1,473
Views
50
CrossRef citations to date
0
Altmetric
Commentary

Glycoconjugate vaccine strategies for protection against invasive Salmonella infections

&
Pages 494-498 | Received 15 Dec 2011, Accepted 23 Dec 2011, Published online: 28 Feb 2012

Abstract

Salmonella enterica serovars Typhi and Paratyphi A and B and certain non-typhoidal Salmonella enterica (NTS) serovars are important causes of invasive Salmonella disease worldwide. NTS serovars Typhimurium and Enteritidis typically cause gastroenteritis in healthy children and adults in industrialized countries but in certain hosts (e.g., young infants, the elderly, immunocompromised individuals) they also cause invasive infections. These two serovars also cause invasive disease in infants and young children in sub-Saharan Africa. Whereas Salmonella surface polysaccharides are poor immunogens in animal models and do not generate immunologic memory, conjugation with carrier proteins overcomes these limitations. S. Typhi expresses a Vi polysaccharide capsule; Vi either alone or as a glycoconjugate protects humans from typhoid fever. In contrast, S. Paratyphi A and B and NTS (with rare exceptions) do not express capsular polysaccharides. Rather, their surface polysaccharides are the O polysaccharide (OPS) of lipopolysaccharide. In animal studies, immunization with Salmonella COPS (core polysaccharide-OPS) conjugated with carrier proteins generates functional immunity and protects against fatal Salmonella challenge. Conjugating to Salmonella proteins (flagellin, porins) may extend immune responses to another relevant target for antibody generation and enhance the glyconjugate’s efficacy.

Introduction

A relatively restricted number of the > 2500 serovars of Salmonella are associated with invasive disease such as bacteremia, septicemia and meningitis. Four fairly distinct clinico-epidemiologic patterns of invasive Salmonella disease are recognized and are caused by distinct serovars: enteric fever; metastatic purulent infections; invasive disease in high risk hosts in industrialized and developing countries; invasive disease in young children in sub-Saharan Africa.

Three human-host-restricted enteric fever serovars (also called “typhoidal” serovars), Salmonella enterica serovar Typhi (S. Typhi), S. Paratyphi A and S. Paratyphi B, cause enteric (typhoid or paratyphoid) fever, manifested by persisting fever, abdominal discomfort and headache. If not treated promptly with effective antibiotics, typhoid and paratyphoid fever may lead to complications and death. In the pre-antibiotic era the case fatality rate of typhoid fever was ~15%. In infants, S. Typhi and S. Paratyphi bacteremic infections may be either clinically mild (with the bacteremia clearing spontaneously)Citation1 or severe.Citation2 Two serovars, S. choleraesuis and S. Paratyphi C, cause metastatic purulent infections, an uncommon clinical form of invasive disease.Citation3-Citation5

In the USA and Europe, gastroenteritis due to NTS serovars, a common disease, may occasionally be accompanied by invasive bacteremic disease. Susceptible hosts for invasive NTS disease include infants < 3 mo of age,Citation6-Citation9 the elderly,Citation9 persons with hemoglobinopathies and those with immunocompromise (inadequately treated HIV infection, etc.).Citation9 The most common NTS serovars associated with invasive disease in the USA include S. Typhimurium, S. Enteritidis, S. Heidelberg, S. Dublin and S. Schwarzengrund.Citation9

Finally, it has also become recognized that NTS commonly cause invasive bacterial disease among children < 3 y of age in many regions of sub-Saharan Africa.Citation10-Citation16 Prior to the introduction of programmatic immunization with Hemophilus influenzae type b (Hib) or Streptococcus pneumoniae conjugate vaccines in countries in sub-Saharan Africa, invasive NTS disease was as common as invasive Hib or pneumococcal disease.Citation10-Citation16 Of these clinico-epidemiologic syndromes caused by different serovars, all represent a sufficiently large burden as to be considered as targets for control by vaccines (except for S. choleraesuis and S. Paratyphi C metastatic purulent infections, which are relatively rare). Whereas licensed vaccines are available to prevent typhoid fever, no specific licensed vaccines are available against S. Paratyphi A or B or NTS serovars.

Vi based conjugate vaccines for protection against S. Typhi infections

Capsular polysaccharides of Hib, S. pneumoniae and Neisseria meningitidis have been linked to carrier proteins as the basis of well tolerated, immunogenic and efficacious licensed conjugate vaccines, documenting that the conjugate vaccine strategy is reliable, robust and flexible for polysaccharide-encapsulated pathogens that invade via the bloodstream. S. Typhi expresses a capsular polysaccharide, Vi antigen, which mediates resistance to bactericidal killing and opsonophagocytic uptake by the alternative arm of the complement system.Citation17 Serum IgG anti-Vi is a correlate of protection in humans.Citation17-Citation19 Like most polysaccharides,Citation20 Vi is poorly immunogenic in infants and fails to induce immunologic memory.Citation21 However, conjugation of Vi to a carrier protein overcomes these limitations,Citation20,Citation21 as has been documented through clinical trials with a pioneering conjugate vaccine consisting of Vi conjugated to recombinant exoprotein A of Pseudomonas aeruginosa (Vi-rEPA) developed at the US. National Institute of Child Health and Human Development. Vi-rEPA was tested in clinical trials in a high typhoid incidence area in Vietnam where, following demonstration of safety and immunogenicity in older children and adults,Citation22-Citation25 it was evaluated for efficacy in a randomized, controlled phase 3 field trial in pre-school children.Citation23,Citation24 A high level of protection was observed over 46 mo of follow-up.Citation23,Citation24 Vi-rEPA is immunogenic in Vietnamese infants when administered concomitantly with other pediatric vaccines that are part of the Vietnamese Expanded Program on Immunization (EPI).Citation26 Several investigators proposed a minimal threshold protective level of serum IgG anti-Vi that can facilitate the clinical development of new Vi conjugates.Citation19,Citation23,Citation26 Carrier proteins utilized in Vi conjugates include diphtheria toxoid (DT),Citation27 tetanus toxoid (TT), and CRM197.Citation28 Phase 1 and 2 clinical trials with Vi-CRM197 have shown its’ safety and immunogenicity in adults and teenagers. Vi-CRM197 elicited comparable levels of antibody at 1/20th of the standard dose of unconjugated Vi polysaccharide vaccine.Citation29 One Vi-TT conjugate has been licensed in India but no peer review publications have presented the safety and immunogenicity data generated with this vaccine. The paucity of published data on this specific conjugate has led to some controversy in India.Citation30-Citation32

S. Paratyphi C and some clones of S. Dublin also express Vi capsular polysaccharide but no field data have documented the efficacy of Vi conjugate vaccines against these serovars. Some have raised the theoretical concern that widespread use of Vi-based parenteral vaccines exert immunologic pressure selecting for the emergence of Vi-negative strains of S. Typhi.Citation33,Citation34 Vi-negative strains are generally rare but one study using molecular diagnostics convincingly detected Vi-negative S. Typhi uncommonly in blood.Citation35

Salmonella O antigens and relevance for developing vaccines to prevent invasive NTS disease and paratyphoid fever

Since NTS and S. Paratyphi A and B do not express capsular polysaccharides, investigators have studied vaccines that contain the repeating polymer of O-polysaccharide (OPS) as the basis of eliciting antibody-based protection in a manner analogous to what Vi polysaccharide and Vi conjugates have been able to accomplish in preventing S. Typhi disease. The lipopolysaccharide (LPS) of Salmonella is comprised of lipid A (endotoxin) attached to a highly conserved core polysaccharide and a repeating OPS polymer. The overwhelmingly majority of invasive Salmonella isolates from humans fall into Salmonella Groups A, B, C or D. OPS of Salmonella groups A, B and D are similar in overall structure. They share a common trisaccharide backbone →2)-α-D-Manp-(1→4)-α-L-Rhap-(1→3)-α-D-Galp-(1→) (which serologically constitutes epitope 12). A dideoxy hexose saccharide linked α-(3→6) at the mannose of the repeating trisaccharideCitation36 results in an immunodominant epitope that confers Salmonella group identity. Thus, if the dideoxy hexose linked to the mannose is a paratose, this provides immunodominant epitope 2, specifying a Group A Salmonella. If the α-(3→6)-linked dideoxyhexose is an abequose, immunodominant epitope 4 specificity is conferred, indicative of Group B. If the α-(3→6)-linked dideoxyhexose is a tyvelose, immunodominant epitope 9 results, putting the isolate into Group D. The rhamnose in the backbone →2)-α-D-Manp-(1→4)-α-L-Rhap-(1→3)-α-D-Galp-(1→) trisaccharide repeat of S. Paratyphi A is also partially O-acetylated; however, there is no antigenic epitope recognized in association with this modification.Citation37,Citation38

In some Group B serovars such as S. Typhimurium, phage conversion modifies the galactose of the trisaccharide backbone epitope 12 so that it becomes α-(1→6) glucosylated and minor epitope 1 can be detected.Citation39 Some Group B serovars also express minor epitope 5, resulting from a chromosomal gene product that acetylates the 2-hydroxyl group of the abequose residue.Citation40,Citation41

OPS of Salmonella serogroups C are structurally and serologically distinct from Groups A, B and D.Citation36,Citation39 Salmonella isolates with OPS exhibiting immunodominant epitopes O:6,7 characterize Salmonella Group C1. Isolates lysogenized with phage 14, resulting in the antigen pattern O:6,7,14, used to be designated group C4 but are presently considered as members of Group C1. Salmonella isolates bearing immunodominant O:8 comprise Group C2, whether or not they also express epitope 6. In older typing regimens, isolates bearing O:6,8 were referred to as C2 to distinguish them from isolates bearing only O:8, which were designated C3.

The critical issues revolving around the use of OPS-based conjugate vaccines to prevent invasive NTS disease and paratyphoid fever include whether O antibodies to NTS and Paratyphi A and B serovars in humans can mediate protection, the biological activities of anti-LPS antibodies in humans and whether antibodies to an OPS-based vaccine made with purified OPS from one serovar cross-protect against other serovars within the same O serogroup, as would be expected.

Biological activity of anti-O antibodies

Although Salmonella are intracellular pathogens, they are vulnerable while extracellular when IgG and IgM directed against the surface polysaccharides of Salmonella can bind them leading to bacteriolysis or opsonophagocytosis. The importance of serum immunity is underscored by the increased virulence seen for Salmonella that can evade the alternative pathway of complement through alteration in the length and structure of their OPS and expression of the resistance to complement killing (rck) gene.Citation42-Citation45 Antibodies to Salmonella surface carbohydrates mediate opsonophagocytosis through Fc receptors on phagocytes that can kill by oxidative burst.Citation46 Activation of the antibody mediated complement pathway by IgM and IgG can also kill directly via formation of the C9 membrane attack complex; surface deposition of C3b also enhances opsonophagocytosis.Citation47,Citation48

Evidence that Salmonella OPS antibodies can protect animals and humans

Passively transferred IgG or IgM monoclonal antibodies specific for S. Typhimurium OPS protected mice against S. Typhimurium challenge.Citation49 A study to assess the protection related to specific epitopes within OPS suggests that antibodies to the immunodominant group-specific epitope constitute the primary protective species; IgG or IgM specific for epitope 4 protected to a greater extent than an IgG to epitope 12.Citation50 A monoclonal IgA directed against epitope 5 has also been shown to prevent mucosal infection with S. Typhimurium given to mice by oral challenge.Citation40,Citation51Polyclonal antibodies elicited by COPS conjugates in rabbits and mice also provide passive immunity against fatal NTS challenge in mice.Citation52,Citation53

While the protective efficacy of antibody against NTS OPS and COPS is well documented in animal studies, the functionality of anti-COPS in humans is less clear. Antibody to S. Typhimurium LPS from HIV positive individuals in Africa was shown to interfere with complement mediated bactericidal killing of a serum sensitive prototype African S. Typhimurium strain.Citation54 Anti-LPS IgG however does not interfere with opsonophagocytosis and oxidative burst in human neutrophils with either complement resistant or sensitive S. Typhimurium strains.Citation46 NTS isolates from the blood also frequently display marked resistance to complement mediated bactericidal killing.Citation55 Further work is needed to better define the role of anti-OPS in serum bactericidal and opsonophagocytic killing in immunity to invasive NTS infection in humans.

Salmonella COPS and OPS as vaccine antigens in humans and in animal models

Little is known regarding the immunogenicity of purified Salmonella COPS or OPS in humans administered parenterally as a polysaccharide vaccine. In the early 1960s, clinical studies assessed the clinical acceptability and immunogenicity of two LPS-based vaccines containing purified S. Typhi LPS. The efficacy of these parenteral vaccines was also examined in large-scale field trials that included killed whole cell S. Typhi vaccines, also administered parenterally.Citation56,Citation57 Whereas the parenteral killed whole cell vaccines conferred a moderate level of protection against typhoid fever, the unconjugated LPS vaccines provided little or no protection. Vi PS expressed by wild type S. Typhi may have interfered with the ability of anti-LPS antibodies to bind to LPS on the bacteria present in blood, perhaps explaining the poor efficacy of these early LPS-based vaccines. However, evidence from studies in mice also suggests that Salmonella COPS as an isolated polysaccharide is a poor immunogen.Citation37,Citation58,Citation59 In contrast, conjugation of Salmonella COPS to protein carriers results in vaccines that have been effective in generating anti-OPS in animal models.Citation37,Citation52,Citation53,Citation58,Citation59 NTS COPS conjugate vaccines have also demonstrated protection against mortality in the mouse model of lethal Salmonella infection. In one study, conjugation of S. Typhimurium COPS to the homologous strain porin proteins elicited increased levels of anti-COPS IgG, and demonstrated protection against an LD100 challenge with virulent S. Typhimurium.Citation53 Antibodies elicited by this conjugate in mice, as well as an OPS conjugate with bovine serum albumin (BSA) in rabbits, exhibited functional opsonophagocytic antibody that could transfer protection by passive immunization.Citation52,Citation53,Citation60 Similar results were seen following immunization of mice with a conjugate of S. Typhimurium COPS with TT.Citation59 A S. Paratyphi A COPS-TT conjugate also increased the immunogenicity of COPS in mice, and elicited antibodies demonstrating complement-mediated bactericidal killing.Citation37 S. Paratyphi A COPS-TT was safe and immunogenic in humans in phase 1 and 2 clinical trials; serum from the vaccinated humans displayed functional bactericidal activity.Citation61 A conjugate vaccine consisting of S. Enteritidis COPS linked to the homologous serovar flagellin FliC elicited LPS-specific IgG and protected mice against otherwise lethal challenge with virulent S. Enteritidis.Citation58

Selecting the carrier protein for Salmonella OPS-based conjugates

Salmonella OPS glyconjugates that use homologous pathogen protective antigens (e.g., flagellins, porins) as carrier proteins can enhance protection by concomitantly eliciting immune responses to a second relevant antigen, thereby providing greater protective efficacy than conjugates constructed with heterologous carrier proteins (e.g., tetanus toxoid, CRM197). Mice immunized with conjugates of S. Typhimurium COPS with homologous strain porins displayed lower mortality to fatal Salmonella challenge than mice immunized with porins alone.Citation53S. Enteritidis COPS-flagellin conjugates that elicited high titers of antibodies to both COPS and flagellin exhibited higher efficacy than conjugate antigens that elicited high antibodies to only one component.Citation58 There is interest to test in humans the hypothesis that antibodies directed toward a carrier protein derived from Salmonella may have an additive or synergistic effect on immunogenicity (and protection). Salmonella flagellins as vaccine antigens are particularly attractive as it is anticipated that they can be economically manufactured at large scaleCitation58,Citation62,Citation63 and are amenable to several biochemical conjugation strategies.

Multivalent Salmonella glycoconjugate vaccine formulations

If COPS-flagellin or COPS-porin conjugates prove to be well tolerated, immunogenic and efficacious against pilot serovars and if antibodies to the immunodominant O serogroup antigens demonstrate cross protection against other clinically important serovars within the same serogroup, one can envision a global multivalent conjugate vaccine. With ca. 5–6 conjugates, such a multivalent conjugate vaccine could offer protection against virtually all the serovars that presently cause invasive disease globally. Thus, for example, a multivalent vaccine formulation consisting of COPS conjugates from S. Paratyphi A (group A), S. Typhimurium (group B), S. Enteritidis (group D), and S. Choleraesuis (Group C), along with a Vi-conjugate, would constitute a broad-based vaccine covering almost all invasive Salmonella disease.

Acknowledgments

Funding: MML and RS received support from Middle Atlantic RCE Program, NIAID/NIH 2 U54 AI057168 (MML PI).

Disclosure of Potential Conflicts of Interest

The authors declare no conflict of interest with regard to this manuscript.

References

  • Ferreccio C, Levine MM, Manterola A, Rodriguez G, Rivara I, Prenzel I, et al. Benign bacteremia caused by Salmonella typhi and paratyphi in children younger than 2 years. J Pediatr 1984; 104:899 - 901; http://dx.doi.org/10.1016/S0022-3476(84)80492-8; PMID: 6427437
  • Owais A, Sultana S, Zaman U, Rizvi A, Zaidi AK. Incidence of typhoid bacteremia in infants and young children in southern coastal Pakistan. Pediatr Infect Dis J 2010; 29:1035 - 9; PMID: 21046701
  • Chiu CH, Chuang CH, Chiu S, Su LH, Lin TY. Salmonella enterica serotype Choleraesuis infections in pediatric patients. Pediatrics 2006; 117:e1193 - 6; http://dx.doi.org/10.1542/peds.2005-2251; PMID: 16717121
  • Chiu CH, Wu TL, Su LH, Liu JW, Chu C. Fluoroquinolone resistance in Salmonella enterica serotype Choleraesuis, Taiwan, 2000-2003. Emerg Infect Dis 2004; 10:1674 - 6; PMID: 15498176
  • Chiu S, Chiu CH, Lin TY. Salmonella enterica serotype Choleraesuis infection in a medical center in northern Taiwan. J Microbiol Immunol Infect 2004; 37:99 - 102; PMID: 15181491
  • Kazemi M, Gumpert G, Marks MI. Clinical spectrum and carrier state of nontyphoidal salmonella infections in infants and children. Can Med Assoc J 1974; 110:1253 - 7; PMID: 4857958
  • Nelson SJ, Granoff D. Salmonella gastroenteritis in the first three months of life. A review of management and complications. Clin Pediatr (Phila) 1982; 21:709 - 12; http://dx.doi.org/10.1177/000992288202101201; PMID: 7140121
  • Kuppermann N. Occult bacteremia in young febrile children. Pediatr Clin North Am 1999; 46:1073 - 109; http://dx.doi.org/10.1016/S0031-3955(05)70176-0; PMID: 10629675
  • Vugia DJ, Samuel M, Farley MM, Marcus R, Shiferaw B, Shallow S, et al, Emerging Infections Program FoodNet Working Group. Invasive Salmonella infections in the United States, FoodNet, 1996-1999: incidence, serotype distribution, and outcome. Clin Infect Dis 2004; 38:Suppl 3 S149 - 56; http://dx.doi.org/10.1086/381581; PMID: 15095184
  • Berkley JA, Lowe BS, Mwangi I, Williams T, Bauni E, Mwarumba S, et al. Bacteremia among children admitted to a rural hospital in Kenya. N Engl J Med 2005; 352:39 - 47; http://dx.doi.org/10.1056/NEJMoa040275; PMID: 15635111
  • Kariuki S, Revathi G, Kariuki N, Kiiru J, Mwituria J, Muyodi J, et al. Invasive multidrug-resistant non-typhoidal Salmonella infections in Africa: zoonotic or anthroponotic transmission?. J Med Microbiol 2006; 55:585 - 91; http://dx.doi.org/10.1099/jmm.0.46375-0; PMID: 16585646
  • Kariuki S, Revathi G, Kariuki N, Kiiru J, Mwituria J, Hart CA. Characterisation of community acquired non-typhoidal Salmonella from bacteraemia and diarrhoeal infections in children admitted to hospital in Nairobi, Kenya. BMC Microbiol 2006; 6:101; http://dx.doi.org/10.1186/1471-2180-6-101; PMID: 17173674
  • Mandomando I, Macete E, Sigaúque B, Morais L, Quintó L, Sacarlal J, et al. Invasive non-typhoidal Salmonella in Mozambican children. Trop Med Int Health 2009; 14:1467 - 74; http://dx.doi.org/10.1111/j.1365-3156.2009.02399.x; PMID: 19793081
  • Walsh AL, Phiri AJ, Graham SM, Molyneux EM, Molyneux ME. Bacteremia in febrile Malawian children: clinical and microbiologic features. Pediatr Infect Dis J 2000; 19:312 - 8; http://dx.doi.org/10.1097/00006454-200004000-00010; PMID: 10783021
  • Enwere G, Biney E, Cheung YB, Zaman SM, Okoko B, Oluwalana C, et al. Epidemiologic and clinical characteristics of community-acquired invasive bacterial infections in children aged 2-29 months in The Gambia. Pediatr Infect Dis J 2006; 25:700 - 5; http://dx.doi.org/10.1097/01.inf.0000226839.30925.a5; PMID: 16874169
  • Tennant SM, Diallo S, Levy H, Livio S, Sow SO, Tapia M, et al. Identification by PCR of non-typhoidal Salmonella enterica serovars associated with invasive infections among febrile patients in Mali. PLoS Negl Trop Dis 2010; 4:e621; http://dx.doi.org/10.1371/journal.pntd.0000621; PMID: 20231882
  • Wilson RP, Winter SE, Spees AM, Winter MG, Nishimori JH, Sanchez JF, et al. The Vi capsular polysaccharide prevents complement receptor 3-mediated clearance of Salmonella enterica serotype Typhi. Infect Immun 2011; 79:830 - 7; http://dx.doi.org/10.1128/IAI.00961-10; PMID: 21098104
  • Robbins JD, Robbins JB. Reexamination of the protective role of the capsular polysaccharide (Vi antigen) of Salmonella typhi. J Infect Dis 1984; 150:436 - 49; http://dx.doi.org/10.1093/infdis/150.3.436; PMID: 6207249
  • Klugman KP, Koornhof HJ, Robbins JB, Le Cam NN. Immunogenicity, efficacy and serological correlate of protection of Salmonella typhi Vi capsular polysaccharide vaccine three years after immunization. Vaccine 1996; 14:435 - 8; http://dx.doi.org/10.1016/0264-410X(95)00186-5; PMID: 8735556
  • Pollard AJ, Perrett KP, Beverley PC. Maintaining protection against invasive bacteria with protein-polysaccharide conjugate vaccines. Nat Rev Immunol 2009; 9:213 - 20; http://dx.doi.org/10.1038/nri2494; PMID: 19214194
  • Szu SC, Taylor DN, Trofa AC, Clements JD, Shiloach J, Sadoff JC, et al. Laboratory and preliminary clinical characterization of Vi capsular polysaccharide-protein conjugate vaccines. Infect Immun 1994; 62:4440 - 4; PMID: 7927707
  • Kossaczka Z, Lin FY, Ho VA, Thuy NT, Van Bay P, Thanh TC, et al. Safety and immunogenicity of Vi conjugate vaccines for typhoid fever in adults, teenagers, and 2- to 4-year-old children in Vietnam. Infect Immun 1999; 67:5806 - 10; PMID: 10531232
  • Lin FY, Ho VA, Khiem HB, Trach DD, Bay PV, Thanh TC, et al. The efficacy of a Salmonella typhi Vi conjugate vaccine in two-to-five-year-old children. N Engl J Med 2001; 344:1263 - 9; http://dx.doi.org/10.1056/NEJM200104263441701; PMID: 11320385
  • Mai NL, Phan VB, Vo AH, Tran CT, Lin FY, Bryla DA, et al. Persistent efficacy of Vi conjugate vaccine against typhoid fever in young children. N Engl J Med 2003; 349:1390 - 1; http://dx.doi.org/10.1056/NEJM200310023491423; PMID: 14523155
  • Canh DG, Lin FY, Thiem VD, Trach DD, Trong ND, Mao ND, et al. Effect of dosage on immunogenicity of a Vi conjugate vaccine injected twice into 2- to 5-year-old Vietnamese children. Infect Immun 2004; 72:6586 - 8; http://dx.doi.org/10.1128/IAI.72.11.6586-6588.2004; PMID: 15501790
  • Thiem VD, Lin FY, Canh G, Son NH, Anh DD, Mao ND, et al. The Vi conjugate typhoid vaccine is safe, elicits protective levels of IgG anti-Vi, and is compatible with routine infant vaccines. Clin Vaccine Immunol 2011; 18:730 - 5; http://dx.doi.org/10.1128/CVI.00532-10; PMID: 21411598
  • Cui C, Carbis R, An SJ, Jang H, Czerkinsky C, Szu SC, et al. Physical and chemical characterization and immunologic properties of Salmonella enterica serovar typhi capsular polysaccharide-diphtheria toxoid conjugates. Clin Vaccine Immunol 2010; 17:73 - 9; http://dx.doi.org/10.1128/CVI.00266-09; PMID: 19889941
  • Micoli F, Rondini S, Pisoni I, Proietti D, Berti F, Costantino P, et al. Vi-CRM 197 as a new conjugate vaccine against Salmonella Typhi. Vaccine 2011; 29:712 - 20; http://dx.doi.org/10.1016/j.vaccine.2010.11.022; PMID: 21115057
  • van Damme P, Kafeja F, Anemona A, Basile V, Hilbert AK, De Coster I, et al. Safety, immunogenicity and dose ranging of a new Vi-CRM₁₉₇ conjugate vaccine against typhoid fever: randomized clinical testing in healthy adults. PLoS One 2011; 6:e25398; http://dx.doi.org/10.1371/journal.pone.0025398; PMID: 21980445
  • Shah N. Indian Vi conjugate typhoid vaccine: misleading claims. Indian Pediatr 2010; 47:447; http://dx.doi.org/10.1007/s13312-010-0068-4; PMID: 20519792
  • Shah NK. Indian conjugate Vi typhoid vaccine: do we have enough evidence?. Indian Pediatr 2009; 46:181 - 2; PMID: 19242041
  • Mathew JL. Conjugate typhoid vaccine(s) in the Indian context. Indian Pediatr 2009; 46:182 - 4; PMID: 19242042
  • Arya SC. Salmonella typhi Vi antigen-negative isolates in India and prophylactic typhoid immunization. Natl Med J India 2000; 13:220; PMID: 11002694
  • Arya SC. Efficacy of Vi polysaccharide vaccine against Salmonella typhi. Vaccine 1999; 17:1015 - 6; PMID: 10195609
  • Baker S, Sarwar Y, Aziz H, Haque A, Ali A, Dougan G, et al. Detection of Vi-negative Salmonella enterica serovar typhi in the peripheral blood of patients with typhoid fever in the Faisalabad region of Pakistan. J Clin Microbiol 2005; 43:4418 - 25; http://dx.doi.org/10.1128/JCM.43.9.4418-4425.2005; PMID: 16145086
  • Lindberg AA, Le Minor L. Serology of Salmonella. In: Bergan T, ed. Methods in Microbiology: Academic Press, 1984:1-141.
  • Konadu E, Shiloach J, Bryla DA, Robbins JB, Szu SC. Synthesis, characterization, and immunological properties in mice of conjugates composed of detoxified lipopolysaccharide of Salmonella paratyphi A bound to tetanus toxoid with emphasis on the role of O acetyls. Infect Immun 1996; 64:2709 - 15; PMID: 8698499
  • Hellerqvist CG, Lindberg B, Samuelsson K, Lindberg AA. Structural studies on the O-specific side-chains of the cell-wall lipopolysaccharide from Salmonella parathyphi A var. durazzo. Acta Chem Scand 1971; 25:955 - 61; http://dx.doi.org/10.3891/acta.chem.scand.25-0955; PMID: 5117491
  • Grimont PAD, Weill FX. Antigenic formulae of the Salmonella serovars. WHO Collaborating Centre for Reference and Research on Salmonella 2007:1-166.
  • Slauch JM, Mahan MJ, Michetti P, Neutra MR, Mekalanos JJ. Acetylation (O-factor 5) affects the structural and immunological properties of Salmonella typhimurium lipopolysaccharide O antigen. Infect Immun 1995; 63:437 - 41; PMID: 7529745
  • Ronholm J, Zhang Z, Cao X, Lin M. Monoclonal antibodies to lipopolysaccharide antigens of Salmonella enterica serotype Typhimurium DT104. Hybridoma (Larchmt) 2011; 30:43 - 52; http://dx.doi.org/10.1089/hyb.2010.0066; PMID: 21466285
  • Heffernan EJ, Harwood J, Fierer J, Guiney D. The Salmonella typhimurium virulence plasmid complement resistance gene rck is homologous to a family of virulence-related outer membrane protein genes, including pagC and ail.. J Bacteriol 1992; 174:84 - 91; PMID: 1729227
  • Grossman N, Schmetz MA, Foulds J, Klima EN, Jimenez-Lucho VE, Leive LL, et al. Lipopolysaccharide size and distribution determine serum resistance in Salmonella montevideo. J Bacteriol 1987; 169:856 - 63; PMID: 2433267
  • Liang-Takasaki CJ, Grossman N, Leive L. Salmonellae activate complement differentially via the alternative pathway depending on the structure of their lipopolysaccharide O-antigen. J Immunol 1983; 130:1867 - 70; PMID: 6187823
  • Liang-Takasaki CJ, Saxén H, Mäkelä PH, Leive L. Complement activation by polysaccharide of lipopolysaccharide: an important virulence determinant of salmonellae.. Infect Immun 1983; 41:563 - 9; PMID: 6347890
  • Gondwe EN, Molyneux ME, Goodall M, Graham SM, Mastroeni P, Drayson MT, et al. Importance of antibody and complement for oxidative burst and killing of invasive nontyphoidal Salmonella by blood cells in Africans. Proc Natl Acad Sci U S A 2010; 107:3070 - 5; http://dx.doi.org/10.1073/pnas.0910497107; PMID: 20133627
  • MacLennan CA, Gondwe EN, Msefula CL, Kingsley RA, Thomson NR, White SA, et al. The neglected role of antibody in protection against bacteremia caused by nontyphoidal strains of Salmonella in African children. J Clin Invest 2008; 118:1553 - 62; http://dx.doi.org/10.1172/JCI33998; PMID: 18357343
  • Casadevall A, Pirofski LA. A reappraisal of humoral immunity based on mechanisms of antibody-mediated protection against intracellular pathogens. Adv Immunol 2006; 91:1 - 44; http://dx.doi.org/10.1016/S0065-2776(06)91001-3; PMID: 16938537
  • Colwell DE, Michalek SM, Briles DE, Jirillo E, McGhee JR. Monoclonal antibodies to Salmonella lipopolysaccharide: anti-O-polysaccharide antibodies protect C3H mice against challenge with virulent Salmonella typhimurium. J Immunol 1984; 133:950 - 7; PMID: 6203984
  • Carlin NI, Svenson SB, Lindberg AA. Role of monoclonal O-antigen antibody epitope specificity and isotype in protection against experimental mouse typhoid. Microb Pathog 1987; 2:171 - 83; http://dx.doi.org/10.1016/0882-4010(87)90019-2; PMID: 2467161
  • Michetti P, Mahan MJ, Slauch JM, Mekalanos JJ, Neutra MR. Monoclonal secretory immunoglobulin A protects mice against oral challenge with the invasive pathogen Salmonella typhimurium. Infect Immun 1992; 60:1786 - 92; PMID: 1373399
  • Svenson SB, Lindberg AA. Artificial Salmonella vaccines: Salmonella typhimurium O-antigen-specific oligosaccharide-protein conjugates elicit protective antibodies in rabbits and mice. Infect Immun 1981; 32:490 - 6; PMID: 6166555
  • Svenson SB, Nurminen M, Lindberg AA. Artificial Salmonella vaccines: O-antigenic oligosaccharide-protein conjugates induce protection against infection with Salmonella typhimurium. Infect Immun 1979; 25:863 - 72; PMID: 387597
  • MacLennan CA, Gilchrist JJ, Gordon MA, Cunningham AF, Cobbold M, Goodall M, et al. Dysregulated humoral immunity to nontyphoidal Salmonella in HIV-infected African adults. Science 2010; 328:508 - 12; http://dx.doi.org/10.1126/science.1180346; PMID: 20413503
  • Roantree RJ, Rantz LA. A Study of the Relationship of the Normal Bactericidal Activity of Human Serum to Bacterial Infection. J Clin Invest 1960; 39:72 - 81; http://dx.doi.org/10.1172/JCI104029; PMID: 16695824
  • Polish Typhoid Committee. Controlled field trials and laboratory studies on the effectiveness of typhoid vaccines in Poland, 1961-64. Bull World Health Organ 1966; 34:211 - 22; PMID: 5296128
  • Hejfec LBSL, Salmin LV, Lejtman MZ, Kuz’minova ML, Vasil’eva AV, Levina LA, et al. A controlled field trial and laboratory study of five typhoid vaccines in the USSR. Bull World Health Organ 1966; 34:321 - 39; PMID: 5296393
  • Simon R, Tennant SM, Wang JY, Schmidlein PJ, Lees A, Ernst RK, et al. 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; 79:4240 - 9; http://dx.doi.org/10.1128/IAI.05484-11; PMID: 21807909
  • Watson DC, Robbins JB, Szu SC. Protection of mice against Salmonella typhimurium with an O-specific polysaccharide-protein conjugate vaccine. Infect Immun 1992; 60:4679 - 86; PMID: 1383154
  • Jörbeck HJ, Svenson SB, Lindberg AA. Artificial Salmonella vaccines: Salmonella typhimurium O-antigen-specific oligosaccharide-protein conjugates elicit opsonizing antibodies that enhance phagocytosis. Infect Immun 1981; 32:497 - 502; PMID: 7019072
  • Konadu EY, Lin FY, Hó VA, Thuy NT, Van Bay P, Thanh TC, et al. Phase 1 and phase 2 studies of Salmonella enterica serovar paratyphi A O-specific polysaccharide-tetanus toxoid conjugates in adults, teenagers, and 2- to 4-year-old children in Vietnam. Infect Immun 2000; 68:1529 - 34; http://dx.doi.org/10.1128/IAI.68.3.1529-1534.2000; PMID: 10678970
  • Tennant SM, Wang JY, Galen JE, Simon R, Pasetti MF, Gat O, et al. Engineering and preclinical evaluation of attenuated nontyphoidal Salmonella strains serving as live oral vaccines and as reagent strains. Infect Immun 2011; 79:4175 - 85; http://dx.doi.org/10.1128/IAI.05278-11; PMID: 21807911
  • Song L, Nakaar V, Kavita U, Price A, Huleatt J, Tang J, et al. Efficacious recombinant influenza vaccines produced by high yield bacterial expression: a solution to global pandemic and seasonal needs. PLoS One 2008; 3:e2257; http://dx.doi.org/10.1371/journal.pone.0002257; PMID: 18493310

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.