becomes a multivalent antigen for a precision vaccine to provide cross-protection against heterogeneous pathotypes or pathogens. By applying this MEFA vaccinology platform, we have constructed multivalent proteins for broad protection against ETEC toxins,Citation204 adhesins,Citation209,Citation275 toxins and adhesins,Citation203,Citation276,Citation277 Shigella spp. and serotypes.Citation278 Preclinical studies demonstrated ETEC adhesin MEFA CFA/I/II/IV and toxoid fusion 3xSTaN12S-mnLTR192G/L211A, when administered subcutaneously or intraperitoneally in mice and intramuscularly in pigs, induced broadly protective antibodies against adherence of the seven most important ETEC adhesins (CFA/I, CS1 to CS6)Citation209,Citation275 or enterotoxicity of LT and STa.Citation204,Citation207,Citation208 Moreover, piglets born to the immunized sows acquired MEFA- and toxoid fusion-derived antibodies and were protected from diarrhea following STa+ or LT+ ETEC challenge.Citation207 Additionally, a MEFA-based vaccine can further broaden its protective coverage by including another antigenically compatible MEFA immunogen to potentially protect against two or more than two groups of pathogens.

There are several advantages of the MEFA vaccinology platform in developing cross-protective vaccines. MEFA vaccinology is different from epitope vaccinology approach which stacks multiple epitopes into a linear peptide. Without assessing the secondary structure and native antigenicity of individual epitopes, epitope vaccines are often shown poor immunogenicity and a low efficacy. MEFAs mimic stable backbone secondary structure and epitope native antigenicity, thus induce strong immune responses to multiple virulence factors or strains represented by individual neutralizing epitopes, protecting against heterogeneous pathotypes. Like other protein-based vaccines, a MEFA-based vaccine consists of a purified protein produced by genetically engineered bacteria; therefore, the vaccine component is well defined. Since MEFA vaccine carries no somatic antigens and has no antibiotic-resistant genes involved, vaccines will not mask host immune responses or introduce the risk of antimicrobial resistance (AMR) to the environment. MEFA proteins can be produced in large quantities at a low cost compared to other strategies that require fermentation of multiple organisms to cover the different species and serotypes. Low-cost vaccines become essential to be licensed in low-income countries. Additionally, because it is protein-based, product purity, immunogenicity, thermostability, and safety characteristics can be easily documented and measured. Perhaps more important practically, MEFA vaccines can potentially be combined and co-administered with other vaccines to fit a single EPI (Expanded Program on Immunization), which becomes very much appreciated since the current EPI is getting overly crowded with the continuous introduction of new vaccines. Because MEFA vaccines would most likely be administered parenterally, they will thus be immunogenic even in infants in low-income to middle-income countries where oral vaccines are often less immunogenic and less protective.Citation279 Furthermore, MEFA is a platform that can be adaptable for multivalent vaccine development for many pathogens including enteric Shigella, ETEC and other enteric pathogens, but potentially respiratory and other infectious pathogens as well.

Conclusion

Gastroenteritis remains a major threat to public health particularly in the low-income to middle-income countries. Parasitic, viral, and bacterial pathogens are the main causes of enteric infections. While WASH (clean drinking water, sanitation, and personal hygiene) may not be quickly implementable in resource-limited countries or regions, cost-effective vaccines and vaccination become a popular practice. However, though we have WHO prequalified vaccines for rotavirus, cholera, and Salmonella typhoid fever, we do not have vaccines licensed for parasites and many other enteric viral and bacterial pathogens including calicivirus, diarrheagenic E. coli, Shigella, Campylobacter, and no-typhoid Salmonella. Developing effective vaccines for enteric pathogens continues to be very challenging due to many reasons. A lack of understanding or characterization at immune correlates to protection from vaccines or vaccine candidates against enteric diseases hampers enteric vaccine development. While vaccine or vaccine-induced IgG antibody responses are more often measured, assessment of IgA particularly mucosal IgA responses should be emphasized. The greatest challenge in the current vaccine development is perhaps the heterogeneity among genotypes or serotypes of viral and bacterial pathogens. An effective vaccine would need to induce broad immunity to cross protect against heterogeneous genotypes or serotypes. Cocktail whole-cell vaccines which mix together a few heterogeneous genotypes or serotypes of an enteric viral or bacterial pathogen certainly broaden vaccine coverage. Whole-cell vaccines often encounter the challenge in inducing strong immunity in children aged less than two years to provide adequate protection to the most vulnerable group. LPS-based vaccines are typically serotype specific; thus, they often fall short in providing cross-protection against heterogeneous serotypes. While protein-based vaccines using conservative antigens for generic protection seem promising, antigens conserved inclusively in virulent strains or isolates may not be easily identified. The newly developed epitope- and structure-based MEFA (multiepitope fusion antigen) vaccinology platform may provide a means to develop broadly protective multivalent vaccines for heterogeneous pathogens or diseases.

Figure 1. A scheme to illustrate vaccinology evolution. From conventional vaccinology to reverse vaccinology and further to structural vaccinology, leading from whole-cell vaccines, to subunit vaccines and then structural epitope vaccines.

Figure 1. A scheme to illustrate vaccinology evolution. From conventional vaccinology to reverse vaccinology and further to structural vaccinology, leading from whole-cell vaccines, to subunit vaccines and then structural epitope vaccines.

Figure 2. MEFA vaccinology applies a backbone immunogen to present neutralizing epitopes of virulence determinants from multiple heterogeneous strains, by substituting backbone surface exposed but less immunogenic backbone epitopes with foreign epitopes, and to mimic foreign epitope native antigenicity assisted with protein modeling and molecule dynamics simulation.

Figure 2. MEFA vaccinology applies a backbone immunogen to present neutralizing epitopes of virulence determinants from multiple heterogeneous strains, by substituting backbone surface exposed but less immunogenic backbone epitopes with foreign epitopes, and to mimic foreign epitope native antigenicity assisted with protein modeling and molecule dynamics simulation.

Disclosure of interest

The authors report no conflict of interest.

Acknowledgments

This manuscript is supported by NIH R01AI121067-01A1.

Supplementary

Supplemental data for this article can be accessed on the publisher’s website.

Additional information

Funding

This work was supported by the National Institutes of Health [R01AI121067-01A1].

References

  • Black RE, Cousens S, Johnson HL, Lawn JE, Rudan I, Bassani DG, Jha P, Campbell H, Walker CF, Cibulskis R, et al. Global, regional, and national causes of child mortality in 2008: a systematic analysis. Lancet. 2010;375(9730):1969–1987. doi:10.1016/S0140-6736(10)60549-1.
  • Kotloff KL, Nataro JP, Blackwelder WC, Nasrin D, Farag TH, Panchalingam S, Wu Y, Sow SO, Sur D, Breiman RF, et al. Burden and aetiology of diarrhoeal disease in infants and young children in developing countries (the Global Enteric Multicenter Study, GEMS): a prospective, case-control study. Lancet. 2013;382(9888):209–222. doi:10.1016/S0140-6736(13)60844-2.
  • Kirk MD, Pires SM, Black RE, Caipo M, Crump JA, Devleesschauwer B, Dopfer D, Fazil A, Fischer-Walker CL, Hald T, et al. World Health Organization Estimates of the Global and Regional Disease Burden of 22 Foodborne Bacterial, Protozoal, and Viral Diseases, 2010: A Data Synthesis. PLoS Med. 2015;12(12):e1001921. doi:10.1371/journal.pmed.1001921.
  • Vos T, Allen C, Arora M, Barber RM, Bhutta ZA, Brown A, Carter A, Casey DC, Charlson FJ, Chen AZ. GBD 2015. Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990-2015: a systematic analysis for the global burden of disease study 2015. Lancet. 2016;388(10053):1545–1602. doi:10.1016/S0140-6736(16)31678-6.
  • Troeger C, Blacker BF, Khalil IA, Rao PC, Cao SJ, Zimsen SRM, Albertson S, Stanaway JD, Deshpande A, Brown A, et al. Estimates of the global, regional, and national morbidity, mortality, and aetiologies of diarrhoea in 195 countries: a systematic analysis for the global burden of disease study 2016. Lancet Infect Dis. 2018;18(11):1211–1228. doi:10.1016/S1473-3099(18)30362-1.
  • Platts-Mills JA, Babji S, Bodhidatta L, Gratz J, Haque R, Havt A, McCormick B, McGrath M, Olortegui M, Samie A, et al. Pathogen-specific burdens of community diarrhoea in developing countries: a multisite birth cohort study (MAL-ED). Lancet Glob Health. 2015;3(9):e564–575. doi:10.1016/S2214-109X(15)00151-5.
  • World Health Organization. Rotavirus vaccines. Wkly Epidemiol Rec. 2007;82(32):285–295.
  • Bernstein DI, Glass RI, Rodgers G, Davidson BL, Sack DA. Evaluation of rhesus rotavirus monovalent and tetravalent reassortant vaccines in US children. JAMA. 1995;273(15):1191–1196. doi:10.1001/jama.1995.03520390051032.
  • Joensuu J, Koskenniemi E, Pang XL, Vesikari T. Randomised placebo-controlled trial of rhesus-human reassortant rotavirus vaccine for prevention of severe rotavirus gastroenteritis. Lancet. 1997;350(9086):1205–1209. doi:10.1016/S0140-6736(97)05118-0.
  • PerezSchael I, Guntinas MJ, Perez M, Pagone V, Rojas AM, Gonzalez R, Cunto W, Hoshino Y, Kapikian AZ. Efficacy of the rhesus rotavirus-based quadrivalent vaccine in infants and young children in Venezuela. N Engl J Med. 1997;337(17):1181–1187. doi:10.1056/NEJM199710233371701.
  • Rennels M, Glass R, Dennehy P, Bernstein D, Pichichero M, Zito E, Mack M, Davidson B, Kapikian A. Safety and efficacy of high-dose rhesus-human reassortant rotavirus vaccines - report of the national multicenter trial. Pediatrics. 1996;97(1):7–13. United States Rotavirus Vaccine Efficacy Group
  • Santosham M, Moulton LH, Reid R, Croll J, Weatherbolt R, Ward R, Forro J, Zito E, Mack M, Brenneman G, et al. Efficacy and safety of high-dose rhesus-human reassortant rotavirus vaccine in Native American populations. J Pediatr. 1997;131(4):632–638. doi:10.1016/S0022-3476(97)70076-3.
  • Banyai K, Laszlo B, Duque J, Steele AD, Nelson EAS, Gentsch JR, Parashar UD. Systematic review of regional and temporal trends in global rotavirus strain diversity in the pre rotavirus vaccine era: insights for understanding the impact of rotavirus vaccination programs. Vaccine. 2012;30S:A122–A130. doi:10.1016/j.vaccine.2011.09.111.
  • Doro R, Laszlo B, Martella V, Leshem E, Gentsch J, Parashar U, Banyai K. Review of global rotavirus strain prevalence data from six years post vaccine licensure surveillance: is there evidence of strain selection from vaccine pressure? Infect Genet Evol. 2014;28:446–461. doi:10.1016/j.meegid.2014.08.017.
  • Parashar UD, Nelson EAS, Kang G. Diagnosis, management, and prevention of rotavirus gastroenteritis in children. BMJ. 2013;347:f7204.
  • Murphy TV, Gargiullo PM, Massoudi MS, Nelson DB, Jumaan AO, Okoro CA, Zanardi LR, Setia S, Fair E, LeBaron CW, et al. Intussusception among infants given an oral rotavirus vaccine. N Engl J Med. 2001;344(8):564–572. doi:10.1056/NEJM200102223440804.
  • Centers for Disease Control and Prevention. NMA recommends withdrawal of rotavirus vaccine. J Natl Med Assoc 1999;91:645.
  • Rothman KJ, Young-Xu Y, Arellano F. Age dependence of the relation between reassortant rotavirus vaccine (RotaShield) and intussusception. J Infect Dis. 2006;193(6):898. doi:10.1086/500217.
  • Armah GE, Kapikian AZ, Vesikari T, Cunliffe N, Jacobson RM, Burlington DB, Ruiz LP. Efficacy, immunogenicity, and safety of two doses of a tetravalent rotavirus vaccine rrv-tv in ghana with the first dose administered during the neonatal period. J Infect Dis. 2013;208(3):423–431. doi:10.1093/infdis/jit174.
  • Ciarlet M, Schodel F. Development of a rotavirus vaccine: clinical safety, immunogenicity, and efficacy of the pentavalent rotavirus vaccine, rotateq®. Vaccine. 2009;27S:G72–G81. doi:10.1016/j.vaccine.2009.09.107.
  • Block SL, Vesikari T, Goveia MG, Rivers SB, Adeyi BA, Dallas MJ, Bauder J, Boslego JW, Heaton PM. Efficacy, immunogenicity, and safety of a pentavalent human-bovine (WC3) reassortant rotavirus vaccine at the end of shelf life. Pediatrics. 2007;119(1):11–18. doi:10.1542/peds.2006-2058.
  • Vesikari T, Clark HF, Offit P, Schodel F, Dallas M, Heaton P, Krah D, Shaw A, Garbarg-Chenon A, Golm G, et al. The effect of dose and composition of a pentavalent rotavirus reassortant vaccine (rotaTeqTM) upon safety, efficacy, and immunogenicity in healthy infants. Presented at: The 37th National Immunization Conference of CDC. 2003 Mar 19; Ontario, Canada.
  • Clark HF, Bernstein DI, Dennehy PH, Offit P, Pichichero M, Treanor J, Ward RL, Krah DL, Shaw A, Dallas MJ, et al. Safety, efficacy, and immunogenicity of a live, quadrivalent human-bovine reassortant rotavirus vaccine in healthy infants. J Pediatr. 2004;144(2):184–190. doi:10.1016/j.jpeds.2003.10.054.
  • Soares-Weiser K, MacLehose H, Bergman H, Ben-Aharon I, Nagpal S, Goldberg E, Pitan F, Cunliffe N. Vaccines for preventing rotavirus diarrhoea: vaccines in use. Cochrane Database Syst Rev. 2012;2:CD008521.
  • Tate JE, Patel MM, Cortese MM, Lopman BA, Gentsch JR, Fleming J, Steele AD, Parashar UD. Remaining issues and challenges for rotavirus vaccine in preventing global childhood diarrheal morbidity and mortality. Expert Rev Vaccines. 2012;11(2):211–220. doi:10.1586/erv.11.184.
  • Patel M, Pedreira C, De Oliveira LH, Umana J, Tate J, Lopman B, Sanchez E, Reyes M, Mercado J, Gonzalez A, et al. Duration of protection of pentavalent rotavirus vaccination in Nicaragua. Pediatrics. 2012;130(2):E365–E372. doi:10.1542/peds.2011-3478.
  • Bernstein DI, Sack DA, Rothstein E, Reisinger K, Smith VE, O’Sullivan D, Spriggs DR, Ward RL. Efficacy of live, attenuated, human rotavirus vaccine 89-12 in infants: a randomised placebo-controlled trial. Lancet. 1999;354(9175):287–290. doi:10.1016/S0140-6736(98)12106-2.
  • Vesikari T, Karvonen A, Puustinen L, Zeng SQ, Szakal ED, Delem A, De Vos B. Efficacy of RIX4414 live attenuated human rotavirus vaccine in Finnish infants. Pediatr Infect Dis J. 2004;23(10):937–943. doi:10.1097/01.inf.0000141722.10130.50.
  • Araujo EC, ClemenS SAC, Oliveira CS, Justino MCA, Rubio P, Gabbay YB, da Silva VB, Mascarenhas JDP, Noronha VL, Clemens R, et al. Safety, immunogenicity, and protective efficacy of two doses of RIX4414 live attenuated human rotavirus vaccine in healthy Brazilian infants. J Pediatr (Rio J). 2007;83(3):217–224. doi:10.2223/JPED.1600.
  • Perez-Schael I, Salinas B, Tomat M, Linhares AC, Guerrero ML, Ruiz-Palacios GM, Bouckenooghe A, Yarzabal JP. Efficacy of the human rotavirus vaccine RIX4414 in malnourished children. J Infect Dis. 2007;196(4):537–540. doi:10.1086/519687.
  • Vesikari T, Karvonen A, Prymula R, Schuster V, Tejedor JC, Cohen R, Meurice F, Han HH, Damaso S, Bouckenooghe A. Efficacy of human rotavirus vaccine against rotavirus gastroenteritis during the first 2 years of life in European infants: randomised, double-blind controlled study. Lancet. 2007;370(9601):1757–1763. doi:10.1016/S0140-6736(07)61744-9.
  • Linhares AC, Velazquez FR, Perez-Schael I, Saez-Llorens X, Abate H, Espinoza F, Lopez P, Macias-Parra M, Ortega-Barria E, Rivera-Medina DM, et al. Efficacy and safety of an oral live attenuated human rotavirus vaccine against rotavirus gastroenteritis during the first 2 years of life in Latin American infants: a randomised, double-blind, placebo-controlled phase III study. Lancet. 2008;371(9619):1181–1189. doi:10.1016/S0140-6736(08)60524-3.
  • O’Ryan M, Linhares AC. Update on rotarix: an oral human rotavirus vaccine. Expert Rev Vaccines. 2009;8(12):1627–1641. doi:10.1586/erv.09.136.
  • Kawamura N, Tokoeda Y, Oshima M, Okahata H, Tsutsumi H, Van Doorn LJ, Muto H, Smolenov I, Suryakiran PV, Han HH. Efficacy, safety and immunogenicity of RIX4414 in Japanese infants during the first two years of life. Vaccine. 2011;29(37):6335–6341. doi:10.1016/j.vaccine.2011.05.017.
  • Cunliffe NA, Witte D, Ngwira BM, Todd S, Bostock NJ, Turner AM, Chimpeni P, Victor JC, Steele AD, Bouckenooghe A, et al. Efficacy of human rotavirus vaccine against severe gastroenteritis in Malawian children in the first two years of life: A randomized, double-blind, placebo controlled trial. Vaccine. 2012;30:A36–A43. doi:10.1016/j.vaccine.2011.09.120.
  • Madhi SA, Kirsten M, Louw C, Bos P, Aspinall S, Bouckenooghe A, Neuzil KM, Steele AD. Efficacy and immunogenicity of two or three dose rotavirus-vaccine regimen in South African children over two consecutive rotavirus-seasons: A randomized, double-blind, placebo-controlled trial. Vaccine. 2012;30:A44–A51. doi:10.1016/j.vaccine.2011.08.080.
  • Phua KB, Lim FS, Lau YL, Nelson EAS, Huang LM, Quak SH, Lee BW, van Doorn LJ, Teoh YL, Tang H, et al. Rotavirus vaccine RIX4414 efficacy sustained during the third year of life: A randomized clinical trial in an Asian population. Vaccine. 2012;30(30):4552–4557. doi:10.1016/j.vaccine.2012.03.030.
  • Lau YL, Nelson EAS, Poon KH, Chan PKS, Chiu S, Sung R, Leung CW, Ng D, Ma YM, Chan D, et al. Efficacy, safety and immunogenicity of a human rotavirus vaccine (RIX4414) in Hong Kong children up to three years of age: A randomized, controlled trial. Vaccine. 2013;31(18):2253–2259. doi:10.1016/j.vaccine.2013.03.001.
  • Li RC, Huang T, Li YP, Luo D, Tao JH, Fu BT, Si GA, Nong Y, Mo ZH, Liao XY, et al. Human rotavirus vaccine (RIX4414) efficacy in the first two years of life A randomized, placebo-controlled trial in China. Hum Vacc Immunother. 2014;10(1):11–18. doi:10.4161/hv.26319.
  • Phua KB, Lim FS, Quak SH, Lee BW, Teoh YL, Suryakiran PV, Han HH, Bock HL. Efficacy, immunogenicity and safety of a human rotavirus vaccine rix4414 in Singaporean Infants. Ann Acad Med Singapore. 2016;45:44–50.
  • Willame C, Noordegraaf-Schouten MV, Gvozdenovic E, Kochems K, Oordt-Speets A, Praet N, van Hoorn R, Rosillon D. effectiveness of the oral human attenuated rotavirus vaccine: a systematic review and meta-analysis-2006-2016. Open Forum Infect Di. 2018;5:ofy292.
  • Luna-Casas G, Juliao P, Carreno-Manjarrez R, Castaneda-Prado A, Cervantes-Apolinar MY, Navarro-Rodriguez R, Sanchez-Gonzalez G, Cortes-Alcala R, DeAntonio R. Vaccine coverage and compliance in Mexico with the two-dose and three-dose rotavirus vaccines. Hum Vaccin Immunother. 2019;15(6):1251–1259. doi:10.1080/21645515.2018.1540827.
  • Clark A, van Zandvoort K, Flasche S, Sanderson C, Bines J, Tate J, Parashar U, Jit M. Efficacy of live oral rotavirus vaccines by duration of follow-up: a meta-regression of randomised controlled trials. Lancet Infect Dis. 2019;19(7):717–727. doi:10.1016/S1473-3099(19)30126-4.
  • Bhandari N, Sharma P, Glass RI, Ray P, Greenberg H, Taneja S, Saksena M, Rao CD, Gentsch JR, Parashar U, et al. Safety and immunogenicity of two live attenuated human rotavirus vaccine candidates, 116E and I321, in infants: results of a randomised controlled trial. Vaccine. 2006;24(31–32):5817–5823. doi:10.1016/j.vaccine.2006.05.001.
  • Bhandari N, Sharma P, Taneja S, Kumar T, Rongsen-Chandola T, Appaiahgari MB, Mishra A, Singh S, Vrati S, Grp RVD. A Dose-Escalation Safety and Immunogenicity Study of Live Attenuated Oral Rotavirus Vaccine 116E in Infants: A Randomized, Double-Blind, Placebo-Controlled Trial. J Infect Dis. 2009;200(3):421–429. doi:10.1086/600104.
  • Bhandari N, Rongsen-Chandola T, Bavdekar A, John J, Antony K, Taneja S, Goyal N, Kawade A, Kang G, Rathore SS, et al. Efficacy of a monovalent human-bovine (116E) rotavirus vaccine in Indian infants: a randomised, double-blind, placebo-controlled trial. Lancet. 2014;383(9935):2136–2143. doi:10.1016/S0140-6736(13)62630-6.
  • Bhandari N, Rongsen-Chandola T, Bavdekar A, John J, Antony K, Taneja S, Goyal N, Kawade A, Kang G, Rathore SS, et al. Efficacy of a monovalent human-bovine (116E) rotavirus vaccine in Indian children in the second year of life. Vaccine. 2014;32:A110–A116. doi:10.1016/j.vaccine.2014.04.079.
  • Ella R, Babji S, Ciarlet M, Blackwelder WC, Vadrevu KM. A randomized, open-labelled, non-inferiority phase 4 clinical trial to evaluate the immunogenicity and safety of the live, attenuated, oral rotavirus vaccine, ROTAVAC® in comparison with a licensed rotavirus vaccine in healthy infants. Vaccine. 2019;37(31):4407–4413. doi:10.1016/j.vaccine.2019.05.069.
  • Luna EJA, Frazatti-Gallina NM, Timenetsky MCST, Cardoso MRA, Veras MASM, Miraglia JL, Escobar AMU, Grisi SJFE, Raw I, Precioso AR. A phase I clinical trial of a new 5-valent rotavirus vaccine. Vaccine. 2013;31(7):1100–1105. doi:10.1016/j.vaccine.2012.12.020.
  • Desai S, Rathi N, Kawad A, Venkatramanan P, Kundu R, Laiwani SK, Dubey AP, Rao JV, Narayanappa D, Ghildiyal R, 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(37):5519–5523. doi:10.1016/j.vaccine.2018.07.064.
  • Kawade A, Babji S, Kamath V, Raut A, Kumar CM, Kundu R, Venkatramanan P, Lalwani SK, Bavdekar A, Juvekar S, et al. Immunogenicity and lot-to-lot consistency of a ready to use liquid bovine-human reassortant pentavalent rotavirus vaccine (ROTASIIL - Liquid) in Indian infants. Vaccine. 2019;37(19):2554–2560. doi:10.1016/j.vaccine.2019.03.067.
  • Kulkarni PS, Desai S, Tewari T, Kawade A, Goyal N, Garg BS, Kumar D, Kanungo S, Kamat V, Kang G, 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(45):6228–6237. doi:10.1016/j.vaccine.2017.09.014.
  • Fu CX, Wang M, Liang JH, He TJ, Wang DH, Xu JX. Effectiveness of Lanzhou lamb rotavirus vaccine against rotavirus gastroenteritis requiring hospitalization: A matched case-control study. Vaccine. 2007;25(52):8756–8761. doi:10.1016/j.vaccine.2007.10.036.
  • Fu CX, Tate JE, Jiang BM. Effectiveness of Lanzhou lamb rotavirus vaccine against hospitalized gastroenteritis Further analysis and update. Hum Vaccin. 2010;6(11):953. doi:10.4161/hv.6.11.12847.
  • Fu CX, He Q, Xu JX, Xie HP, Ding P, Hu WS, Dong ZQ, Liu XY, Wang M. Effectiveness of the lanzhou lamb rotavirus vaccine against gastroenteritis among children. Vaccine. 2012;31(1):154–158. doi:10.1016/j.vaccine.2012.10.078.
  • Li J, Zhang Y, Yang Y, Liang ZC, Tian Y, Liu BW, Gao ZY, Jia L, Chen LJ, Wang QY. Effectiveness of Lanzhou lamb rotavirus vaccine in preventing gastroenteritis among children younger than 5 years of age. Vaccine. 2019;37:3611–3616.
  • Le LT, Nguyen TV, Nguyen PM, Huong NT, Huong NT, Huong NTM, Hanh TB, Ha DN, Anh DD, Gentsch JR, et al. Development and characterization of candidate rotavirus vaccine strains derived from children with diarrhoea in Vietnam. Vaccine. 2009;27:F130–F138. doi:10.1016/j.vaccine.2009.08.086.
  • Anh DD, Trang NV, Thiem VD, Nguyen THA, Mao ND, Wang YH, Jiang BM, Hien ND, Luan LT, Grp R-MVT. A dose-escalation safety and immunogenicity study of a new live attenuated human rotavirus vaccine (Rotavin-M1) in Vietnamese children. Vaccine. 2012;30:A114–A121. doi:10.1016/j.vaccine.2011.07.118.
  • Groome MJ, Koen A, Fix A, Page N, Jose L, Madhi SA, McNeal M, Dally L, Cho I, Power M, et al. Safety and immunogenicity of a parenteral P2-VP8-P[8] subunit rotavirus vaccine in toddlers and infants in South Africa: a randomised, double-blind, placebo-controlled trial. Lancet Infect Dis. 2017;17(8):843–853. doi:10.1016/S1473-3099(17)30242-6.
  • Groome MJ, Fairlie L, Morrison J, Fix A, Koen A, Masenya M, Page N, Jose L, Madhi SA, McNeal M, et al. Safety and Immunogenicity of a parenteral P2-VP8 subunit rotavirus vaccine. Presented at: The 13th International Rotavirus Symposium; 2018 Aug 29-31; Minsk, Belarus.
  • Burke RM, Tate JE, Kirkwood CD, Steele AD, Parashar UD. Current and new rotavirus vaccines. Curr Opin Infect Dis. 2019;32(5):435–444. doi:10.1097/QCO.0000000000000572.
  • Glass RI, Noel J, Ando T, Fankhauser R, Belliot G, Mounts A, Parashar UD, Bresee JS, Monroe SS. The epidemiology of enteric caliciviruses from humans: A reassessment using new diagnostics. J Infect Dis. 2000;181(s2):S254–S261. doi:10.1086/315588.
  • Koopmans M, Vinje J, de Wit M, Leenen I, van der Poel W, van Duynhoven Y. Molecular epidemiology of human enteric caliciviruses in The Netherlands. J Infect Dis. 2000;181(s2):S262–S269. doi:10.1086/315573.
  • Dolin R. Perspective: noroviruses - Challenges to control. N Engl J Med. 2007;357(11):1072–1073. doi:10.1056/NEJMp078050.
  • Hall AJ, Lopman BA, Payne DC, Patel MM, Gastanaduy PA, Vinje J, Parashar UD. Norovirus disease in the United States. Emerg Infect Dis. 2013;19(8):1198–1205. doi:10.3201/eid1908.130465.
  • Kroneman A, Vega E, Vennema H, Vinje J, White PA, Hansman G, Green K, Martella V, Katayama K, Koopmans M. Proposal for a unified norovirus nomenclature and genotyping. Arch Virol. 2013;158(10):2059–2068. doi:10.1007/s00705-013-1708-5.
  • Cortes-Penfield NW, Ramani S, Estes MK, Atmar RL. Prospects and Challenges in the development of a Norovirus Vaccine. Clin Ther. 2017;39(8):1537–1549. doi:10.1016/j.clinthera.2017.07.002.
  • Tran TNH, Trainor E, Nakagomi T, Cunliffe NA, Nakagomi O. Molecular epidemiology of noroviruses associated with acute sporadic gastroenteritis in children: global distribution of genogroups, genotypes and GII.4variants. J Clin Virol. 2013;56(3):185–193. doi:10.1016/j.jcv.2012.11.011.
  • Ball JM, Graham DY, Opekun AR, Gilger MA, Guerrero RA, Estes MK. Recombinant Norwalk virus-like particles given orally to volunteers: phase I study. Gastroenterology. 1999;117(1):40–48. doi:10.1016/S0016-5085(99)70548-2.
  • El-Kamary SS, Pasetti MF, Mendelman PM, Frey SE, Bernstein DI, Treanor JJ, Ferreira J, Chen WH, Sublett R, Richardson C, et al. Adjuvanted intranasal norwalk virus-like particle vaccine elicits antibodies and antibody-secreting cells that express homing receptors for mucosal and peripheral lymphoid tissues. J Infect Dis. 2010;202(11):1649–1658. doi:10.1086/657087.
  • Ramirez K, Wahid R, Richardson C, Bargatze RF, El-Kamary SS, Sztein MB, Pasetti MF. Intranasal vaccination with an adjuvanted Norwalk virus-like particle vaccine elicits antigen-specific B memory responses in human adult volunteers. Clin Immunol. 2012;144(2):98–108. doi:10.1016/j.clim.2012.05.006.
  • Parra GI, Bok K, Taylor R, Haynes JR, Sosnovtsev SV, Richardson C, Green KY. Immunogenicity and specificity of norovirus Consensus GII.4 virus-like particles in monovalent and bivalent vaccine formulations. Vaccine. 2012;30(24):3580–3586. doi:10.1016/j.vaccine.2012.03.050.
  • Treanor JJ, Atmar RL, Frey SE, Gormley R, Chen WH, Ferreira J, Goodwin R, Borkowski A, Clemens R, Mendelman PM. A Novel intramuscular bivalent norovirus virus-like particle vaccine candidate-reactogenicity, safety, and immunogenicity in a phase 1 trial in healthy adults. J Infect Dis. 2014;210(11):1763–1771. doi:10.1093/infdis/jiu337.
  • Sundararajan A, Sangster MY, Frey S, Atmar RL, Chen WH, Ferreira J, Bargatze R, Mendelman PM, Treanor JJ, Topham DJ. Robust mucosal-homing antibody-secreting B cell responses induced by intramuscular administration of adjuvanted bivalent human norovirus-like particle vaccine. Vaccine. 2015;33(4):568–576. doi:10.1016/j.vaccine.2014.09.073.
  • Ramani S, Neill FH, Ferreira J, Treanor JJ, Frey SE, Topham DJ, Goodwin RR, Borkowski A, Baehner F, Mendelman PM, et al. B-cell responses to intramuscular administration of a bivalent virus-like particle Human Norovirus Vaccine. Clin Vaccine Immunol. 2017;24(5):e00571–16. doi:10.1128/CVI.00571-16.
  • Bernstein DI, Atmar RL, Lyon GM, Treanor JJ, Chen WH, Jiang X, Vinje J, Gregoricus N, Frenck RW, Moe CL, et al. Norovirus vaccine against experimental human gii.4 virus illness: a challenge study in healthy adults. J Infect Dis. 2015;211(6):870–878. doi:10.1093/infdis/jiu497.
  • Blazevic V, Lappalainen S, Nurminen K, Huhti L, Vesikari T. Norovirus VLPs and rotavirus VP6 protein as combined vaccine for childhood gastroenteritis. Vaccine. 2011;29(45):8126–8133. doi:10.1016/j.vaccine.2011.08.026.
  • Tamminen K, Lappalainen S, Huhti L, Vesikari T, Blazevic V. Trivalent combination vaccine induces broad heterologous immune responses to norovirus and rotavirus in mice. PLoS One. 2013;8(7):e70409. doi:10.1371/journal.pone.0070409.
  • Tan M, Jiang X. The P domain of norovirus capsid protein forms a subviral particle that binds to histo-blood group antigen receptors. J Virol. 2005;79(22):14017–14030. doi:10.1128/JVI.79.22.14017-14030.2005.
  • Fang H, Tan M, Xia M, Wang LH, Jiang X. Norovirus P Particle efficiently elicits innate, humoral and cellular immunity. PLoS One. 2013;8(4):e63269. doi:10.1371/journal.pone.0063269.
  • Baric RS, Yount B, Lindesmith L, Harrington PR, Greene SR, Tseng FC, Davis N, Johnston RE, Klapper DG, Moe CL. Expression and self-assembly of norwalk virus capsid protein from venezuelan equine encephalitis virus replicons. J Virol. 2002;76(6):3023–3030. doi:10.1128/JVI.76.6.3023-3030.2002.
  • Guo L, Wang JW, Zhou HL, Si HL, Wang M, Song JD, Han BJ, Shu Y, Ren LL, Qu J, et al. Intranasal administration of a recombinant adenovirus expressing the norovirus capsid protein stimulates specific humoral, mucosal, and cellular immune responses in mice. Vaccine. 2008;26(4):460–468. doi:10.1016/j.vaccine.2007.11.039.
  • Ramani S, Estes MK, Atmar RL. Norovirus vaccine development. In: Viral Gastroenteritis. MA: Academic Press; 2016. p. 447–469.
  • Hansman GS, Natori K, Shirato-Horikoshi H, Ogawa S, Oka T, Katayama K, Tanaka T, Miyoshi T, Sakae K, Kobayashi S, et al. Genetic and antigenic diversity among noroviruses. J Gen Virol. 2006;87(4):909–919. doi:10.1099/vir.0.81532-0.
  • Parrino TA, Schreiber DS, Trier JS, Kapikian AZ, Blacklow NR. Clinical immunity in acute gastroenteritis caused by norwalk agent. N Engl J Med. 1977;297(2):86–89. doi:10.1056/NEJM197707142970204.
  • Cukor G, Nowak NA, Blacklow NR. Immunoglobulin-m responses to the norwalk virus of gastroenteritis. Infect Immun. 1982;37(2):463–468. doi:10.1128/IAI.37.2.463-468.1982.
  • Johnson PC, Mathewson JJ, Dupont HL, Greenberg HB. Multiple-challenge study of host susceptibility to norwalk gastroenteritis in United-States Adults. J Infect Dis. 1990;161(1):18–21. doi:10.1093/infdis/161.1.18.
  • Lindesmith L, Moe C, Marionneau S, Ruvoen N, Jiang X, Lindbland L, Stewart P, LePendu J, Baric R. Human susceptibility and resistance to Norwalk virus infection. Nat Med. 2003;9(5):548–553. doi:10.1038/nm860.
  • Simmons K, Gambhir M, Leon J, Lopman B. Duration of Immunity to Norovirus Gastroenteritis. Emerg Infect Dis. 2013;19(8):1260–1267. doi:10.3201/eid1908.130472.
  • Reeck A, Kavanagh O, Estes MK, Opekun AR, Gilger MA, Graham DY, Atmar RL. Serological correlate of protection against norovirus-induced gastroenteritis. J Infect Dis. 2010;202(8):1212–1218. doi:10.1086/656364.
  • Atmar RL, Bernstein DI, Lyon GM, Treanor JJ, Al-Ibrahim MS, Graham DY, Vinje J, Jiang X, Gregoricus N, Frenck RW, et al. Serological correlates of protection against a gii.4 norovirus. Clin Vaccine Immunol. 2015;22(8):923–929. doi:10.1128/CVI.00196-15.
  • Ramani S, Estes MK, Atmar RL. Correlates of protection against norovirus infection and disease-where are we now, where do we go? PLoS Pathog. 2016;12(4):e1005334. doi:10.1371/journal.ppat.1005334.
  • Xia M, Wei C, Wang L, Cao D, Meng XJ, Jiang X, Tan M. A trivalent vaccine candidate against hepatitis E virus, norovirus, and astrovirus. Vaccine. 2016;34(7):905–913. doi:10.1016/j.vaccine.2015.12.068.
  • Bidokhti MRM, Ullman K, Hammer AS, Jensen TH, Chriel M, Byrareddy SN, Baule C. Immunogenicity and efficacy evaluation of subunit astrovirus vaccines. Vaccines (Basel). 2019;7(3):79. doi:10.3390/vaccines7030079.
  • Raqib R, Mia SMS, Qadri F, Alam TI, Alam NH, Chowdhury AK, Mathan MM, Andersson J. Innate immune responses in children and adults with shigellosis. Infect Immun. 2000;68(6] has been updated. OK?):3620–3629. doi:10.1128/IAI.68.6.3620-3629.2000.
  • TranVan Nhieu G, Sansonetti P. The cellular microbiology of Shigella invasion of epithelial cells. In: William D, Picking WLP, editors. Shigella: molecular and cellular biology. Lawrence (KS): Caister Academic Press; 2015. p. 151–168.
  • Scheiring J, Andreoli SP, Zimmerhackl LB. Treatment and outcome of shiga-toxin-associated hemolytic uremic syndrome (HUS). Pediatr Nephrol. 2008;23(10):1749–1760. doi:10.1007/s00467-008-0935-6.
  • Dupont HL, Levine MM, Hornick RB, Formal SB. Inoculum size in shigellosis and Implications for expected mode of transmission. J Infect Dis. 1989;159(6):1126–1128. doi:10.1093/infdis/159.6.1126.
  • Cohen D, Green M, Block C, Slepon R, Ambar R, Wasserman SS, Levine MM. Reduction of transmission of shigellosis by control of houseflies (Musca-Domestica). Lancet. 1991;337:993–997.
  • Farag TH, Faruque AS, Wu Y, Das SK, Hossain A, Ahmed S, Ahmed D, Nasrin D, Kotloff KL, Panchilangam S, et al. Housefly population density correlates with shigellosis among children in Mirzapur, Bangladesh: a time series analysis. PLoS Negl Trop Dis. 2013;7:e2280.
  • Tribble DR. Resistant pathogens as causes of traveller’s diarrhea globally and impact(s) on treatment failure and recommendations. J Travel Med. 2017;24(suppl_1):S6–S12. doi:10.1093/jtm/taw090.
  • Humphries RM, Schuetz AN. Antimicrobial susceptibility testing of bacteria that cause gastroenteritis. Clin Lab Med. 2015;35(2):313–331. doi:10.1016/j.cll.2015.02.005.
  • Niyogi SK. Increasing antimicrobial resistance-an emerging problem in the treatment of shigellosis. Clin Microbiol Infect. 2007;13(12):1141–1143. doi:10.1111/j.1469-0691.2007.01829.x.
  • Puzari M, Sharma M, Chetia P. Emergence of antibiotic resistant Shigella species: A matter of concern. J Infect Public Health. 2018;11(4):451–454. doi:10.1016/j.jiph.2017.09.025.
  • Baker S, The HC. Recent insights into Shigella: a major contributor to the global diarrhoeal disease burden. Curr Opin Infect Dis. 2018;31(5):449–454. doi:10.1097/QCO.0000000000000475.
  • Shiferaw B, Solghan S, Palmer A, Joyce K, Barzilay EJ, Krueger A, Cieslak P. Antimicrobial susceptibility patterns of shigella isolates in foodborne diseases active surveillance network (FoodNet) sites, 2000-2010. Clin Infect Dis. 2012;54(suppl_5):S458–S463. doi:10.1093/cid/cis230.
  • Kakoullis L, Papachristodoulou E, Chra P, Panos G. Shiga toxin-induced haemolytic uraemic syndrome and the role of antibiotics: a global overview. J Infect. 2019;79(2):75–94. doi:10.1016/j.jinf.2019.05.018.
  • Grif K, Dierich MP, Karch H, Allerberger F. Strain-specific differences in the amount of Shiga toxin released from enterohemorrhagic escherichia coli O157 following exposure to subinhibitory concentrations of antimicrobial agents. Eur J Clin Microbiol Infect Dis. 1998;17(11):761–766. doi:10.1007/s100960050181.
  • Ferreccio C, Prado V, Ojeda A, Cayyazo M, Abrego P, Guers L, Levine MM. Epidemiologic patterns of acute diarrhea and endemic shigella infections in children in a poor periurban setting in Santiago, Chile. Am J Epidemiol. 1991;134(6):614–627. doi:10.1093/oxfordjournals.aje.a116134.
  • Abu-Elyazeed RR, Wierzba TF, Frenck RW, Putnam SD, Rao MR, Savarino SJ, Kamal KA, Peruski LF Jr., Abd-El Messih IA, El-Alkamy SA, et al. Epidemiology of Shigella-associated diarrhea in rural Egyptian children. Am J Trop Med Hyg. 2004;71(3):367–372. doi:10.4269/ajtmh.2004.71.367.
  • Formal SB, Oaks EV, Olsen RE, Wingfield-Eggleston M, Snoy PJ, Cogan JP. Effect of prior infection with virulent Shigella flexneri 2a on the resistance of monkeys to subsequent infection with Shigella sonnei. J Infect Dis. 1991;164(3):533–537. doi:10.1093/infdis/164.3.533.
  • Kotloff KL, Nataro JP, Losonsky GA, Wasserman SS, Hale TL, Taylor DN, Sadoff JC, Levine MM. A modified Shigella volunteer challenge model in which the inoculum is administered with bicarbonate buffer: clinical experience and implications for Shigella infectivity. Vaccine. 1995;13(16):1488–1494. doi:10.1016/0264-410X(95)00102-7.
  • Cohen D, Green MS, Block C, Rouach T, Ofek I. Serum antibodies to lipopolysaccharide and natural immunity to shigellosis in an Israeli military population. J Infect Dis. 1988;157(5):1068–1071. doi:10.1093/infdis/157.5.1068.
  • Cohen D, Green MS, Block C, Slepon R, Ofek I. Prospective study of the association between serum antibodies to lipopolysaccharide O antigen and the attack rate of shigellosis. J Clin Microbiol. 1991;29(2):386–389. doi:10.1128/JCM.29.2.386-389.1991.
  • Livio S, Strockbine NA, Panchalingam S, Tennant SM, Barry EM, Marohn ME, Antonio M, Hossain A, Mandomando I, Ochieng JB, et al. Shigella isolates from the global enteric multicenter study inform vaccine development. Clin Infect Dis. 2014;59(7):933–941. doi:10.1093/cid/ciu468.
  • Levine MM, Kotloff KL, Barry EM, Pasetti MF, Sztein MB. Clinical trials of Shigella vaccines: two steps forward and one step back on a long, hard road. Nat Rev Microbiol. 2007;5(7):540–553. doi:10.1038/nrmicro1662.
  • Mani S, Wierzba T, Walker RI. Status of vaccine research and development for Shigella. Vaccine. 2016;34(26):2887–2894. doi:10.1016/j.vaccine.2016.02.075.
  • Turbyfill KR, Hartman AB, Oaks EV. Isolation and characterization of a Shigella flexneri invasin complex subunit vaccine. Infect Immun. 2000;68(12):6624–6632. doi:10.1128/IAI.68.12.6624-6632.2000.
  • Li A, Rong ZC, Ekwall E, Forsum U, Lindberg AA. Serum antibody-responses against shigella lipopolysaccharides and invasion plasmid-coded antigens in shigella infected swedish patients. Scand J Infect Dis. 1993;25(5):569–577. doi:10.3109/00365549309008545.
  • Li A, Zhao CR, Ekwall E, Lindberg AA. Serum IgG antibody responses to shigella invasion plasmid-coded antigens detected by immunoblot. Scand J Infect Dis. 1994;26(4):435–445. doi:10.3109/00365549409008617.
  • Oberhelman RA, Kopecko DJ, Salazarlindo E, Gotuzzo E, Buysse JM, Venkatesan MM, Yi A, Fernandezprada C, Guzman M, Leonbarua R, et al. Prospective study of systemic and mucosal immune responses in dysenteric patients to specific Shigella invasion plasmid antigens and lipopolysaccharides.. Infect Immun. 1991;59(7] has been updated. OK?):2341–2350. doi:10.1128/IAI.59.7.2341-2350.1991.
  • Vandeverg LL, Herrington DA, Boslego J, Lindberg AA, Levine MM. Age-Specific prevalence of serum antibodies to the invasion plasmid and lipopolysaccharide antigens of shigella species in chilean and North American Populations. J Infect Dis. 1992;166(1] has been updated. OK?):158–161. doi:10.1093/infdis/166.1.158.
  • Martinez-Becerra FJ, Chen XT, Dickenson NE, Choudhari SP, Harrison K, Clements JD, Picking WD, Van De Verg LL, Walker RI, Picking WL. Characterization of a novel fusion protein from IpaB and IpaD of Shigella spp.and its potential as a pan-Shigella vaccine. Infect Immun. 2013;81(12):4470–4477. doi:10.1128/IAI.00859-13.
  • Roy C, Clements JD, Picking WD, Freytag LC. Protective vaccine against shigellosis composed of a Shigella IpaB-IpaD fusion protein (DBF) in combination with the adjuvant dmLT. Presented at: Vaccines for Enteric Diseases; 2017 Oct 9-11; Albufeira, Portugal.
  • Hartman AB, Venkatesan MM. Construction of a Stable attenuated shigella sonnei δvirg vaccine strain, wrss1, and protective efficacy and immunogenicity in the guinea pig keratoconjunctivitis model. Infect Immun. 1998;66(9):4572–4576. doi:10.1128/IAI.66.9.4572-4576.1998.
  • Kotloff KL, Taylor DN, Sztein MB, Wasserman SS, Losonsky GA, Nataro JP, Venkatesan M, Hartman A, Picking WD, Katz DE, et al. Phase I evaluation of δvirg shigella sonnei live, attenuated, oral vaccine strain wrss1 in healthy adults. Infect Immun. 2002;70(4):2016–2021. doi:10.1128/IAI.70.4.2016-2021.2002.
  • Orr N, Katz DE, Atsmon J, Radu P, Yavzori M, Halperin T, Sela T, Kayouf R, Klein Z, Ambar R, et al. Community-based safety, immunogenicity, and transmissibility study of the Shigella sonnei WRSS1 vaccine in Israeli volunteers. Infect Immun. 2005;73(12):8027–8032. doi:10.1128/IAI.73.12.8027-8032.2005.
  • Raqib R, Sarker P, Zaman K, Alam NH, Wierzba TF, Maier N, Talukder K, Baqui AH, Suvarnapunya AE, Qadri F, et al. A phase I trial of WRSS1, a Shigella sonnei live oral vaccine in Bangladeshi adults and children. Hum Vaccin Immunother. 2019;15(6):1326–1337. doi:10.1080/21645515.2019.1575165.
  • Pitisuttithum P, Islam D, Chamnanchanunt S, Ruamsap N, Khantapura P, Kaewkungwal J, Kittitrakul C, Luvira V, Dhitavat J, Venkatesan MM, et al. Clinical trial of an oral live shigella sonnei vaccine candidate, wrss1, in thai adults. Clin Vaccine Immunol. 2016;23(7):564–575. doi:10.1128/CVI.00665-15.
  • Barnoy S, Jeong KI, Helm RF, Suvarnapunya AE, Ranallo RT, Tzipori S, Venkatesan MM. Characterization of WRSs2 and WRSs3, new second-generation virG(icsA)-based Shigella sonnei vaccine candidates with the potential for reduced reactogenicity. Vaccine. 2010;28(6):1642–1654. doi:10.1016/j.vaccine.2009.11.001.
  • Barnoy S, Baqar S, Kaminski RW, Collins T, Nemelka K, Hale TL, Ranallo RT, Venkatesan MM. Shigella sonnei vaccine candidates WRSs2 and WRSs3 are as immunogenic as WRSS1, a clinically tested vaccine candidate, in a primate model of infection. Vaccine. 2011;29(37):6371–6378. doi:10.1016/j.vaccine.2011.04.115.
  • Frenck RW, Baqar S, Alexander W, Dickey M, McNeal M, El-Khorazaty J, Baughman H, Hoeper A, Barnoy S, Suvarnapunya AE, et al. A Phase I trial to evaluate the safety and immunogenicity of WRSs2 and WRSs3; two live oral candidate vaccines against Shigella sonnei. Vaccine. 2018;36(32):4880–4889. doi:10.1016/j.vaccine.2018.06.063.
  • Kotloff KL, Simon JK, Pasetti MF, Sztein MB, Wooden SL, Livio S, Nataro JP, Blackwelder WC, Barry EM, Picking W, et al. Safety and immunogenicity of CVD 1208s, a live, oral δguaba δsen δset shigella flexneri 2a vaccine grown on animal-free media. Hum Vaccin. 2007;3(6):268–275. doi:10.4161/hv.4746.
  • Kotloff KL, Pasetti MF, Barry EM, Nataro JP, Wasserman SS, Sztein MB, Picking WD, Levine MM. Deletion in the shigella enterotoxin genes further attenuates shigella flexneri 2a bearing guanine auxotrophy in a phase 1 trial of CVD 1204 and CVD 1208. J Infect Dis. 2004;190(10):1745–1754. doi:10.1086/424680.
  • Dharmasena MN, Osorio M, Takeda K, Stibitz S, Kopecko DJ. Stable chromosomal expression of shigella flexneri 2a and 3a o-antigens in the live salmonella oral vaccine vector Ty21a. Clin Vaccine Immunol. 2017;24(12):e00181–17. doi:10.1128/CVI.00181-17.
  • Wu Y, Chakravarty S, Li ML, Wai TT, Hoffman SL, Sim BKL. Development of a live attenuated bivalent oral vaccine against Shigella sonnei shigellosis and typhoid fever. J Infect Dis. 2017;215(2):259–268. doi:10.1093/infdis/jiw528.
  • Wu Y, Wai TT, Jackson JM, Li M, Chakravarty S, James ER, Huang H, Laney V, Zhang W, Sack DA, et al. Development and characterization of Salmonella enterica serovar Typhi Ty21a vaccine platform: the promise and insight for vaccines against Shigellosis, ETEC diarrhea, typhoid fever, and non-typhoid salmonellosis. Presented at: Vaccines for Enteric Diseases; 2017 Oct 9-11; Albufeira, Portugal.
  • Girardi P, Harutyunyan S, Neuhauser I, Szijártó V, Nagy G, Nagy E, Henics T Pre-clinical findings with ShigETEC, a combined Shigella and ETEC vaccine that represents paradigm shifts in Shigella vaccine approaches. Presented at: Vaccines for Enteric Diseases; 2017 Oct 9-11; Albufeira, Portugal.
  • McKenzie R, Walker RI, Nabors GS, Van de Verg LL, Carpenter C, Gomes G, Forbes E, Tian JH, Yang HH, Pace JL, et al. Safety and immunogenicity of an oral, inactivated, whole-cell vaccine for Shigella sonnei: preclinical studies and a Phase I trial. Vaccine. 2006;24(18):3735–3745. doi:10.1016/j.vaccine.2005.07.014.
  • Kaminski RW, Wu M, Turbyfill KR, Clarkson K, Tai B, Bourgeois AL, Van De Verg LL, Walker RI, Oaks EV. Development and Preclinical Evaluation of a Trivalent, Formalin-Inactivated Shigella Whole-Cell Vaccine. Clin Vaccine Immunol. 2014;21(3):366–382. doi:10.1128/CVI.00683-13.
  • Kim MJ, Moon YH, Kim H, Rho S, Shin YK, Song M, Walker R, Czerkinsky C, Kim DW, Kim JO. Cross-protective shigella whole-cell vaccine with a truncated o-polysaccharide chain. Front Microbiol. 2018;9:2609. doi:10.3389/fmicb.2018.02609.
  • Barman S, Koley H, Ramamurthy T, Chakrabarti MK, Shinoda S, Nair GB, Takeda Y. Protective immunity by oral immunization with heat-killed Shigella strains in a guinea pig colitis model. Microbiol Immunol. 2013;57(11):762–771. doi:10.1111/1348-0421.12095.
  • Barman S, Koley H, Nag D, Shinoda S, Nair GB, Takeda Y. Passive immunity with multi-serotype heat-killed Shigellae in neonatal mice. Microbiol Immunol. 2014;58(8):463–466. doi:10.1111/1348-0421.12164.
  • Nag D, Sinha R, Mitra S, Barman S, Takeda Y, Shinoda S, Chakrabarti MK, Koley H. Heat killed multi-serotype Shigella immunogens induced humoral immunity and protection against heterologous challenge in rabbit model. Immunobiology. 2015;220(11):1275–1283. doi:10.1016/j.imbio.2015.07.002.
  • Taylor DN, Trofa AC, Sadoff J, Chu CY, Bryla D, Shiloach J, Cohen D, Ashkenazi S, Lerman Y, Egan W, et al. Synthesis, Characterization, and Clinical-Evaluation of Conjugate Vaccines Composed of the O-Specific Polysaccharides of Shigella dysenteriae type 1, Shigella flexneri type 2a, and Shigella sonnei (Plesiomonas-Shigelloides) Bound to Bacterial Toxoids. Infect Immun. 1993;61(9):3678–3687. doi:10.1128/IAI.61.9.3678-3687.1993.
  • Cohen D, Ashkenazi S, Green M, Lerman Y, Slepon R, Robin G, Orr N, Taylor DN, Sadoff JC, Chu CY, et al. Safety and immunogenicity of investigational Shigella conjugate vaccines in Israeli volunteers.. Infect Immun. 1996;64(10):4074–4077. doi:10.1128/IAI.64.10.4074-4077.1996.
  • Cohen D, Ashkenazi S, Green MS, Gdalevich M, Robin G, Slepon R, Yavzori M, Orr N, Block C, Ashkenazi I, et al. Double-blind vaccine-controlled randomised efficacy trial of an investigational Shigella sonnei conjugate vaccine in young adults. Lancet. 1997;349(9046):155–159. doi:10.1016/S0140-6736(96)06255-1.
  • Passwell JH, Ashkenzi S, Banet-Levi Y, Ramon-Saraf R, Farzam N, Lerner-Geva L, Even-Nir H, Yerushalmi B, Chu CY, Shiloach J, et al. Age-related efficacy of Shigella O-specific polysaccharide conjugates in 1–4-year-old Israeli children. Vaccine. 2010;28(10):2231–2235. doi:10.1016/j.vaccine.2009.12.050.
  • Pavliakova D, Chu CY, Bystricky S, Tolson NW, Shiloach J, Kaufman JB, Bryla DA, Robbins JB, Schneerson R. Treatment with succinic anhydride improves the immunogenicity ofshigella flexneri type 2a o-specific polysaccharide–protein conjugates in mice. Infect Immun. 1999;67(10):5526–5529. doi:10.1128/IAI.67.10.5526-5529.1999.
  • Pavliakova D, Moncrief JS, Lyerly DM, Schiffman G, Bryla DA, Robbins JB, Schneerson R. Clostridium difficile recombinant toxin A repeating units as a carrier protein for conjugate vaccines: studies of pneumococcal type 14, Escherichia coli K1, and Shigella flexneri type 2a polysaccharides in mice. Infect Immun. 2000;68:2161–2166.
  • Feldman MF, Wacker M, Hernandez M, Hitchen PG, Marolda CL, Kowarik M, Morris HR, Dell A, Valvano MA, Aebi M. Engineering N-linked protein glycosylation with diverse O antigen lipopolysaccharide structures in Escherichia coli. Proc Natl Acad Sci USA. 2005;102(8):3016–3021. doi:10.1073/pnas.0500044102.
  • Vozza NF, Feldman MF. Glyco-engineering O-antigen-based vaccines and diagnostics in E. coli. Methods Mol Biol. 2015;1321:57–70.
  • Barel LA, Mulard LA. Classical and novel strategies to develop Shigella glycoconjugate vaccine: from concept to efficacy in human. Hum Vaccin Immunother. 2019;15(6):1338–1356. doi:10.1080/21645515.2019.1606972.
  • Hatz CFR, Bally B, Rohrer S, Steffen R, Kramme S, Siegrist CA, Wacker M, Alaimo C, Fonck VG. Safety and immunogenicity of a candidate bioconjugate vaccine against Shigella dysenteriae type 1 administered to healthy adults: A single blind, partially randomized Phase I study. Vaccine. 2015;33(36):4594–4601. doi:10.1016/j.vaccine.2015.06.102.
  • Riddle MS, Kaminski RW, Di Paolo C, Porter CK, Gutierrez RL, Clarkson KA, Weerts HE, Duplessis C, Castellano A, Alaimo C, et al. Safety and immunogenicity of a candidate bioconjugate vaccine against shigella flexneri 2a administered to healthy adults: a single-blind, randomized phase i study. Clin Vaccine Immunol. 2016;23(12] has been updated. OK?):908–917. doi:10.1128/CVI.00224-16.
  • Pozsgay V, Chu CY, Pannell L, Wolfe J, Robbins JB, Schneerson R. Protein conjugates of synthetic saccharides elicit higher levels of serum IgG lipopolysaccharide antibodies in mice than do those of the O-specific polysaccharide from Shigella dysenteriae type 1. Proc Natl Acad Sci U S A. 1999;96(9] has been updated. OK?):5194–5197. doi:10.1073/pnas.96.9.5194.
  • Phalipon A, Tanguy M, Grandjean C, Guerreiro C, Belot F, Cohen D, Sansonetti PJ, Mulard LA. A synthetic carbohydrate-protein conjugate vaccine candidate against shigella flexneri 2a Infection. J Immunol. 2009;182(4):2241–2247. doi:10.4049/jimmunol.0803141.
  • Gerke C, Colucci AM, Giannelli C, Sanzone S, Vitali CG, Sollai L, Rossi O, Martin LB, Auerbach J, Di Cioccio V, et al. Production of a shigella sonnei vaccine based on Generalized Modules for Membrane Antigens (GMMA), 1790GAHB. PLoS One. 2015;10(8):e0134478. doi:10.1371/journal.pone.0134478.
  • Obiero CW, Ndiaye AGW, Scire AS, Kaunyangi BM, Marchetti E, Gone AM, Schutte LD, Riccucci D, Auerbach J, Saul A, et al. A Phase 2a randomized study to evaluate the safety and immunogenicity of the 1790gahb generalized modules for membrane antigen vaccine against shigella sonnei administered intramuscularly to adults from a shigellosis-Endemic Country. Front Immunol. 2017;8:1884.
  • Oaks EV, Picking WD, Picking WL. Antibody response of monkeys to invasion plasmid antigen D after infection with Shigella spp.. Clin Diagn Lab Immun. 1996;3(2):242–245. doi:10.1128/CDLI.3.2.242-245.1996.
  • Turbyfill KR, Kaminski RW, Oaks EV. Immunogenicity and efficacy of highly purified invasin complex vaccine from Shigella flexneri 2a. Vaccine. 2008;26(10):1353–1364. doi:10.1016/j.vaccine.2007.12.040.
  • Hueck CJ. Type III protein secretion systems in bacterial pathogens of animals and plants. Microbiol Mol Biol Rev. 1998;62:379–433.
  • Oaks EV, Turbyfill KR. Development and evaluation of a Shigella flexneri 2a and S. sonnei bivalent invasin complex (Invaplex) vaccine. Vaccine. 2006;24(13):2290–2301. doi:10.1016/j.vaccine.2005.11.040.
  • Tribble D, Kaminski R, Cantrell J, Nelson M, Porter C, Baqar S, Williams C, Arora R, Saunders J, Ananthakrishnan M, et al. Safety and immunogenicity of a Shigella flexneri 2a Invaplex 50 intranasal vaccine in adult volunteers. Vaccine. 2010;28(37):6076–6085. doi:10.1016/j.vaccine.2010.06.086.
  • Riddle MS, Kaminski RW, Williams C, Porter C, Baqar S, Kordis A, Gilliland T, Lapa J, Coughlin M, Soltis C, et al. Safety and immunogenicity of an intranasal Shigella flexneri 2a Invaplex 50 vaccine. Vaccine. 2011;29(40):7009–7019. doi:10.1016/j.vaccine.2011.07.033.
  • Turbyfill KR, Clarkson KA, Vortherms AR, Oaks EV, Kaminski RW. Assembly, Biochemical Characterization, Immunogenicity, Adjuvanticity, and Efficacy of Shigella Artificial Invaplex. Msphere. 2018;3(2):e00583–17. doi:10.1128/mSphere.00583-17.
  • World Health Organization. Future directions for research on enterotoxigenic Escherichia coli vaccines for developing countries.. Wkly Epidemiol Rec. 2006;81(11):97–107.
  • Sanders JW, Putnam SD, Riddle MS, Tribble DR. Military importance of diarrhea: lessons from the Middle East. Curr Opin Gastroenterol. 2005;21:9–14.
  • Jiang ZD, DuPont HL. Etiology of travellers’ diarrhea. J Travel Med. 2017;24(suppl_1):S13–S16. doi:10.1093/jtm/tax003.
  • Nataro JP, Kaper JB. Diarrheagenic Escherichia coli. Clin Microbiol Rev. 1998;11:142–201.
  • Svennerholm AM, Tobias J. Vaccines against enterotoxigenic Escherichia coli. Expert Rev Vaccines. 2008;7(6):795–804. doi:10.1586/14760584.7.6.795.
  • Zhang W, Sack DA. Progress and hurdles in the development of vaccines against enterotoxigenic Escherichia coli in humans. Expert Rev Vaccines. 2012;11(6):677–684. doi:10.1586/erv.12.37.
  • Walker RI. An assessment of enterotoxigenic Escherichia coli and Shigella vaccine candidates for infants and children. Vaccine. 2015;33(8):954–965. doi:10.1016/j.vaccine.2014.11.049.
  • Zhang W, Sack DA. Current progress in developing subunit vaccines against enterotoxigenic escherichia coli -associated diarrhea. Clin Vaccine Immunol. 2015;22(9):983–991. doi:10.1128/CVI.00224-15.
  • Walker R, Dull P. Combination vaccine strategies to prevent enteric infections. Vaccine. 2017;35(49):6790–6792. doi:10.1016/j.vaccine.2017.06.076.
  • Qadri F, Svennerholm AM, Faruque AS, Sack RB. Enterotoxigenic Escherichia coli in developing countries: epidemiology, microbiology, clinical features, treatment, and prevention. Clin Microbiol Rev. 2005;18(3):465–483. doi:10.1128/CMR.18.3.465-483.2005.
  • Svennerholm AM, Lundgren A. Recent progress toward an enterotoxigenic Escherichia coli vaccine. Expert Rev Vaccines. 2012;11(4] has been updated. OK?):495–507. doi:10.1586/erv.12.12.
  • Isidean SD, Riddle MS, Savarino SJ, Porter CK. A systematic review of ETEC epidemiology focusing on colonization factor and toxin expression. Vaccine. 2011;29(37):6167–6178. doi:10.1016/j.vaccine.2011.06.084.
  • Jertborn M, Ahren C, Holmgren J, Svennerholm AM. Safety and immunogenicity of an oral inactivated enterotoxigenic Escherichia coli vaccine. Vaccine. 1998;16(2–3):255–260. doi:10.1016/S0264-410X(97)00169-2.
  • Lebens M, Shahabi V, Backstrom M, Houze T, Lindblad M, Holmgren J. Synthesis of hybrid molecules between heat-labile enterotoxin and cholera toxin B subunits: potential for use in a broad-spectrum vaccine. Infect Immun. 1996;64(6] has been updated. OK?):2144–2150. doi:10.1128/IAI.64.6.2144-2150.1996.
  • Ahren C, Jertborn M, Svennerholm AM. Intestinal immune responses to an inactivated oral enterotoxigenic Escherichia coli vaccine and associated immunoglobulin A responses in blood. Infect Immun. 1998;66(7 et al., 2016] has been updated. OK?):3311–3316. doi:10.1128/IAI.66.7.3311-3316.1998.
  • Cohen D, Orr N, Haim M, Ashkenazi S, Robin G, Green MS, Ephros M, Sela T, Slepon R, Ashkenazi I, et al. Safety and immunogenicity of two different lots of the oral, killed enterotoxigenic Escherichia coli-Cholera toxin B subunit vaccine in Israeli young adults. Infect Immun. 2000;68(8):4492–4497. doi:10.1128/IAI.68.8.4492-4497.2000.
  • Qadri F, Wenneras C, Ahmed F, Asaduzzaman M, Saha D, Albert MJ, Sack RB, Svennerholm A. Safety and immunogenicity of an oral, inactivated enterotoxigenic Escherichia coli plus cholera toxin B subunit vaccine in Bangladeshi adults and children. Vaccine. 2000;18(24):2704–2712. doi:10.1016/S0264-410X(00)00056-6.
  • Savarino SJ, Hall ER, Bassily S, Brown FM, Youssef F, Wierzba TF, Peruski L, El-Masry NA, Safwat M, Rao M, et al. Oral, Inactivated, whole cell enterotoxigenic escherichia coli plus cholera toxin b subunit vaccine: results of the initial evaluation in children. J Infect Dis. 1999;179(1):107–114. doi:10.1086/314543.
  • Qadri F, Ahmed T, Ahmed F, Bradley Sack R, Sack DA, Svennerholm AM. Safety and immunogenicity of an oral, inactivated enterotoxigenic Escherichia coli plus cholera toxin B subunit vaccine in Bangladeshi children 18-36 months of age. Vaccine. 2003;21(19–20] has been updated. OK?):2394–2403. doi:10.1016/S0264-410X(03)00077-X.
  • Savarino SJ, Hall ER, Bassily S, Wierzba TF, Youssef FG, Peruski LF Jr., Abu-Elyazeed R, Rao M, Francis WM, El Mohamady H, et al. Introductory evaluation of an oral, killed whole cell enterotoxigenic Escherichia coli plus cholera toxin B subunit vaccine in Egyptian infants. Pediatr Infect Dis J. 2002;21(4):322–330. doi:10.1097/00006454-200204000-00012.
  • Sack DA, Shimko J, Torres O, Bourgeois AL, Francia DS, Gustafsson B, Karnell A, Nyquist I, Svennerholm AM. Randomised, double-blind, safety and efficacy of a killed oral vaccine for enterotoxigenic E. coli diarrhoea of travellers to Guatemala and Mexico. Vaccine. 2007;25(22):4392–4400. doi:10.1016/j.vaccine.2007.03.034.
  • Holmgren J, Bourgeois L, Carlin N, Clements J, Gustafsson B, Lundgren A, Nygren E, Tobias J, Walker R, Svennerholm AM. Development and preclinical evaluation of safety and immunogenicity of an oral ETEC vaccine containing inactivated E. coli bacteria overexpressing colonization factors CFA/I, CS3, CS5 and CS6 combined with a hybrid LT/CT B subunit antigen, administered alone and together with dmLT adjuvant.. Vaccine. 2013;31(20):2457–2464. doi:10.1016/j.vaccine.2013.03.027.
  • Lundgren A, Bourgeois L, Carlin N, Clements J, Gustafsson B, Hartford M, Holmgren J, Petzold M, Walker R, Svennerholm AM. Safety and immunogenicity of an improved oral inactivated multivalent enterotoxigenic Escherichia coli (ETEC) vaccine administered alone and together with dmLT adjuvant in a double-blind, randomized, placebo-controlled Phase I study. Vaccine. 2014;32(52):7077–7084. doi:10.1016/j.vaccine.2014.10.069.
  • Akhtar M, Chowdhury MI, Bhuiyan TR, Kaim J, Ahmed T, Rafique TA, Khan A, Rahman SIA, Khanam F, Begum YA, et al. Evaluation of the safety and immunogenicity of the oral inactivated multivalent enterotoxigenic Escherichia coli vaccine ETVAX in Bangladeshi adults in a double-blind, randomized, placebo-controlled Phase I trial using electrochemiluminescence and ELISA assays for immunogenicity analyses.. Vaccine. 2019;37(37):5645–5656. doi:10.1016/j.vaccine.2018.11.040.
  • Leach S, Lundgren A, Carlin N, Lofstrand M, Svennerholm AM. Cross-reactivity and avidity of antibody responses induced in humans by the oral inactivated multivalent enterotoxigenic Escherichia coli (ETEC) vaccine ETVAX. Vaccine. 2017;35(32):3966–3973. doi:10.1016/j.vaccine.2017.06.006.
  • Qadri F, Akhtar M, Bhuiyan TR, Chowdhury MI, Ahmed T, Rafique TA, Khan A, Rahman SIA, Khanam F, Lundgren A, et al. Safety and immunogenicity of the oral, inactivated, enterotoxigenic Escherichia coli vaccine ETVAX in Bangladeshi children and infants: a double-blind, randomised, placebo-controlled phase 1/2 trial. Lancet Infect Dis. 2020;20(2):208–219. doi:10.1016/S1473-3099(19)30571-7.
  • Seo H, Zhang W. Development of effective vaccines for enterotoxigenic Escherichia coli. Lancet Infect Dis. 2020;20(2):150–152. doi:10.1016/S1473-3099(19)30631-0.
  • Turner AK, Stephens JC, Beavis JC, Greenwood J, Gewert C, Randall R, Freeman D, Darsley MJ. Generation and Characterization of a Live Attenuated Enterotoxigenic Escherichia coli Combination Vaccine Expressing Six Colonization Factors and Heat-Labile Toxin Subunit B. Clin Vaccine Immunol. 2011;18(12):2128–2135. doi:10.1128/CVI.05345-11.
  • Harro C, Sack D, Bourgeois AL, Walker R, Denearing B, Feller A, Chakraborty S, Buchwaldt C, Darsley MJ. A combination vaccine consisting of three live attenuated enterotoxigenic Escherichia coli strains expressing a range of colonization factors and heat-labile toxin subunit B is well tolerated and immunogenic in a placebo-controlled double-blind phase I trial in healthy adults.. Clin Vaccine Immunol. 2011;18(12] has been updated. OK?):2118–2127. doi:10.1128/CVI.05342-11.
  • Harro C, David S, Darsley M, Bourgeois AL, DeNearing B, Feller A, Chakraborty S, Marcum A, Comendador R, Buchwaldt C, et al. Volunteers receiving live attenuated ETEC vaccine (ACE527) have reduced severity of illness following H10407 challenge. Presented at: Vaccines for Enteric Diseases; 2011 Sep 14–16; Cannes, France.
  • Darsley MJ, Chakraborty S, DeNearing B, Sack DA, Feller A, Buchwaldt C, Bourgeois AL, Walker R, Harro CD. The oral, live attenuated enterotoxigenic Escherichia coli vaccine ACE527 reduces the incidence and severity of diarrhea in a human challenge model of diarrheal disease. Clin Vaccine Immunol. 2012;19(12):1921–1931. doi:10.1128/CVI.00364-12.
  • Harro C, Louis Bourgeois A, Sack D, Walker R, DeNearing B, Brubaker J, Maier N, Fix A, Dally L, Chakraborty S, et al. Live attenuated enterotoxigenic Escherichia coli (ETEC) vaccine with dmLT adjuvant protects human volunteers against virulent experimental ETEC challenge. Vaccine. 2019;37(14):1978–1986. doi:10.1016/j.vaccine.2019.02.025.
  • Sincock SA, Hall ER, Woods CM, O’Dowd A, Poole ST, McVeigh AL, Nunez G, Espinoza N, Miller M, Savarino SJ. Immunogenicity of a prototype enterotoxigenic Escherichia coli adhesin vaccine in mice and nonhuman primates. Vaccine. 2016;34(2):284–291. doi:10.1016/j.vaccine.2015.11.017.
  • Rollenhagen JE, Woods CM, O’Dowd A, Poole ST, Tian JH, Guebre-Xabier M, Ellingsworth L, Prouty MG, Glenn G, Savarino SJ. Evaluation of transcutaneous immunization as a delivery route for an enterotoxigenic E. coli adhesin-based vaccine with CfaE, the colonization factor antigen 1 (CFA/I) tip adhesin. Vaccine. 2019;37(42):6134–6138. doi:10.1016/j.vaccine.2019.08.057.
  • Savarino SJ, McKenzie R, Tribble DR, Porter CK, O’Dowd A, Cantrell JA, Sincock SA, Poole ST, DeNearing B, Woods CM, et al. prophylactic efficacy of hyperimmune bovine colostral antiadhesin antibodies against enterotoxigenic escherichia coli diarrhea: a randomized, double-blind, placebo-controlled, phase 1 trial. J Infect Dis. 2017;216(1):7–13. doi:10.1093/infdis/jix144.
  • Anantha RP, McVeigh AL, Lee LH, Agnew MK, Cassels FJ, Scott DA, Whittam TS, Savarino SJ. Evolutionary and functional relationships of colonization factor antigen i and other class 5 adhesive fimbriae of enterotoxigenic Escherichia coli. Infect Immun. 2004;72(12):7190–7201. doi:10.1128/IAI.72.12.7190-7201.2004.
  • Seo H, Ruan X, Duan Q, Garcia C, Sack DA, Zhang W MecVax, a broadly protective subunit vaccine against enterotoxigenic Escherichia coli (ETEC) diarrhea. Presented at: Vaccines for Enteric Diseases; 2019 Oct 16-18; Lausanne, Switzerland.
  • Ruan X, Robertson DC, Nataro JP, Clements JD, Zhang W. Characterization of Heat-Stable (STa) Toxoids of Enterotoxigenic Escherichia coli Fused to Double Mutant Heat-Labile Toxin Peptide in Inducing Neutralizing Anti-STa Antibodies. Infect Immun. 2014;82(5):1823–1832. doi:10.1128/IAI.01394-13.
  • Nandre R, Ruan X, Duan Q, Zhang W. Enterotoxigenic Escherichia coli heat-stable toxin and heat-labile toxin toxoid fusion 3xSTa N12S -dmLT induces neutralizing anti-STa antibodies in subcutaneously immunized mice. FEMS Microbiol Lett. 2016;363(21] has been updated. OK?):1–6. doi:10.1093/femsle/fnw246.
  • Duan Q, Huang J, Xiao N, Seo H, Zhang W. Neutralizing anti-STa antibodies derived from enterotoxigenic Escherichia coli (ETEC) toxoid fusions with heat-stable toxin (STa) mutant STaN12S, STaL9A/N12S or STaN12S/A14T show little cross-reactivity with guanylin or uroguanylin. Appl Environ Microbiol. 2018;84:e01737–17.
  • Nandre RM, Duan Q, Wang Y, Zhang W. Passive antibodies derived from intramuscularly immunized toxoid fusion 3xSTa N12S -dmLT protect against STa+ enterotoxigenic Escherichia coli (ETEC) diarrhea in a pig model. Vaccine. 2017;35(4):552–556. doi:10.1016/j.vaccine.2016.12.021.
  • Seo H, Lu T, Nandre RM, Duan Q, Zhang W. Immunogenicity characterization of genetically fused or chemically conjugated heat-stable toxin toxoids of enterotoxigenic Escherichia coli in mice and pigs. FEMS Microbiol Lett. 2019;366(4):fnz037. doi:10.1093/femsle/fnz037.
  • Ruan X, Knudsen DE, Wollenberg KM, Sack DA, Zhang W. Multiepitope fusion antigen induces broadly protective antibodies that prevent adherence of escherichia coli strains expressing colonization factor antigen I (CFA/I), CFA/II, and CFA/IV. Clin Vaccine Immunol. 2014;21(2):243–249. doi:10.1128/CVI.00652-13.
  • Seo H, Lu T, Mani S, Bourgeois AL, Walker R, Sack DA, Zhang W. Adjuvant effect of enterotoxigenic Escherichia coli (ETEC) double-mutant heat-labile toxin (dmLT) on systemic immunogenicity induced by the CFA/I/II/IV MEFA ETEC vaccine: dose-related enhancement of antibody responses to seven ETEC adhesins (CFA/I, CS1-CS6). Hum Vaccin Immunother. 2019;23:1–7.
  • Seo H, Nandre RM, Nietfeld J, Chen Z, Duan Q, Zhang W. Antibodies induced by enterotoxigenic Escherichia coli (ETEC) adhesin major structural subunit and minor tip adhesin subunit equivalently inhibit bacteria adherence in vitro. PLoS One. 2019;14(5):e0216076. doi:10.1371/journal.pone.0216076.
  • Duan Q, Lu T, Garcia C, Yanez C, Nandre RM, Sack DA, Zhang W. Co-administered Tag-Less Toxoid Fusion 3xSTaN12S-mnLTR192G/L211A and CFA/I/II/IV MEFA (Multiepitope Fusion Antigen) Induce Neutralizing Antibodies to 7 Adhesins (CFA/I, CS1-CS6) and Both Enterotoxins (LT, STa) of Enterotoxigenic Escherichia coli (ETEC). Front Microbiol. 2018;9:1198. doi:10.3389/fmicb.2018.01198.
  • Fleckenstein JM, Rasko DA. Overcoming Enterotoxigenic Escherichia coli Pathogen Diversity: translational Molecular Approaches to Inform Vaccine Design. Methods Mol Biol. 2016;1403:363–383.
  • Kuhlmann FM, Martin J, Hazen TH, Vickers TJ, Pashos M, Okhuysen PC, Gomez-Duarte OG, Cebelinski E, Boxrud D, Del Canto F, et al. Conservation and global distribution of non-canonical antigens in Enterotoxigenic Escherichia coli. PLoS Negl Trop Dis. 2019;13(11):e0007825. doi:10.1371/journal.pntd.0007825.
  • Govasli ML, Diaz Y, Puntervoll P. Virus-like particle-display of the enterotoxigenic Escherichia coli heat-stable toxoid STh-A14T elicits neutralizing antibodies in mice. Vaccine. 2019;37(43):6405–6414. doi:10.1016/j.vaccine.2019.09.004.
  • Diaz Y, Govasli ML, Zegeye ED, Sommerfelt H, Steinsland H, Puntervoll P. Immunizations with enterotoxigenic escherichia coli heat-stable toxin conjugates engender toxin-neutralizing antibodies in mice that also cross-react with guanylin and uroguanylin. Infect Immun. 2019;87(7):e00099–19. doi:10.1128/IAI.00099-19.
  • Donnenberg MS, Finlay BB. Combating enteropathogenic Escherichia coli (EPEC) infections: the way forward. Trends Microbiol. 2013;21(7):317–319. doi:10.1016/j.tim.2013.05.003.
  • Sanchez-Villamil JI, Tapia D, Torres AG. Development of a Gold Nanoparticle Vaccine against Enterohemorrhagic Escherichia coli O157:H7. mBio. 2019;10(4):e01869–19. doi:10.1128/mBio.01869-19.
  • Ali M, Nelson AR, Lopez AL, Sack DA. Updated global burden of cholera in endemic countries. PLoS Negl Trop Dis. 2015;9(6):e0003832. doi:10.1371/journal.pntd.0003832.
  • Clemens JD, Harris JR, Khan MR, Kay BA, Yunus MD, Svennerholm AM, Sack DA, Chakraborty J, Stanton BF, Khan MU, et al. Field trial of oral cholera vaccines in Bangladesh. Lancet. 1986;2(8499):124–127. doi:10.1016/S0140-6736(86)91944-6.
  • Clemens JD, Sack DA, Harris JR, Vanloon F, Chakraborty J, Ahmed F, Rao MR, Khan MR, Yunus M, Huda N, et al. Field Trial of Oral Cholera Vaccines in Bangladesh - Results from 3-Year Follow-Up. Lancet. 1990;335(8684):270–273.
  • Clemens JD, Sack DA, Harris JR, Van Loon F, Chakraborty J, Ahmed F, Rao MR, Khan MR, Yunus M, Huda N, et al. Field trial of oral cholera vaccines in Bangladesh: results from three-year follow-up. Lancet. 1990;335(8684):270–273. doi:10.1016/0140-6736(90)90080-O.
  • Trach DD, Cam PD, Ke NT, Rao MR, Dinh D, Hang PV, Hung NV, Canh DG, Thiem VD, Naficy A, et al. Investigations into the safety and immunogenicity of a killed oral cholera vaccine developed in Viet Nam. Bull World Health Organ. 2002;80(1):2–8.
  • Anh DD, Canh DG, Lopez AL, Thiem VD, Long PT, Son NH, Deen J, von Seidlein L, Carbis R, Han SH, et al. Safety and immunogenicity of a reformulated Vietnamese bivalent killed, whole-cell, oral cholera vaccine in adults. Vaccine. 2007;25(6):1149–1155. doi:10.1016/j.vaccine.2006.09.049.
  • Shamsuzzaman S, Ahmed T, Mannoor K, Begum YA, Bardhan PK, Sack RB, Sack DA, Svennerholm AM, Holmgren J, Qadri F. Robust gut associated vaccine-specific antibody-secreting cell responses are detected at the mucosal surface of Bangladeshi subjects after immunization with an oral killed bivalent V. cholerae O1/O139 whole cell cholera vaccine: comparison with other mucosal and systemic responses.. Vaccine. 2009;27(9):1386–1392. doi:10.1016/j.vaccine.2008.12.041.
  • Saha A, Chowdhury MI, Khanam F, Bhuiyan MS, Chowdhury F, Khan AI, Khan IA, Clemens J, Ali M, Cravioto A, et al. Safety and immunogenicity study of a killed bivalent (O1 and O139) whole-cell oral cholera vaccine Shanchol, in Bangladeshi adults and children as young as 1 year of age. Vaccine. 2011;29(46):8285–8292. doi:10.1016/j.vaccine.2011.08.108.
  • Sur D, Kanungo S, Sah B, Manna B, Ali M, Paisley AM, Niyogi SK, Park JK, Sarkar B, Puri MK, et al. Efficacy of a low-cost, inactivated whole-cell oral cholera vaccine: results from 3 years of follow-up of a randomized, controlled trial. PLoS Negl Trop Dis. 2011;5(10):e1289. doi:10.1371/journal.pntd.0001289.
  • Sur D, Lopez AL, Kanungo S, Paisley A, Manna B, Ali M, Niyogi SK, Park JK, Sarkar B, Puri MK, 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(9702):1694–1702. doi:10.1016/S0140-6736(09)61297-6.
  • Bhattacharya SK, Sur D, Ali M, Kanungo S, You YA, Manna B, Sah B, Niyogi SK, Park JK, Sarkar 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(12):1050–1056. doi:10.1016/S1473-3099(13)70273-1.
  • Qadri F, Ali M, Chowdhury F, Khan AI, Saha A, Khan IA, Begum YA, Bhuiyan TQR, Chowdhury MI, Uddin MJ, et al. Feasibility and effectiveness of oral cholera vaccine in an urban endemic setting in Bangladesh: a cluster randomised open-label trial. Lancet. 2015;386(10001):1362–1371. doi:10.1016/S0140-6736(15)61140-0.
  • Qadri F, Wierzba TF, Ali M, Chowdhury F, Khan AI, Saha A, Khan IA, Asaduzzaman M, Akter A, Khan A, et al. Efficacy of a Single-Dose, Inactivated Oral Cholera Vaccine in Bangladesh. N Engl J Med. 2016;374(18):1723–1732. doi:10.1056/NEJMoa1510330.
  • Baik YO, Choi SK, Kim JW, Yang JS, Kim IY, Kim CW, Hong JH. Safety and Immunogenicity Assessment of an Oral Cholera Vaccine through Phase I Clinical Trial in Korea. J Korean Med Sci. 2014;29(4):494–501. doi:10.3346/jkms.2014.29.4.494.
  • Russo P, Ligsay AD, Olveda R, Choi SK, Kim DR, Park JY, Park JY, Syed KA, Dey A, Kim YH, et al. A randomized, observer-blinded, equivalence trial comparing two variations of Euvichol®, a bivalent killed whole-cell oral cholera vaccine, in healthy adults and children in the Philippines. Vaccine. 2018;36(29):4317–4324. doi:10.1016/j.vaccine.2018.05.102.
  • Baik YO, Choi SK, Olveda RM, Espos RA, Ligsay AD, Montellano MB, Yeam JS, Yang JS, Park JY, Kim DR, 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(46):6360–6365. doi:10.1016/j.vaccine.2015.08.075.
  • Levine MM, Herrington D, Losonsky G, Tall B, Kaper JB, Ketley J, Tacket CO, Cryz S. Safety, immunogenicity, and efficacy of recombinant live oral cholera vaccines, CVD-103 and CVD-103-Hgr. Lancet. 1988;2:468–470.
  • Tacket CO, Losonsky G, Nataro JP, Cryz SJ, Edelman R, Kaper JB, Levine MM. Onset and duration of protective immunity in challenged volunteers after vaccination with live oral cholera vaccine CVD l03-HgR. J Infect Dis. 1992;166(4):837–841. doi:10.1093/infdis/166.4.837.
  • Tacket CO, Cohen MB, Wasserman SS, Losonsky G, Livio S, Kotloff K, Edelman R, Kaper JB, Cryz SJ, Giannella RA, et al. Randomized, double-blind, placebo-controlled, multicentered trial of the efficacy of a single dose of live oral cholera vaccine CVD 103-HgR in preventing cholera following challenge with Vibrio cholerae O1 El Tor Inaba three months after vaccination. Infect Immun. 1999;67(12):6341–6345. doi:10.1128/IAI.67.12.6341-6345.1999.
  • Richie E, Punjabi NH, Sidharta Y, Peetosutan K, Sukandar M, Wasserman SS, Lesmana M, Wangsasaputra F, Pandam S, Levine MM, 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(22):2399–2410. doi:10.1016/S0264-410X(00)00006-2.
  • Kenner JR, Coster TS, Taylor DN, Trofa AF, Barreraoro M, Hyman T, Adams JM, Beattie DT, Killeen KP, Spriggs DR, et al. Peru-15, an Improved Live Attenuated Oral Vaccine Candidate for Vibrio Cholerae O1. J Infect Dis. 1995;172(4):1126–1129. doi:10.1093/infdis/172.4.1126.
  • Chowdhury MI, Sheikh A, Qadri F. Development of Peru-15 (CholeraGarde®), a live-attenuated oral cholera vaccine: 1991–2009. Expert Rev Vaccines. 2009;8(12):1643–1652. doi:10.1586/erv.09.137.
  • Cohen MB, Giannella RA, Bean J, Taylor DN, Parker S, Hoeper A, Wowk S, Hawkins J, Kochi SK, Schiff G, et al. Randomized, controlled human challenge study of the safety, immunogenicity, and protective efficacy of a single dose of Peru-15, a live attenuated oral cholera vaccine. Infect Immun. 2002;70(4):1965–1970. doi:10.1128/IAI.70.4.1965-1970.2002.
  • Qadri F, Chowdhury MI, Faruque SM, Salam MA, Ahmed T, Begum YA, Saha A, Al Tarique A, Seidlein LV, Park E, et al. Peru-15, a live attenuated oral cholera vaccine, is safe and immunogenic in Bangladeshi toddlers and infants. Vaccine. 2007;25(2):231–238. doi:10.1016/j.vaccine.2006.08.031.
  • Qadri F, Chowdhury MI, Faruque SM, Salam MA, Ahmed T, Begum YA, Saha A, Alam MS, Zaman K, Seidlein LV, et al. Randomized, controlled study of the safety and immunogenicity of Peru-15, a live attenuated oral vaccine candidate for cholera, in adult volunteers in Bangladesh. J Infect Dis. 2005;192(4):573–579. doi:10.1086/432074.
  • Crim SM, Iwamoto M, Huang JY, Griffin PM, Gilliss D, Cronquist AB, Cartter M, Tobin-D’Angelo M, Blythe D, Smith K, et al. Incidence and trends of infection with pathogens transmitted commonly through food-Foodborne Diseases Active Surveillance Network, 10 U.S. sites, 2006-2013. MMWR Morb Mortal Wkly Rep. 2014;63(15):328–332.
  • Tack DM, Marder EP, Griffin PM, Cieslak PR, Dunn J, Hurd S, Scallan E, Lathrop S, Muse A, Ryan P, et al. Preliminary incidence and trends of infections with pathogens transmitted commonly through food-foodborne diseases active surveillance network, 10 U.S. sites, 2015-2018. MMWR Morb Mortal Wkly Rep. 2019;68(16):369–373. doi:10.15585/mmwr.mm6816a2.
  • Coker AO, Isokpehi RD, Thomas BN, Amisu KO, Obi CL. Human campylobacteriosis in developing countries. Emerg Infect Dis. 2002;8(3):237–244. doi:10.3201/eid0803.010233.
  • Klena JD, Parker CT, Knibb K, Ibbitt JC, Devane PM, Horn ST, Miller WG, Konkel ME. Differentiation of Campylobacter coli, Campylobacter jejuni, Campylobacter lari, and Campylobacter upsaliensis by a multiplex PCR developed from the nucleotide sequence of the lipid A gene lpxA. J Clin Microbiol. 2004;42(12] has been updated. OK?):5549–5557. doi:10.1128/JCM.42.12.5549-5557.2004.
  • Dasti JI, Tareen AM, Lugert R, Zautner AE, Gross U. Campylobacter jejuni: a brief overview on pathogenicity-associated factors and disease-mediating mechanisms. Int J Med Microbiol. 2010;300(4):205–211. doi:10.1016/j.ijmm.2009.07.002.
  • Konkel ME, Garvis SG, Tipton SL, Anderson, Jr DE Jr., Cieplak, Jr W Jr. Identification and molecular cloning of a gene encoding a fibronectin-binding protein (CadF) from Campylobacter jejuni. Mol Microbiol. 1997;24(5):953–963. doi:10.1046/j.1365-2958.1997.4031771.x.
  • Pei Z, Blaser MJ. PEB1, the major cell-binding factor of Campylobacter jejuni, is a homolog of the binding component in gram-negative nutrient transport systems.. J Biol Chem. 1993;268:18717–18725.
  • Jin S, Joe A, Lynett J, Hani EK, Sherman P, Chan VL. JlpA, a novel surface-exposed lipoprotein specific to Campylobacter jejuni, mediates adherence to host epithelial cells. Mol Microbiol. 2001;39(5):1225–1236. doi:10.1111/j.1365-2958.2001.02294.x.
  • Cortes-Bratti X, Karlsson C, Lagergard T, Thelestam M, Frisan T. The Haemophilus ducreyi Cytolethal Distending Toxin Induces Cell Cycle Arrest and Apoptosis via the DNA Damage Checkpoint Pathways. J Biol Chem. 2001;276(7):5296–5302. doi:10.1074/jbc.M008527200.
  • Korlath JA, Osterholm MT, Judy LA, Forfang JC, Robinson RA. A point-source outbreak of campylobacteriosis associated with consumption of raw milk. J Infect Dis. 1985;152(3):592–596. doi:10.1093/infdis/152.3.592.
  • Baqar S, Applebee LA, Bourgeois AL. Immunogenicity and protective efficacy of a prototype Campylobacter killed whole-cell vaccine in mice.. Infect Immun. 1995;63(9):3731–3735. doi:10.1128/IAI.63.9.3731-3735.1995.
  • Baqar S, Bourgeois AL, Schultheiss PJ, Walker RI, Rollins DM, Haberberger RL, Pavlovskis OR. Safety and immunogenicity of a prototype oral whole-cell killed Campylobacter vaccine administered with a mucosal adjuvant in non-human primates. Vaccine. 1995;13(1):22–28. doi:10.1016/0264-410X(95)80006-Y.
  • Tribble DR, Baqar S, Thompson SA. Development of a Human Vaccine.. In: Nachamkin I, Szymanski CM, Blaser MJ editors. Campylobacter. 3rd. Washington (DC): American Society of Microbiology; 2008. p. 429–444.
  • Lee LH, Burg E 3rd, Baqar S, Bourgeois AL, Burr DH, Ewing CP, Trust TJ, Guerry P, Orndorff PE. Evaluation of a truncated recombinant flagellin subunit vaccine against Campylobacter jejuni. Infect Immun. 1999;67(11):5799–5805. doi:10.1128/IAI.67.11.5799-5805.1999.
  • Monteiro MA, Baqar S, Hall ER, Chen YH, Porter CK, Bentzel DE, Applebee L, Guerry P. Capsule polysaccharide conjugate vaccine against diarrheal disease caused by Campylobacter jejuni. Infect Immun. 2009;77(3):1128–1136. doi:10.1128/IAI.01056-08.
  • Poly F, Noll AJ, Riddle MS, Porter CK. Update on Campylobacter vaccine development. Hum Vaccin Immunother. 2019;15(6):1389–1400. doi:10.1080/21645515.2018.1528410.
  • Brunette GW. CDC Yellow Book 2020: health Information for International Travel. NY: Oxford University Press; 2020.
  • Scallan E, Hoekstra RM, Angulo FJ, Tauxe RV, Widdowson MA, Roy SL, Jones JL, Griffin PM. Foodborne illness acquired in the United States-major pathogens. Emerg Infect Dis. 2011;17(1):7–15. doi:10.3201/eid1701.P11101.
  • Gordon MA, Banda HT, Gondwe M, Gordon SB, Boeree MJ, Walsh AL, Corkill JE, Hart CA, Gilks CF, Molyneux ME. Non-typhoidal Salmonella bacteraemia among HIV-infected Malawian adults: high mortality and frequent recrudescence. AIDS. 2002;16(12):1633–1641. doi:10.1097/00002030-200208160-00009.
  • MacLennan CA, Levine MM. Invasive nontyphoidal Salmonella disease in Africa: current status. Expert Rev Anti Infect Ther. 2013;11(5):443–446. doi:10.1586/eri.13.27.
  • Kingsley RA, Msefula CL, Thomson NR, Kariuki S, Holt KE, Gordon MA, Harris D, Clarke L, Whitehead S, Sangal V, et al. Epidemic multiple drug resistant Salmonella Typhimurium causing invasive disease in sub-Saharan Africa have a distinct genotype. Genome Res. 2009;19(12):2279–2287. doi:10.1101/gr.091017.109.
  • de Jong HK, Parry CM, van der Poll T, Wiersinga WJ. Host-pathogen interaction in invasive Salmonellosis. PLoS Pathog. 2012;8(10):e1002933. doi:10.1371/journal.ppat.1002933.
  • Hindle Z, Chatfield SN, Phillimore J, Bentley M, Johnson J, Cosgrove CA, Ghaem-Maghami M, Sexton A, Khan M, Brennan FR, et al. Characterization of Salmonella enterica derivatives harboring defined aroC and Salmonella pathogenicity island 2 type III secretion system (ssaV) mutations by immunization of healthy volunteers. Infect Immun. 2002;70(7):3457–3467. doi:10.1128/IAI.70.7.3457-3467.2002.
  • Chatfield SN, Fairweather N, Charles I, Pickard D, Levine M, Hone D, Posada M, Strugnell RA, Dougan G. Construction of a genetically defined Salmonella typhi Ty2 aroA, aroC mutant for the engineering of a candidate oral typhoid-tetanus vaccine. Vaccine. 1992;10(1):53–60. doi:10.1016/0264-410X(92)90420-O.
  • Tennant SM, Wang JY, Galen JE, Simon R, Pasetti MF, Gat O, Levine MM, Fang FC. Engineering and preclinical evaluation of attenuated nontyphoidal Salmonella strains serving as live oral vaccines and as reagent strains. Infect Immun. 2011;79(10):4175–4185. doi:10.1128/IAI.05278-11.
  • Simon R, Tennant SM, Wang JY, Schmidlein PJ, Lees A, Ernst RK, Pasetti MF, Galen JE, Levine MM. Salmonella enterica Serovar Enteritidis Core O Polysaccharide Conjugated to H:g,m Flagellin as a Candidate Vaccine for Protection against Invasive Infection with S.Enteritidis. Infect Immun. 2011;79(10):4240–4249. doi:10.1128/IAI.05484-11.
  • Martinez-Becerra FJ, Kumar P, Vishwakarma V, Kim JH, Arizmendi O, Middaugh CR, Picking WD, Picking WL, Payne SM. Characterization and Protective Efficacy of Type III Secretion Proteins as a Broadly Protective Subunit Vaccine against Salmonella enterica Serotypes. Infect Immun. 2018;86(3):e00473–17. doi:10.1128/IAI.00473-17.
  • Wu HJ, Wang AH, Jennings MP. Discovery of virulence factors of pathogenic bacteria. Curr Opin Chem Biol. 2008;12(1):93–101. doi:10.1016/j.cbpa.2008.01.023.
  • Chao CC, Chelius D, Zhang T, Mutumanje E, Ching WM. Insight into the virulence of Rickettsia prowazekii by proteomic analysis and comparison with an avirulent strain. Biochim Biophys Acta. 2007;1774(3):373–381. doi:10.1016/j.bbapap.2007.01.001.
  • Duan Q, Lee KH, Nandre RM, Garcia C, Chen J, Zhang W. MEFA (multiepitope fusion antigen)-Novel Technology for Structural Vaccinology, Proof from Computational and Empirical Immunogenicity Characterization of an Enterotoxigenic Escherichia coli (ETEC) Adhesin MEFA. J Vaccines Vaccin. 2017;8:367.
  • Ruan X, Sack DA, Zhang W. Genetic fusions of a CFA/I/II/IV MEFA (multiepitope fusion antigen) and a toxoid fusion of heat-stable toxin (STa) and heat-labile toxin (LT) of enterotoxigenic Escherichia coli (ETEC) retain broad anti-CFA and antitoxin antigenicity. PLoS One. 2015;10(3] has been updated. OK?):e0121623. doi:10.1371/journal.pone.0121623.
  • Nandre RM, Ruan X, Duan Q, Sack DA, Zhang W. Antibodies derived from an enterotoxigenic Escherichia coli (ETEC) adhesin tip MEFA (multiepitope fusion antigen) against adherence of nine ETEC adhesins: CFA/I, CS1, CS2, CS3, CS4, CS5, CS6, CS21 and EtpA. Vaccine. 2016;34(31):3620–3625. doi:10.1016/j.vaccine.2016.04.003.
  • Rausch D, Ruan X, Nandre R, Duan Q, Hashish E, Casey TA, Zhang W. Antibodies derived from a toxoid MEFA (multiepitope fusion antigen) show neutralizing activities against heat-labile toxin (LT), heat-stable toxins (STa, STb), and Shiga toxin 2e (Stx2e) of porcine enterotoxigenic Escherichia coli (ETEC). Vet Microbiol. 2017;202:79–89. doi:10.1016/j.vetmic.2016.02.002.
  • Nandre R, Ruan X, Lu T, Duan Q, Sack D, Zhang W. Enterotoxigenic Escherichia coli Adhesin-Toxoid Multiepitope Fusion Antigen CFA/I/II/IV-3xSTaN12S-mnLTG192G/L211A-Derived Antibodies Inhibit Adherence of Seven Adhesins, Neutralize Enterotoxicity of LT and STa Toxins, and Protect Piglets against Diarrhea. Infect Immun. 2018;86:e00550–17.
  • Seo H, Anvari S, Li S, Lu T, Sack DA, Zhang W Immunogenicity characterization and pre-clinical evaluation of a combo vaccine for Shigella and enterotoxigenic Escherichia coli. Presented at: Vaccines for Enteric Diseases; 2019 Oct 16-18; Lausanne, Switzerland.
  • Qadri F, Bhuiyan TR, Sack DA, Svennerholm AM. Immune responses and protection in children in developing countries induced by oral vaccines. Vaccine. 2013;31(3):452–460. doi:10.1016/j.vaccine.2012.11.012.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.