Salmonella enterica serovar Typhi (typhoid) vaccines

55
Immunization, Vaccines and Biologicals The Immunological Basis for Immunization Series Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines

Transcript of Salmonella enterica serovar Typhi (typhoid) vaccines

Page 1: Salmonella enterica serovar Typhi (typhoid) vaccines

Immunization, Vaccines and Biologicals

The Immunological Basisfor Immunization Series

Module 20:

Salmonella enterica serovar Typhi (typhoid) vaccines

Page 2: Salmonella enterica serovar Typhi (typhoid) vaccines

Immunization, Vaccines and Biologicals

The Immunological Basisfor Immunization Series

Module 20:

Salmonella enterica serovar Typhi (typhoid) vaccines

Page 3: Salmonella enterica serovar Typhi (typhoid) vaccines

WHO Library Cataloguing-in-Publication Data

The immunological basis for immunization series: module 20: salmonella enterica serovar Typhi (typhoid) vaccines.

(Immunological basis for immunization series ; module 20)

1.Salmonella typhi - microbiology. 2.Typhoid fever - immunology. 3.Typhoid-paratyphoid vaccines - therapeutic use. 4.Vaccination. I.World Health Organization. II.Series.

ISBN 978 92 4 150261 0 (NLM classification: WC 270)

© World Health Organization 2011All rights reserved. Publications of the World Health Organization are available on the WHO web site (www.who.int) or can be purchased from WHO Press, World Health Organization, 20 Avenue Appia, 1211 Geneva 27, Switzerland (tel.: +41 22 791 3264; fax: +41 22 791 4857; e-mail: [email protected]). Requests for permission to reproduce or translate WHO publications – whether for sale or for noncommercial distribution – should be addressed to WHO Press through the WHO web site (http://www.who.int/about/licensing/copyright_form/en/index.html).

The designations employed and the presentation of the material in this publication do not imply the expression of any opinion whatsoever on the part of the World Health Organization concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted lines on maps represent approximate border lines for which there may not yet be full agreement.

The mention of specific companies or of certain manufacturers’ products does not imply that they are endorsed or recommended by the World Health Organization in preference to others of a similar nature that are not mentioned. Errors and omissions excepted, the names of proprietary products are distinguished by initial capital letters.

All reasonable precautions have been taken by the World Health Organization to verify the information contained in this publication. However, the published material is being distributed without warranty of any kind, either expressed or implied. The responsibility for the interpretation and use of the material lies with the reader. In no event shall the World Health Organization be liable for damages arising from its use.

Page 4: Salmonella enterica serovar Typhi (typhoid) vaccines

ii

The Department of Immunization, Vaccines and Biologicals thanks the donors whose unspecified financial support

has made the production of this document possible.

This module was produced for Immunization, Vaccines and Biologicals, WHO, by:

Professor Myron M Levine with inputs from Professor Marcelo B Sztein and Dr Marcela F Pasetti.

Printed in October 2011

Copies of this publication as well as additional materials on immunization, vaccines and biological may be requested from:

World Health Organization Department of Immunization, Vaccines and Biologicals

CH-1211 Geneva 27, Switzerland • Fax: + 41 22 791 4227 • Email: [email protected]

© World Health Organization 2010

All rights reserved. Publications of the World Health Organization can be obtained from WHO Press, World Health Organization, 20 Avenue Appia, 1211 Geneva 27, Switzerland (tel: +41 22 791 3264; fax: +41 22 791 4857; email: [email protected]). Requests for permission to reproduce or translate WHO publications – whether for sale or for noncommercial distribution – should be addressed to WHO Press, at the above address (fax: +41 22 791 4806; email: [email protected]).

The designations employed and the presentation of the material in this publication do not imply the expression of any opinion whatsoever on the part of the World Health Organization concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted lines on maps represent approximate border lines for which there may not yet be full agreement.

The mention of specific companies or of certain manufacturers’ products does not imply that they are endorsed or recommended by the World Health Organization in preference to others of a similar nature that are not mentioned. Errors and omissions excepted, the names of proprietary products are distinguished by initial capital letters.

All reasonable precautions have been taken by the World Health Organization to verify the information contained in this publication. However, the published material is being distributed without warranty of any kind, either expressed or implied. The responsibility for the interpretation and use of the material lies with the reader. In no event shall the World Health Organization be liable for damages arising from its use.

The named authors alone are responsible for the views expressed in this publication.

Printed by the WHO Document Production Services, Geneva, Switzerland

Page 5: Salmonella enterica serovar Typhi (typhoid) vaccines

iii

Contents

Abbreviations and acronyms .............................................................................................vPreface .............................................................................................................................. vii

1. The organism and the disease ...............................................................................11.1 Introduction .................................................................................................11.2 Bacteriology ..................................................................................................21.3 Epidemiology................................................................................................31.4 Epidemiology of other enteric fever agents: Salmonella Paratyphi A, B

and C ............................................................................................................6

2. The pathogenesis and response to infection .......................................................72.1 Pathogenesis of Salmonella Typhi and Salmonella Paratyphi disease .....72.2 Clinical and epidemiologic significance of Vi-negative S. Typhi .............92.3 Immune response to wild type Salmonella Typhi ......................................9

3. Typhoid vaccines ..................................................................................................113.1 Current WHO guidelines for typhoid vaccine use ..................................113.2 Licensed typhoid vaccines .........................................................................11

4. Immune responses to currently licensed typhoid vaccines ............................144.1 Ty21a live oral vaccine ..............................................................................144.2 Purified Vi polysaccharide .........................................................................15

5. Vaccine efficacy .....................................................................................................175.1 Ty21a ...........................................................................................................175.2 Vi vaccine ..................................................................................................21

6. Vaccine formulation issues ..................................................................................236.1 Ty21a ...........................................................................................................236.2 Vi .................................................................................................................23

7. Vaccine schedule issues ........................................................................................247.1 Ty21a ..........................................................................................................247.2 Vi ................................................................................................................25

8. Vaccine effectiveness .............................................................................................278.1 Ty21a ...........................................................................................................278.2 Vi ................................................................................................................27

9. Future prospects ...................................................................................................289.1 New generation vaccines under development .........................................289.2 Immune response to the new generation typhoid vaccines ....................29

References ........................................................................................................................31

Page 6: Salmonella enterica serovar Typhi (typhoid) vaccines

iv

Page 7: Salmonella enterica serovar Typhi (typhoid) vaccines

v

Abbreviations and acronyms

ASC antibody secreting cell

CFU colony-forming unit

CI confidence interval

CMI cell-mediated immune response

CTL cytotoxic T-lymphocyte

DNA deoxyribonucleic acid

ELISA enzyme-linked immunosorbent assay

EPI Expanded Programme on Immunization

EU ELISA units

GM geometric mean

GMT geometric mean titre

Hib Haemophilus influenzae type b

IFN interferon

Ig immunoglobulin

IL interleukin

LPS lipopolysaccharide

MLST multilocus sequence typing

NTS non-typhoidal Salmonella

PBMC peripheral blood mononuclear cell

PCR polymerase chain reaction

PFGE pulsed field gel electrophoresis

rEPA recombinant exoprotein

TCM T central memory

TCR T-cell receptor

TEM T effector memory

Th T helper

TM memory T-cells

TNF tumour necrosis factor

Page 8: Salmonella enterica serovar Typhi (typhoid) vaccines

vi

Page 9: Salmonella enterica serovar Typhi (typhoid) vaccines

vii

Preface

This module is part of the series The Immunological Basis for Immunization, which was initially developed in 1993 as a set of eight modules focusing on the vaccines included in the Expanded Programme on Immunization (EPI)1. In addition to a general immunology module, each of the seven other modules covered one of the vaccines recommended as part of the EPI programme — diphtheria, measles, pertussis, polio, tetanus, tuberculosis and yellow fever. The modules have become some of the most widely used documents in the field of immunization.

With the development of the Global Immunization Vision and Strategy (GIVS) (2005–2015) (http://www.who.int/vaccines-documents/DocsPDF05/GIVS_Final_EN.pdf) and the expansion of immunization programmes in general, as well as the large accumulation of new knowledge since 1993, the decision was taken to update and extend this series.

The main purpose of the modules — which are published as separate disease/vaccine-specific modules — is to give immunization managers and vaccination professionals a brief and easily-understood overview of the scientific basis of vaccination, and also of the immunological basis for the World Health Organization (WHO) recommendations on vaccine use that, since 1998, have been published in the Vaccine Position Papers (http://www.who.int/immunization/documents/positionpapers_intro/en/index.html).

WHO would like to thank all the people who were involved in the development of the initial Immunological Basis for Immunization series, as well as those involved in its updating, and the development of new modules.

1 This programme was established in 1974 with the main aim of providing immunization for children in developing countries.

Page 10: Salmonella enterica serovar Typhi (typhoid) vaccines

viii

Page 11: Salmonella enterica serovar Typhi (typhoid) vaccines

1

1.1 Introduction

Typhoid fever is an acute generalized infection of the reticuloendothelial system, intestinal lymphoid tissue and gallbladder caused by Salmonella enterica serovar Typhi (Salmonella Typhi). This communicable disease is restricted to human hosts and humans (chronic carriers) constitute the reservoir of infection. A broad spectrum of clinical illness can follow ingestion of Salmonella Typhi, with more severe forms being characterized by persisting high fever, abdominal discomfort, malaise and headache. Prior to the availability of antibiotics, the disease ran its course over several weeks, resulting in a case-fatality rate of approximately 10%–20% (1,2). Paratyphoid fever is the clinically identical febrile infection caused by Salmonella Paratyphi A or B (or more rarely C) (3,4). Typhoid and paratyphoid fevers are often referred to collectively as enteric fevers. In most endemic areas, typhoid comprises approximately 75%– 80% of enteric fever. However, in some regions, particularly in Asia, Paratyphi A is starting to contribute a larger proportion of all enteric fevers (5,6). Paratyphoid fever caused by Salmonella Paratyphi A has emerged in recent years as an increasingly common severe infection among travellers from industrialized countries who visit developing countries, particularly in Asia (7–10).

Typhoid infection is acquired by ingestion of food or water vehicles contaminated by human excreta that contain Salmonella Typhi. Typhoid fever was endemic in virtually all countries of Europe and North and South America in the late nineteenth and early twentieth centuries. A single intervention — the treatment of water supplies (by chlorination, sand filtration, or other means) — which became increasingly popular in the late nineteenth and early twentieth century, broke the cycle of endemicity and caused the incidence of typhoid fever to plummet drastically, even though the prevalence of chronic carriers in the populations remained high for decades thereafter (11,12).

Typhoid fever is extremely uncommon in modern industrialized countries where populations have access to treated water supplies and to sanitation that removes human waste. By contrast, among populations in less-developed countries, that lack such amenities, typhoid fever is often endemic and, from the public-health perspective, typically constitutes the most important enteric disease problem of school-age children that is severe enough to result in visits to health-care providers and in hospitalizations (13). In some parts of Asia a notable proportion of cases also occur in preschool children (14,15). Besides children in less-developed countries, two other populations that are recognized to be at increased risk of developing typhoid fever are travellers (16,17,18) and clinical microbiologists (19,20).

1. The organism and the disease

Page 12: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines2

If strains are susceptible, typhoid fever and paratyphoid fever are readily treated by prompt administration of appropriate antibiotics (21). Since the late 1980s, strains of Salmonella Typhi exhibiting resistance to most of the antimicrobials that previously were clinically effective, have spread aggressively throughout the Indian subcontinent, South-East Asia and the Middle East (22–25). Whereas earlier epidemics of antibiotic resistant typhoid fever in Mexico (26) and Viet Nam (27) in the 1970s and Peru (28) in the 1980s were limited in time, and antibiotic-sensitive strains once again predominated within a few years, the current multiply-resistant Asian pandemic strains have persisted. The few antibiotics that remain effective against these multiply-resistant strains are expensive and not readily available in rural areas of developing countries. Thus, the arrival on the scene and dissemination of these multiply-resistant Salmonella Typhi strains constitutes an increasing public-health crisis in many developing countries.

1.2 Bacteriology

The taxonomy of Salmonella has changed repeatedly during the past 30 years (29). Originally, speciation was based on association with distinct clinical syndromes. With the Kauffman-White serologic classification, each distinct O:H serotype was given species status, a situation that became ponderous. The current official taxonomic designation is Salmonella enterica, sub-species enterica, serovar Typhi, to refer to the serovar that causes typhoid fever. Since it is now considered a serovar and not a separate species, Typhi is written non-italicized and with a capital T. Thus, Salmonella Typhi or S. Typhi are shortened forms that can be used. Serologically, Salmonella Typhi falls into Salmonella group D, based on its expressing O antigen 9, the immunodominant epitope diagnostic of Group D; it also bears minor O antigen 12. S. Typhi is motile and its peritrichous flagellae bear Phase 1 flagellar (H) antigen H:d. A few percent of isolates from Indonesia have flagella that bear other antigens including H:j (which results from an internal deletion within the the fliC gene encoding (d) (30) and z66 (which may be found in conjunction with H:d or H:j (31). Strains freshly isolated from patients possess on their surface an acidic polysaccharide capsule, the Vi (for virulence) antigen (32). Vi prevents O antibody from binding to the O antigen and inhibits the C3 component of complement from fixing to the surface of S. Typhi (33).

Salmonella Typhi exhibits a remarkable degree of homogeneity with respect to its biochemical and antigenic characteristics and genome (34,35). Exceptional are the few percent of isolates from Indonesia that bear Phase 1 flagellar antigen H:j rather than H:d. Application of multilocus sequence typing (MLST) to a worldwide collection of S. Typhi isolates spanning many decades revealed only four MLST patterns (35), two of which were predominant. Phage typing, using Vi phages, has historically offered reference laboratories a tool for differentiating strains from different geographic areas (36), while pulsed field gel electrophoresis (PFGE) allows greater discernment among S. Typhi isolates (37). However, it is the analysis of single nucleotide polymorphisms identified by high throughput methodology that is finally offering a molecular epidemiologic tool that can discern in detail among strains circulating in an endemic area (38).

Page 13: Salmonella enterica serovar Typhi (typhoid) vaccines

3

Analysis of the complete genome of two S. Typhi strains shows an impressive degree of homogeneity (39,40). The complete sequences of the ~ 4.8 million bp genomes of multiply antibiotic-resistant S. Typhi strain CT 18 (39) and of antibiotic-sensitive strain Ty2 (40) have been reported. Approximately 70% to 80% of the genome shares a backbone containing genes similar in sequence and order to those in Escherichia coli and other salmonellae. Along this backbone are intermittent clusters of S. enterica–specific and S. Typhi–specific genes (i.e. genes not found in S. Typhimurium or other S. enterica serovars) and more than 200 pseudogenes (including several that play a role in virulence in S. Typhimurium). In addition to the 218 150 bp plasmid encoding antibiotic resistance in CT 18, another 106 516 bp plasmid was found that shows common derivation with the pFra virulence plasmid of Yersinia pestis (39,41).

1.3 Epidemiology

Typhoid fever is transmitted by consumption of contaminated food or water vehicles. Wherever primitive or defective sanitary facilities exist and water supplies are subject to faecal contamination, the opportunity exists for enhanced transmission of S. Typhi. Prior to the availability of treated water and sewer systems, all strata of society were at risk. Endemic regions persist in the developing world primarily because segments of the population remain underserved by adequate water supply and sanitation services, and there is lack of detection and restriction of carriers. Provision of means to dispose of human faecal waste and to assure access to treated, bacteriologically monitored water, precludes widespread transmission and virtually eliminates typhoid fever from loci where it was previously endemic.

Transmission also occurs among clinical microbiologists in hospital diagnostic bacteriology laboratories, including in developed countries (19,20). The handling of specimens containing S. Typhi has caused numerous cases of typhoid fever.

The true incidence of typhoid fever in most regions of developing countries is not known, and attempts to provide global estimates of disease burden have had to make gross extrapolations from limited data (42). One global estimate suggested that there occur annually ~ 21 650 000 cases of typhoid fever (with ~ 216 500 deaths) and ~ 5 410 000 cases of paratyphoid fever. Several factors lead to an underestimation of the burden, while others lead to overestimation. Since clinical bacteriologic methods that isolate the causative organism are necessary to differentiate typhoid from paratyphoid fever and to confirm the clinical diagnosis of enteric fever, the paucity of bacteriological capability in much of the developing world, particularly in rural areas, introduces a high level of uncertainty about disease incidence. It can be argued that lack of clinical bacteriology infrastructure results in an underestimation of burden. On the other hand, several other febrile syndromes can be confused with enteric fever, particularly in children, so that if clinical bacteriology is not available for confirmation, many febrile illnesses caused by other bacterial, viral or rickettsial agents may be counted incorrectly as cases of typhoid or paratyphoid fever. Even if clinical bacteriology is available to isolate S. Typhi and S. Paratyphi from suspect cases, blood culture alone, the most commonly collected clinical specimen, is only 60%–70% sensitive in isolating the bacteria from otherwise confirmed cases (e.g., confirmed by a bone marrow or bile containing duodenal fluid culture) (43,44). Suppression of disease by self-medication with antibiotics or by health-practitioner treatment without obtaining cultures, further distorts the true situation.

Page 14: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines4

Nevertheless, despite the above caveats, a series of systematic surveys that have been carried out during the past two decades have provided invaluable information on the epidemiology of endemic typhoid (14,15,45,46,47). These systematic culture-based surveillance efforts were limited to geographic areas (mainly urban) and populations where prior evidence indicated that typhoid fever was endemic. Recognizing that S. Typhi infections exhibit a range of clinical severity, and that passive surveillance that detects ill patients seeking care at fixed health-care facilities tends to detect more severe cases than are detected by active household surveillance or other more active forms of surveillance, these published reports utilized systematic passive surveillance (14,15,45,46,47). Three studies, two carried out in urban slums in Kolkata and North Jakarta and the other in rural Dong Thap Province in the rural Mekong Delta of Viet Nam, included surveillance in all age groups from infancy through adulthood (14,15). The data indicate that the peak age incidence of typhoid fever in Asia is seen in school-age children. A fourth surveillance study in Pakistan that did not include infants 0–11 months of age and toddlers 12–23 months of age showed the peak incidence to be in preschool children (14).

Systematic health facility-based passive surveillance during efficacy trials of typhoid vaccines has established the incidence of confirmed typhoid in populations of more limited age range, particularly in school-age children (48–54). In several other typhoid vaccine field trials, active surveillance methods were used to detect cases based on systematic weekly or biweekly visits to participants’ households to detect subjects with fever; blood cultures were then collected from the febrile subjects (55,56). Since vaccine efficacy trials are typically carried out in known “hot spots” of endemicity, caution must be taken in extrapolating such incidence rates to other populations. Collectively, these studies indicate that in most endemic areas typhoid fever of a severity that leads to seeking health care peaks in school-age children and this age group must be an important target for specific intervention with typhoid vaccines.

Some other population-based studies not associated with vaccine trials have also been carried out, particularly in Asia, that have utilized active household surveillance to detect persons with fever, who then had blood cultures to look for S. Typhi bacteraemia (57,58). These studies have demonstrated a high incidence of S. Typhi bacteraemia in toddlers and preschool children (12–59 months of age) with a low incidence in infants < 12 months of age (57,58,59); an exception was household-based surveillance in Karachi, Pakistan that detected a high rate of S. Typhi and S. Paratyphi bacteraemia in infants (59a). It is difficult to assess the severity of clinical illness in the young paediatric patients in whom S. Typhi bacteraemia detected by active surveillance in which any elevation of temperature > 38 °C, irrespective of duration, led to the collection of a blood culture (57,58). In the 1980s, systematic health centre-based surveillance for S. Typhi bacteraemia in febrile infants and toddlers < 24 months of age in Chile and Peru indicated that in those endemic settings the S. Typhi (and S. Paratyphi) bacteraemias were clinically mild and self-limited (resolving before antibiotics were administered) (45,60), and were designated “benign bacteraemia” (45,60). In some other geographic areas where bacteraemia in febrile infants was systematically studied, the isolation of S. Typhi was distinctly rare in infants < 12 months of age (15,47,57,58,61). In several health centre and hospital-based systematic studies of bacteraemia in sub-Saharan Africa where rates of isolation of other invasive bacteria, such as Haemophilus influenzae type b (Hib), Streptococcus pneumoniae and non-typhoidal Salmonella (NTS) were impressively high, S. Typhi and S. Paratyphi were either absent or only rarely isolated (61,62,63); several of those studies were population-based

Page 15: Salmonella enterica serovar Typhi (typhoid) vaccines

5

(61,62). Other hospital-based systematic surveillance blood-culture studies among febrile patients carried out elsewhere in Africa revealed S. Typhi and S. Paratyphi, as well as NTS, mainly among preschool and school-age children (46,64,65). Young children with typhoid exhibit less prominent pathology of the small intestine (45). Together, these features suggest that young children readily become infected with S. Typhi, but that a distinct pathogenesis usually produces a mild illness.

If the appropriate typhoid vaccines become available, infants are an attractive target age group for programmatic immunization because of the existing contacts within the Expanded Programme on Immunization (EPI). Immunization of infants should be useful in those areas in Asia (e.g., urban slums in some cities) and elsewhere, that exhibit a high incidence of disease in preschool children. For infant immunization with typhoid vaccines in the EPI to protect against the peak age-specific incidence as observed in most endemic areas, school-age children, the vaccine will have to confer long-term protection. If the typhoid vaccines elicit strong immunologic memory, long-term protection may perhaps also be achieved if repetitive contact of vaccinated individuals with S. Typhi in endemic areas naturally boosts and extends vaccine-induced protection.

1.3.1 Occurrence in industrialized countries

In industrialized countries, a large proportion of sporadic cases occur among travellers (10,17,66–70). When outbreaks occur in the United States of America and other industrialized countries that have high levels of sanitation, controlled water supplies, and sophisticated food hygiene practices, a careful epidemiological investigation almost always incriminates the food or water vehicle responsible for the outbreak, and often identifies a chronic carrier responsible for its contamination (71,72,73). Similarly, epidemiological investigation of sporadic, non travel-associated cases in industrialized countries often identifies a chronic carrier, such as an elderly family member, who serves as the reservoir of infection (74).

1.3.2 Chronic carriers

Only a few quantitative studies have been made of the prevalence of chronic typhoid and paratyphoid carriers in less-developed countries. One such survey carried out in Santiago, Chile, during a period when typhoid fever was highly endemic, estimated a crude prevalence of 694 typhoid carriers/105 population (75). This high prevalence was related to the high overall prevalence of cholelithiasis in the adult female population of Santiago, as well as the high prevalence of chronic S. Typhi and S. Paratyphi carriers documented among 1000 consecutive patients with gallbladder disease undergoing cholecystectomy (76). Among 404 patients (316 female and 88 male) with gallbladder disease undergoing cholecystectomy in Kathmandu, Nepal, a high prevalence of S. Typhi (3.0%) and S. Paratyphi A (2.2%) was also documented (77). However, since the overall prevalence of cholelithiasis in the Kathmandu population was not known, an overall prevalence of chronic carriage in that population could not be calculated.

Page 16: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines6

1.4 Epidemiology of other enteric fever agents: Salmonella Paratyphi A, B and C

An individual case of paratyphoid fever due to S. Paratyphi A or B appears clinically indistinguishable from a case of typhoid fever caused by S. Typhi (3,4), although at the population level paratyphoid infections appear to be somewhat less virulent as judged by lower rates of complications and of hospitalizations. In general, in most endemic areas, approximately one clinical case of enteric fever is attributed to S. Paratyphi A or B for every four cases of S. Typhi disease. In recent years there has been a surge of enteric fever cases in Asia caused by S. Paratyphi A, with the strains carrying R factors encoding resistance to multiple, clinically-relevant antibiotics (78). This has also been accompanied by an increase in the frequency of S. Paratyphi A (particularly multi-resistant strains) as a cause of enteric fever in North American and European travellers who visit Asia.

S. Paratyphi C is also capable of causing enteric fever. However, more often the clinical syndromes of patients infected with this pathogen are sepsis or focal metastatic purulent infections (similar to the clinical illnesses in humans caused by S. Choleraesuis).

Page 17: Salmonella enterica serovar Typhi (typhoid) vaccines

7

2.1 Pathogenesis of Salmonella Typhi and Salmonella Paratyphi disease

S. Typhi and S. Paratyphi A and B are highly invasive bacteria that rapidly and efficiently pass through the intestinal mucosa of humans to eventually reach the reticuloendothelial system, where, after an 8- to 14-day incubation period, they precipitate a systemic illness. Salmonella Typhi is a highly host-adapted pathogen; humans comprise the only natural host and reservoir of this infection. Our comprehension of the steps involved in the pathogenesis of typhoid fever comes from four sources: (1) clinicopathological observations in humans (79); (2) volunteer studies (80); (3) studies of a chimpanzee model (81); (4) analogies drawn from S. Typhimurium and S. Enteriditis infection in mice—the “mouse typhoid” model (82). The probable steps in the pathogenesis of S. Typhi infection in humans are summarized below.

Susceptible human hosts ingest the causative organisms in contaminated food and water. The inoculum size and the type of vehicle in which it is ingested greatly influence the attack rate for typhoid fever and also affect the incubation period, a relationship that was documented in volunteer studies in the 1960s. Doses of 109 and 108 pathogenic S. Typhi ingested by volunteers in 45 ml of skim milk induced clinical illness in 98% and 89% of individuals, respectively; doses of 105 caused typhoid fever in 28% to 55% of volunteers, whereas none of 14 subjects who ingested 103 organisms developed clinical illness (80).

In the fasting normochlorhydric stomach, gastric acid undoubtedly inactivates many of the ingested typhoid bacilli. However, some foods may effectively buffer this otherwise formidable protective acid barrier, allowing many S. Typhi to survive. After passing through the pylorus and reaching the small intestine, the bacilli rapidly penetrate the mucosa by two main mechanisms to arrive in the lamina propria. One mechanism involves typhoid bacilli being actively taken up by M cells (83), the dome-like epithelial cells that cover Peyer’s patches and other organized lymphoid tissue of the gut. They are then passed to the underlying dendritic cells, macrophages and lymphoid cells. In the second, quite distinct mechanism, S. Typhi invade enterocytes and enter endocytic vacuoles that pass through the enterocytes to ultimately release the bacilli in the lamina propria without destroying the enterocyte (84). In yet a third mode of entry, it has been proposed that Salmonella may sometimes take a paracellular route, passing between enterocytes (85).

2. The pathogenesis and response to infection

Page 18: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines8

Upon reaching the lamina propria in the nonimmune host, typhoid bacilli elicit an influx of macrophages and dendritic cells that ingest the organisms but are generally unable to kill them. Some bacilli apparently remain within macrophages of the small intestinal lymphoid tissue. Other typhoid bacilli are drained into mesenteric lymph nodes where further multiplication and ingestion by macrophages take place. Eventually, there is a release of tumour necrosis factor-alpha, interleukin-2 (IL-2), IL-6, and other inflammatory cytokines by the mononuclear cells (and perhaps by enterocytes).

Postmortem studies have documented the inflammatory responses that occur in the distal ileum in the Peyer’s patches and other organized lymphoid aggregations. Presumably, cytokine release elicited by the intracellular S. Typhi is responsible for these cellular changes. It is from these lesions in the distal ileum and ascending colon that haemorrhage can occur later in the disease course. Gross bleeding comes from eroded vessels in or near the Peyer’s patches. This complication occurs late in the second week in about 1%–3% of untreated patients and rarely in patients whose treatment with an appropriate antibiotic is begun late in their clinical course. Perforations of the bowel wall occur in the same sections of the gut as the haemorrhages.

Shortly after invasion of the intestinal mucosa, a primary bacteraemia is believed to take place in which S. Typhi are filtered from the circulation by fixed phagocytic cells of the reticuloendothelial system. The main route by which typhoid bacilli reach the bloodstream in this early stage is by lymph drainage from mesenteric nodes entering the thoracic duct and then the general circulation. Conceivably, ingestion of a massive inoculum followed by widespread invasion of the intestinal mucosa could result in rapid and direct invasion of the bloodstream. As a result of this primary bacteraemia, the pathogen rapidly attains an intracellular haven throughout the organs of the reticuloendothelial system where it resides during the relatively long incubation period (usually 8–14 days) until the onset of clinical typhoid fever. Clinical illness is accompanied by a fairly sustained, albeit low level (1–10 organisms/ml) “secondary” bacteraemia.

Relapses were observed in about eight percent of patients suffering from typhoid fever in the pre-antibiotic era. The rate in patients treated with the first (chloramphenicol) and second (ampicillin, amoxicillin and trimethoprim/sulfamethoxazole) generation of antibiotics used for therapy of typhoid ranged from 10% to 35%. The organisms that initiate relapse appear to come from within the reticuloendothelial system. Typhoid bacilli can be isolated from liver biopsies during the relapse, and also from bone marrow, many months after the patient has fully recovered from the symptoms. Relapses usually occur about three weeks after the last febrile day or about two weeks after cessation of antibiotics. Most relapses are milder than the initial illness, last for only a few days, and respond promptly to appropriate antibiotic therapy. Relapse after treatment with oral fluoroquinolones and azithromycin and after parenteral ceftriaxone therapy is noticeably uncommon.

Page 19: Salmonella enterica serovar Typhi (typhoid) vaccines

9

During the primary bacteraemia that follows ingestion of typhoid bacilli and seeds the reticuloendothelial system, organisms also reach the gallbladder, an organ for which S. Typhi has a remarkable predilection (81,86). Salmonella Typhi can be readily cultured from bile or from bile-stained duodenal fluid in patients with acute typhoid fever (44,87,88). In approximately 2%–5% of patients, the gallbladder infection becomes chronic (89). The propensity to become a chronic carrier is greater in females and increases with age at the time of acute S. Typhi infection, thereby resembling the epidemiology of gallbladder disease (76,89). The infection tends to become chronic in those individuals who have pre-existent gallbladder pathology at the time of acute S. Typhi infection. Carriers shed as many as 109 organisms/g faeces (90). These organisms are propelled the length of the gastrointestinal tract without penetrating and without causing disease in the host.

2.2 Clinical and epidemiologic significance of Vi-negative S. Typhi

The Vi antigen is a virulence property and in most endemic areas and outbreaks almost all strains freshly isolated from patients possess this polysaccharide capsule (32). Nevertheless, Vi-negative strains have been occasionally reported, both in association with sporadic cases and with outbreaks (91). However, until recently, it was considered by many that these rare Vi-negative strains were regulatory mutants that occur during culture in vitro but that they are derived from strains that were Vi-positive in vivo. The fact that some Vi-negative strains could be induced to revert to a Vi-positive state following parenteral inoculation of mice supported this contention (92). Genetically stable Vi-negative strains were shown to be capable of causing typhoid fever in volunteers who participated in experimental challenge studies (80) and some authorities have cautioned that the widespread programmatic use of Vi polysaccharide and Vi conjugate vaccines might select for the emergence of Vi-negative strains (93,94). The recent application of modern microbiologic and molecular biologic methods has convincingly established that in some geographic foci in Asia a small proportion of the S. Typhi strains harbour deletions that make them indeed incapable of expressing Vi polysaccharide (95,96,97); these include S. Typhi detected directly in blood by polymerase chain reaction (PCR) techniques (95,97). Epidemiological observations and studies in volunteers support the contention that S. Typhi strains that possess Vi are more virulent than strains lacking this polysaccharide (80).

In the various controlled field trials of Vi and Vi conjugate vaccine carried out heretofore, there has been no report of the emergence of Vi-negative S. Typhi strains (93,94). On the other hand, the total number of subjects in the published field trials is small compared to the numbers of persons who would be immunized in programmatic public-health implementation of Vi vaccine for extended periods of time in large populations suffering a high incidence of typhoid fever. It is under those conditions that careful epidemiologic and microbiologic surveillance must be maintained in order to verify or refute the stated concern that Vi-negative strains might emerge.

2.3 Immune response to wild type Salmonella Typhi

Most of the measurements of immune response to natural infection with wild type S. Typhi were carried out in an era before there existed modern methods for characterizing and quantifying serum antibody, SIgA intestinal antibody and cell-mediated immune responses. The serum antibody, SIgA intestinal antibody and cell-mediated immune responses are relatively strong following natural infection (80,98–105).

Page 20: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines10

High titres of serum IgG antibodies directed at the capsular Vi polysaccharide antigen that covers Salmonella Typhi are encountered in ~ 80% of chronic biliary carriers of Salmonella Typhi (106,107) in endemic areas but only rarely in healthy individuals (106,107,108); 12%–38% of persons with acute typhoid fever manifest elevated Vi titres (106,107). Therefore, the measurement of Vi antibodies has been repeatedly utilized with success as a screening test to detect chronic carriers in nonendemic and endemic areas (71,106,107,109–112). In endemic areas a variable proportion of healthy subjects exhibit low titres of Vi antibody and the prevalence increases with age (55,113). The low titres of serum Vi antibody elicited in response to infection with wild type S. Typhi infection among persons living in endemic areas appears to reside mainly in the IgG2 sub-class (113).

Serum H (anti-flagella) antibodies following clinical infection are IgG and long-lived and have been used for seroepidemiologic surveys (98,114). Murphy and associates (115,116) observed that the peripheral blood mononuclear cells (PBMCs) from healthy adults living in typhoid-endemic areas who have no known history of acute typhoid fever often specifically proliferate when exposed to S. Typhi antigens. This corroborates results of antibody prevalence studies that indicate that considerable mild or subclinical infection occurs in such areas.

Page 21: Salmonella enterica serovar Typhi (typhoid) vaccines

11

3.1 Current WHO guidelines for typhoid vaccine use

The most recent WHO guidelines for use of the typhoid vaccines were published in 2008 (117).

3.2 Licensed typhoid vaccines

3.2.1 Killed whole cell parenteral vaccines

Inactivated (heat-killed, phenol-preserved) Salmonella Typhi were utilized as parenteral vaccines as far back as 1896 by Pfeiffer and Kolle in Germany and Wright in England (118). Randomized, placebo-controlled field trials in the 1960s sponsored by the World Health Organization established the efficacy of the heat-phenolized and acetone-inactivated parenteral whole cell typhoid vaccines (119–124). Although these vaccines were shown to confer moderate protection that persisted for up to seven years (124), they never became well-accepted public-health tools because they frequently caused systemic adverse reactions (about 25% of recipients developed fever and malaise) (121,122,125). The parenteral killed whole cell vaccines have been supplanted by two newer vaccines: attenuated Salmonella Typhi strain Ty21a used as a live oral vaccine and purified Vi capsular polysaccharide utilized as a parenteral vaccine (13,126). These two newer vaccines, which are at least as effective as the parenteral whole cell vaccines, but are well-tolerated, are licensed and available in many countries and constitute credible public-health tools.

The venerable inactivated whole cell parenteral vaccines, including heat-inactivated, phenol-preserved vaccine and acetone-inactivated vaccine, conferred moderate levels of protection for up to seven years (120–124). However, they were distinctly unsatisfactory vaccines because they caused adverse reactions at such high frequency. In controlled trials, approximately 25% of recipients of the inactivated whole cell vaccines developed fever and systemic reactions, and about 15% had to miss work or school (124,125). For this reason, these vaccines are no longer being manufactured and have been replaced by two modern licensed vaccines: live oral vaccine strain Ty21a and parenteral purified Vi capsular polysaccharide vaccine (13). The early type of killed whole cell parenteral vaccines mainly stimulated serum antibodies, including to S. Typhi O and H antigens and to a variable degree to Vi polysaccharide (127). Some data suggested a correlation between H antibody responses elicited by killed whole cell vaccines in animals and humans and protection against typhoid fever in field trials (128). Other data argued for serum Vi antibody elicited by killed whole cell vaccines as being a correlate of protection (129). These killed whole cell parenteral vaccines are mentioned for historical context only, since they are practically no longer available.

3. Typhoid vaccines

Page 22: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines12

3.2.2 Attenuated Salmonella Typhi strain Ty21a live oral vaccine

Ty21a, an attenuated strain of S. Typhi that is safe and protective as a live oral vaccine, was developed in the early 1970s by chemical mutagenesis of a pathogenic S. Typhi strain Ty2 (130). The characteristic mutations in this strain include an inactivation of the galE gene that encodes an enzyme involved in lipopolysaccharide (LPS) synthesis and by the inability to express Vi polysaccharide. However, approximately two dozen additional mutations are also present in Ty21a (131). Although Ty21a has proved to be remarkably well tolerated in placebo-controlled clinical trials (13), it is not clear precisely what mutations in addition to inactivation of galE and of Vi synthesis are most responsible for the stable, impressive attenuation of this vaccine.

Ty21a provides significant protection without causing adverse reactions. Results of multiple double-blind, placebo-controlled studies that utilized various surveillance methods to assess the reactogenicity of Ty21a in adults and children showed that adverse reactions for any symptom or sign were not observed significantly more often in the vaccine recipients than the placebo group (48–51,132). Controlled field trials of Ty21a emphasize that the formulation of the vaccine, the number of doses administered and the spacing of the doses markedly influence the level of protection that can be achieved (48–51,133,134,209). Two formulations are licensed, including enteric-coated capsules and a “liquid” formulation in which lyophilized vaccine is reconstituted along with buffer powder into a vaccine cocktail; however, in recent years, only the enteric-coated capsule formulation has been manufactured. Based on a field trial in Chile that demonstrated that three doses of Ty21a in enteric-coated capsules given on an every other day schedule conferred 67% efficacy over three years of follow-up and 62% protection over seven years of follow-up (48,135), this formulation and schedule are used worldwide, with the exception of North America. Canada and the USA recommend a four-dose regimen with an interval of 48 hours between doses. This was based on the results of a large-scale, randomized comparative trial carried out in Santiago, Chile whereby four doses of Ty21a in enteric-coated capsules given within seven days were shown to be significantly more protective than two or three doses (134).

In the mid 1980s industry succeeded in preparing a “liquid suspension” formulation of Ty21a for large-scale field trials that was amenable to large-scale manufacture. The new formulation consisted of two packets, one containing a dose of lyophilized vaccine and the other containing buffer. Contents of the two packets are mixed in a cup containing 100 ml of water and the suspension is then ingested by the subject to be vaccinated. Randomized placebo-controlled field trials were carried out in Santiago, Chile (49) and Plaju, Indonesia (51) to directly compare this new liquid formulation of Ty21a, similar to the formulation used in the first, highly encouraging field trial of Ty21a in Alexandria, Egypt (133), with the enteric-coated capsule formulation. In both trials the vaccine administered as a liquid suspension was superior to the vaccine in enteric-coated capsules. In the Santiago trial, over three years of follow-up, the liquid suspension formulation conferred 77% efficacy (95% confidence interval [CI], 60%–87%; p<0.0001), whereas in the Indonesian trial the liquid formulation conferred 53% efficacy (95% CI, 35%–66%; p<0.0001). Over two additional years of follow-up in the Santiago trial, the vaccine conferred 79% protection (95% CI, 56%–90%; p<0.001), giving a total five-year point estimate of vaccine efficacy of 78% (65%–87%; p<0.001).

Page 23: Salmonella enterica serovar Typhi (typhoid) vaccines

13

3.2.3 Purified Vi polysaccharide parenteral vaccine

In the 1970s and early 1980s, methods were developed to purify Vi capsular polysaccharide so that it was 99.8% free of contaminating LPS and was not denatured (32,55,136,137,138). This was an important breakthrough because as little as 5% impurity with LPS can result in systemic adverse reactions in a small percentage of recipients (137). By contrast, when Vi vaccine is highly purified, it is well-tolerated and febrile reactions are observed in only 1%–2% of subjects. In clinical trials, well-tolerated 25 µg and 50 µg single parenteral doses of purified Vi stimulated rises of serum Vi antibodies in the vast majority of vaccinated adults and school-age children (55,137,138). Administration of subsequent parenteral doses did not achieve boosts in antibody (139); thus, the Vi polysaccharide functions as a T cell-independent antigen. The antibodies elicited by a single dose persist in some subjects for up to three years (140,141,142). However, follow-up of groups of adults from non-endemic areas who were immunized with a dose of Vi vaccine show that serum Vi antibodies in general drop markedly after two years and less than one-half of subjects still have protective levels (141,142). Passive surveillance carried out during field trials showed the Vi vaccine to be as well-tolerated as the licensed (meningococcal and pneumococcal) polysaccharide vaccines that served as the control preparations in these trials (55,138).

Two randomized, controlled, double-blind field trials were carried out in Nepal (55) and South Africa to assess the efficacy of a single 25 µg dose of non-denatured purified Vi vaccine. Over 17 months of surveillance in Nepal, the vaccine conferred 72% vaccine efficacy (95% CI, 42%–86%; p=0.004) (55). In South Africa, the vaccine provided 64% protection (95% CI, 36%–79%; p<0.001) over 21 months of follow-up (138) and 55% efficacy (95% CI, 30%–71%; p<0.0001) over three years (143). The Nepal trial included all ages from preschool to adulthood (55), whereas the South African trial was performed in schoolchildren (138). A third controlled field trial, carried out in subjects 3–50 years of age in Guangxi, China, evaluated the protective efficacy of a single 30 µg dose of a Vi polysaccharide vaccine manufactured in China (52). The vaccine conferred 69% efficacy (95% CI = 28%–87%; p<0.0038) over 19 months of follow-up.

An advantage of the Vi vaccine is that it provides a moderate level of protection after just a single dose. On the other hand, there are no robust data to suggest that the efficacy of Vi persists beyond three years; efficacy was 55% at three years in the South African trial (143). This concern over the relatively short duration of protection of Vi and the need for reimmunization to extend protection has been heightened following an epidemiologic investigation of an outbreak of typhoid fever that occurred among Vi-vaccinated French soldiers deployed to the Ivory Coast (144). Prior to the outbreak, the standard operating procedure had been to immunize French soldiers with Vi vaccine every five years. The outbreak investigation revealed that receipt of Vi more than three years earlier was associated with a significantly increased risk of developing typhoid fever during the outbreak (144).

Public-health authorities in China responded to an outbreak of typhoid fever in schoolchildren by offering Vi vaccine. A comparison of the attack rate of culture-confirmed and possible typhoid fever in recipients of Vi, and non-recipients, suggested that the vaccine may have conferred ~ 70% protection during the outbreak (145). Vi vaccine may therefore be useful in controlling epidemics of typhoid fever.

Page 24: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines14

Immunity to S. Typhi is complex and, depending on the vaccine, involves serum (mainly IgG) antibodies, intestinal secretory IgA antibodies or systemic cell-mediated immune (CMI) responses that include contributions from CD4+ T helper (Th) and CD8+ cytotoxic T-cells (CTL). Serum antibodies to S. Typhi surface antigens such as Vi play an important role in defense against typhoid bacilli when they are extracellular. By contrast, it is CMI that eliminates S. Typhi when they are intracellular in the human host and are shielded from contact with antibodies and complement. As described below, oral Ty21a vaccine elicits a broad immune response against S. Typhi that includes both humoral and CMI components, while Vi polysaccharide vaccine stimulates serum IgG Vi antibodies that appear to attack S. Typhi while the bacteria are extracellular.

4.1 Ty21a live oral vaccine

4.1.1 Serum antibody responses

Ty21a is not a powerful stimulator of serum antibody responses. Nevertheless, measured by enzyme-linked immunosorbent assay (ELISA), the majority of recipients of Ty21a manifest modest but significant rises in serum IgG antibody against the O polysaccharide of S. Typhi (120,132,146). The rate of seroconversion of IgG anti-O antibody increases as more doses are administered within a period of seven days (120), or if more Ty21a organisms are administered per dose (147). Since Ty21a does not express Vi polysaccharide, anti-Vi antibody responses are not induced.

4.1.2 Mucosal antibody responses

Intestinal (jejunal fluid or faecal) secretory IgA antibody responses have been reported following oral immunization with various formulations and regimens of Ty21a, including several known to be protective (147–150).

4.1.3 Gut-derived IgA antibody secreting cells following oral immunization with Ty21a

Approximately 7–10 days following oral immunization with Ty21a, antibody secreting cells (ASCs) that produce specific IgA to S. Typhi O polysaccharide, can be readily detected among peripheral blood mononuclear cells. The magnitude of the IgA O ASC response was greater in individuals who received formulations that gave better efficacy in field trials (151–156). The demonstration of trafficking IgA ASCs that make antibody to Typhi O polysaccharide is considered a measure of priming of the mucosal immune system. Most of these ASCs carry integrin α4β7 homing markers that direct the cells back to the intestinal mucosa (156,157,158).

4. Immune responses to currently licensed typhoid

vaccines

Page 25: Salmonella enterica serovar Typhi (typhoid) vaccines

15

4.1.4 Cell-mediated immune responses (CMI)

Oral Ty21a vaccine stimulates both CD4+ helper T-cells and CD8+ cytotoxic T-cell responses, both of which are believed to play roles in defence against S. Typhi. The CMI responses that follow oral immunization of subjects with Ty21a include lymphoproliferative responses, secretion of Th type-1 cytokines (e.g. interferon-γ [IFN-γ]), tumour necrosis factor-α (TNF-α), classical class Ia-restricted CD8+ CTLs (159,160) and novel, non-classical class-Ib HLA-E-restricted CD8+ CTLs (160,161). Based on their expression of homing molecules (CD62L, CCR7), CD45RO or CD45RA, two main populations of memory T-cells (TM) are recognized: T central memory (TCM) and T effector memory (TEM) (162,163). Although TEM populations appear ready for immediate effector action, TCM are important in the generation of a new wave of effector T-cells. Flow cytometric analysis has shown that T-cell clones derived from Ty21a vaccinees exhibit a T-effector memory phenotype, i.e. CCR7(-), CD27(-) CD45RO(+) CD62L(-), and co-express gut homing molecules (i.e. high levels of integrin α4β7, intermediate levels of CCR9, and low levels of CD103) (161). The long-term T-cell responses to Ty21a are oligoclonal and involve multiple T-cell receptor (TCR) Vbeta families. The S. Typhi-specific CD8(+) T-cells bearing defined Vbeta specificities that appear after oral immunization with Ty21a are phenotypically and functionally consistent with T effector memory (TEM) cells with gut homing potential (161). Some of these key findings were confirmed by other investigators (164,165). These responses are likely to be important in resistance to S. Typhi by several mechanisms, including increased macrophage bactericidal activity, enhancement of antigen presentation and lysis of S. Typhi-infected targets. Notably, no correlations were observed between CMI and serum antibody responses in multiple studies (166,167,168).

4.1.5 Antibody-dependent cellular cytotoxicity responses

Another interesting immune response has been described following oral immunization with Ty21a in which post-immunization sera from vaccinees mixed with peripheral blood mononuclear cells and wild type Salmonella Typhi allows intracellular killing of the bacteria (148,169,170). Hence, this can be viewed as a form of antibody-dependent cellular cytotoxicity. Pre-immunization sera are not able to support intracellular killing. Notably, the specificity and biological activity of the post-vaccination serum antibodies have been reported to reside within IgA class.

4.2 Purified Vi polysaccharide

4.2.1 Serum antibodies

Parenteral administration of purified Vi polysaccharide stimulates serum antibodies, including IgG, in approximately 85%–95% of vaccinees > 2 years of age (55,108,137,138,139,142,171–176); by contrast, responses in toddlers were weak and shortlived (171). The serum IgG antibodies elicited by this vaccine are believed to mediate the observed protection (55,138,143). As this polysaccharide behaves as a T-independent antigen, immunologic memory is not stimulated (139,172). Vi polysaccharide, like other purified polysaccharide vaccines, stimulate generally shortlived B-cell responses by cross-linking the B-cell receptor (177). This drives the B-cells to differentiate to plasma cells that produce Vi antibody and results in a depletion of the pool of memory B-cells without the generation of new memory B-cells (177). Thus, without the induction of immunologic memory, whatever serum antibody levels are reached after an initial parenteral inoculation with Vi cannot be boosted

Page 26: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines16

further by the administration of additional doses. Moreover, the depletion of memory B-cells implies that the administration of subsequent doses of the T-independent polysaccharide vaccine may be accompanied by lower serologic responses than were observed after the initial vaccination (hyporesponsiveness). This is a phenomenon that has been well described with meningococcal polysaccharide (178,179) and pneumococcal polysaccharide (180,181) vaccines. Hyporesponsiveness has not been specifically looked for with Vi vaccine. However, there are some data to suggest that the response to a re-immunization with Vi two to three years after the initial immunization, when antibody titres have fallen, does not fully restore the titres to the peak level observed after the initial immunization (142). By contrast, another study showed that the titres following a re-immunization with Vi 27 months after the initial immunization did restore serum anti-Vi to the same peak titre recorded after the initial immunization with Vi more than two years earlier (139). A study with locally-produced Vi vaccine was carried out in China to address the safety and immune responses of re-vaccination of school-age children three years after an initial Vi vaccination versus response to a primary immunization with Vi (182). Re-vaccination was well tolerated. The geometric mean titre (GMT) of serum Vi antibodies measured by passive haemagglutination was 1:40 in primary vaccines and 1:29 in re-vaccinated children (p=0.24); the fold rise in anti-Vi was 7.7-fold in primary vaccinees but was 3.1-fold in re-vaccinated children (p<0.001) (182).

Although the administration of repetitive spaced doses of Vi vaccine fail to boost the levels of IgG Vi antibody above the level achieved following the initial dose, when protective levels of serum IgG antibody drop after ~ 2–3 years, re-immunization with a dose of Vi vaccine returns the antibody titres to the level observed after the initial dose (139) or to a level about two-thirds of the level initially achieved (geometric mean 1.81 mcg/mL versus 2.72 mcg/mL) (142).

Several investigators have proposed levels of serum Vi antibody titres following administration of Vi polysaccharide vaccine that might be considered a threshold for protection, including: 1.0 mcg/ml (143) and 1.5 mcg/ml (176). (Notably, based on the results of an efficacy trial with a Vi conjugate vaccine and serological measurements, a much lower threshold, 3.52 EU, equivalent to 0.11 mcg/mL of serum Vi antibody, has been proposed to constitute a protective threshold following administration of that vaccine) (54). Most of the serum IgG antibody stimulated by Vi vaccine is purported to be IgG1 subtype (183). Little is known regarding the biologic activity and the sites of action (e.g., bloodstream, intestinal surface, gallbladder) of the serum antibodies in humans stimulated by the Vi vaccine.

Page 27: Salmonella enterica serovar Typhi (typhoid) vaccines

17

When considering the public-health use of Ty21a and Vi typhoid vaccines, two critical biological parameters to be considered with respect to the efficacy of each of these vaccines are the point estimate of vaccine efficacy derived from large-scale, randomized controlled field trials, and the duration of efficacy.

5.1 Ty21a

5.1.1 Preliminary assessment of efficacy in experimental challenge studies in volunteers

The initial demonstration of the biological capacity of Ty21a to protect immunologically naive subjects against typhoid fever came from the experimental challenge of healthy adult volunteers who ingested multiple (five to eight) 1010 colony-forming unit (CFU) doses of Ty21a administered as freshly harvested organisms, and were challenged 5–9 weeks later with 105 CFU of Quailes strain wild type S. Typhi (184). Vaccinated subjects had a significantly lower attack rate than unimmunized control subjects (vaccine efficacy 87%, p=0.0002).

5.1.2 The different formulations of Ty21a that were assessed for efficacy in large-scale, controlled field trials

In pre-licensure studies, four different formulations of Ty21a were evaluated for efficacy in large-scale, randomized, double-blind, controlled field trials, including: (1) the “Alexandria” formulation consisted of vials containing ~ 3 x 109 CFU of lyophilized vaccine that were reconstituted with 30 ml of phosphate-sucrose diluent and administered after paediatric subjects chewed a sodium bicarbonate tablet to neutralize gastric acid (133,185).; (2) a gelatin capsule formulation containing ~ 1–3 x 109 CFU of lyophilized vaccine that was co-administered with two other gelatin capsules, each containing 0.5 gm of sodium bicarbonate to neutralize gastric acid (48); (3) a hydroxyprophymethylcellulosephthalate-coated, acid-resistant, enteric capsule formulation containing ~ 1–3 x 109 CFU of lyophilized vaccine (48). In vitro these enteric capsules resist gastric acid (pH 1.5) for at least two hours but dissolve within 10 minutes of exposure to artificial intestinal fluid of pH > 6.0. (4) A “liquid” formulation consisting of an aluminium foil sachet containing 1–3 x 109 CFU of lyophilized Ty21a and another aluminium foil sachet containing buffer powder to neutralize gastric acid. The contents of the two sachets are mixed together in a cup with 100 ml of water to create a “vaccine cocktail” that is ingested by the subject to be vaccinated (49).

Two of the above-mentioned formulations (enteric-coated capsules and “liquid” formulation) are currently licensed by regulatory authorities but only the enteric-coated capsule formulation is currently manufactured and marketed.

5. Vaccine efficacy

Page 28: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines18

5.1.3 Efficacy of the Alexandria formulation

In Alexandria, Egypt, ~ 32 000 primary school children 6–7 years of age were randomly allocated to receive three doses of vaccine or placebo administered in an every other day schedule (133,185). In total, 16 486 children were allocated to receive Ty21a vaccine, of whom 14 735 got all three doses, while 15 902 children were allocated to the control group and 14 557 of these ingested all three doses of placebo. Children seen at health clinics with fever of at least three days duration had blood cultures obtained to detect S. Typhi, and only bacteriologically-confirmed cases were considered in the assessment of vaccine efficacy. Over three years of follow-up, there were 22 cases of bacteriologically-confirmed typhoid fever in the 15 902 placebo recipients versus only a single confirmed case in the 16 486 vaccine recipients, yielding a point estimate of vaccine efficacy of 95.6% (CI, 77%–99%; p,0.001) (133). The breakthrough case of typhoid in the vaccine group was in a child who received only two doses of vaccine, and the illness occurred during the third year of follow-up.

Although the “Alexandria” formulation was not amenable to large-scale manufacture, the results of this first field trial corroborated the encouraging results observed in the immunologically-naive adult volunteer challenge study and demonstrated that the vaccine had the ability to protect children at risk in a typhoid-endemic area.

5.1.4 Efficacy of the gelatin capsule/bicarbonate formulation

The efficacy of the gelatin capsule/bicarbonate formulation was assessed in the same randomized, placebo-controlled field trial that also assessed the efficacy of the enteric-coated capsule formulation (48). The gelatin capsule/bicarbonate formulation gave poor protection and so further work with this formulation was abandoned (48).

5.1.5 Efficacy of the enteric-coated capsule formulation

The efficacy of the enteric-coated capsule formulation was evaluated in a large randomized, controlled trial in Area Occidente, Santiago, Chile, in which schoolchildren 6–19 years of age received three doses of this formulation of Ty21a according to two different immunization schedules (48 hours between doses, as in Alexandria, Egypt, or 21 days between the doses), or placebo. A total of 22 170 children received three doses of enteric-coated capsules by the short interval regimen (48 hours between doses), 21 598 received three doses by the long interval regimen (21 days between doses) and 21 906 children received placebo. Over three years of follow-up, there were 68 cases of bacteriologically-confirmed typhoid fever in the placebo group and 23 cases in the short interval vaccine group, yielding a point estimate of vaccine efficacy of 67% (CI, 47%–79%; p<0.0001). There were 34 cases of confirmed typhoid fever in the long interval group, providing a point estimate of vaccine efficacy of 49% (CI, 24%–66%; p=0.0006).

Based on these data, the enteric-coated capsule formulation of Ty21a was commercialized and licensed in many countries, with a recommendation to administer the three doses with an every other day spacing between doses.

Page 29: Salmonella enterica serovar Typhi (typhoid) vaccines

19

5.1.6 Efficacy of the sachet “liquid” formulation

Following the success of the enteric-coated capsule formulation, leading to its licensure by many countries, a new sachet formulation was developed that bore certain similarities to the “Alexandria” formulation except that the dose of lyophilized vaccine was contained in a sachet, rather than in a vial, and the buffer was different. The so-called sachet “liquid” formulation was tested for efficacy in two randomized, controlled field trials in Santiago, Chile (49) and Plaju, Indonesia (51). Besides the possible enhanced efficacy that might be expected, this formulation was amenable for immunizing infants and toddlers, as well as older children and adults.

In the Santiago field trial (49), 36 623 schoolchildren 6–19 years of age were allocated to receive three doses (109 CFU per dose) of the liquid formulation of vaccine (administered as a vaccine cocktail with buffer), while 10 302 children of the same age were allocated to get an identically appearing placebo; doses of vaccine or placebo were given in an every other day schedule. Over three years of follow-up, the incidence of bacteriologically-confirmed typhoid fever was 272 cases per 105 schoolchildren, while the incidence of confirmed typhoid in the “liquid” formulation vaccine group was 63 cases per 105 schoolchildren. This difference yields a point estimate of vaccine efficacy of 77% (CI, 60%–87%; p<0.0001).

In the field trial in Plaju, Indonesia, subjects (3–44 years of age) were randomly allocated to receive three doses of liquid formulation of vaccine (N=5066) or three doses of “liquid” placebo (N=5146); an additional 5122 children received placebo in capsule formulation. Doses of vaccine and placebo were administered one week apart (51). Over 30 months of follow-up, among the 10 268 combined placebo recipients, the incidence of confirmed typhoid fever was 810 cases per 105 persons, while among the 5066 recipients of liquid formulation vaccine the incidence of confirmed typhoid was 379 cases per 105 persons. This difference indicates a point estimate of efficacy of 53% (CI, 36%–66%; p<0.0001) for the liquid formulation (51).

5.1.7 Duration of efficacy of the enteric-coated capsule and liquid formulations

Follow-up of the cohorts in the Area Occidente trial and in the liquid formulation field trial were continued for a total of seven years and five years, respectively, allowing a robust estimate of the protection over that extended period. Over seven years of follow-up, the enteric-coated capsule formulation of Ty21a conferred 62% vaccine efficacy (CI, 48%–73%; p<0.0001) (135,209). In the field trial in Area Suroriente, follow-up over five years demonstrated a point estimate of vaccine efficacy of 78% (CI, 65%–87%; p<0.0001) (135). These extended follow-up data document that Ty21a provides an impressive duration of protection.

Page 30: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines20

5.1.8 Evidence for cross protection against S. Paratyphi

During the field trials of Ty21a in Area Occidente and Area Norte in Santiago, Chile there was a considerable amount of paratyphoid fever due to S. Paratyphi B, thereby allowing an estimation of vaccine efficacy against S. Paratyphi B (186). There was a trend in each trial indicating protection against S. Paratyphi B disease (56% efficacy in Norte and 38% efficacy in Occidente) (186). To enhance statistical power, an analysis was performed of pooled data from the two trials. The pooled analysis was justified by the following facts: epidemiologic surveillance and microbiologic methods in the two trials were identical; the trials overlapped during 22 of the 36 months of follow-up in each trial; the estimates of efficacy against Paratyphoid B in the two trials were roughly similar, and the ratio of follow-up of vaccinees to controls in both trials was approximately 1:1. In the pooled analysis, Ty21a conferred significant protection against Paratyphoid B disease; 49% vaccine efficacy (CI, 8%–73%; p=0.019).

There was not enough disease caused by S. Paratyphi A in the Chilean field trials to assess vaccine efficacy against that pathogen. However, there were enough cases of S. Paratyphi A in the Plaju, Indonesia trial to allow an assessment of whether Ty21a appears to offer cross protection against this pathogen (51). In the Indonesian venue there was no hint that Ty21a can confer protection against paratyphoid A illness (51).

5.1.9 Indirect protection of non-vaccinated subjects

Evidence that Ty21a can provide indirect protection for non-vaccinated subjects comes from monitoring of the incidence rate in the placebo control group in the first of four large-scale field trials carried out in Santiago, Chile, and observing the indirect effect as mass immunization with Ty21a was carried out in other administrative areas of the city as part of additional large-scale field trials. During the first year of follow-up in a randomized, placebo-controlled field trial of efficacy in Area Norte, the incidence of confirmed typhoid in the control group was 227 cases per 105 school children (50). This incidence rate was similar to the incidence seen in the previous four years in school-age children in Area Norte. Follow-up during the second year of the field trial in Area Norte occurred coincident with the performance of a very large field trial in adjacent Area Occidente involving > 109 594 children, 80% of whom received one or another formulation or regimen of Ty21a vaccine and 20% of whom received placebo. During this second year of follow-up in Area Norte the incidence of confirmed typhoid fell to 139 cases per 105 schoolchildren, a decrease of 39% (120). Shortly before the third season of follow-up began in the Area Norte trial, ~ 217 000 children in Area Sur and Area Central of Santiago received two, three or four doses of the enteric-coated capsule formulation of Ty21a (134). The incidence of confirmed typhoid in the Area Norte control group fell by 50% from the previous year, to reach a rate of only 70 cases per 105 schoolchildren (120). A rate this low had not been seen in Area Norte for many years. During the fourth year of follow-up in Area Norte no additional field trial was carried out elsewhere in Santiago, and the incidence of confirmed typhoid rose by 30% over the incidence observed in the previous year (120). During the fifth year of follow-up an additional field trial was performed, mostly in Area Suroriente but also including six and seven year olds in Area Norte. The incidence of typhoid fell once again by 40% from the rate of the previous year (120). Collectively these data demonstrate that large-scale vaccination with Ty21a elsewhere in the city exerts an indirect herd immunity effect that diminishes the incidence rate in non-vaccinated subjects, an observation that is important from the public-health perspective.

Page 31: Salmonella enterica serovar Typhi (typhoid) vaccines

21

5.2 Vi vaccine

5.2.1 Pre-licensure efficacy trials in Nepal and South Africa of Vi vaccine manufactured in France

In five villages near Kathmandu, Nepal, 6907 subjects were randomized to receive a single intramuscular injection of 25 mcg of Vi polysaccharide (N=3457) or a dose of 23-valent pneumococcal polysaccharide vaccine (25 mcg per capsular type) (N=3450); 6438 of the subjects were between three and 44 years of age. Active surveillance was utilized to detect cases. Health workers visited the households every two days, except Sundays, to take temperatures. Subjects with a temperature of 37.8o C or above for at least three days were asked to have 6 ml of blood collected for culture. Over 17 months of follow-up there were 32 bacteriologically-confirmed cases of typhoid fever in the control group and nine confirmed cases of typhoid in the Vi vaccine group (55,209). These data indicate a point estimate of vaccine efficacy of 72% (CI, 42%–86%; p=0.004).

In the eastern Transvaal area of South Africa, 5692 schoolchildren (99% were 5–16 years of age) were randomly allocated to receive a 25 mcg dose of Vi vaccine, while 5692 control children received a dose of meningococcal A + C polysaccharide vaccine. Surveillance to detect cases was maintained by identifying children absent from school for at least three days and visiting them, and by drawing blood cultures from children who visited health clinics with fever (37.8o C or above) for at least three days. Over 21 months of follow-up, 47 cases of bacteriologically-confirmed typhoid fever were recorded in the control group versus 19 cases in the Vi vaccine group (138). This indicates a point estimate of vaccine efficacy of 60% (CI, 31%–76%; p<0.001). If cases are limited to subjects whose symptoms began more than six weeks after vaccination, the difference was 44 cases in the control group and 16 cases in the Vi vaccine group, yielding a vaccine efficacy estimate of 64% (CI, 36%–79%; p<0.001) (138).

5.2.2 Efficacy trials of a Vi vaccine manufactured in China

A randomized, placebo-controlled field trial was performed in Guangxi Zhuang Autonomous Region in southwestern China. Enrolled subjects were children, who have the highest incidence of typhoid fever in this setting. A total of 131 271 subjects ranging in age from 3–50 years (but the majority of school age) were randomly allocated to receive a single 30 mcg dose of Vi polysaccharide (manufactured in China) or saline placebo (52). The subjects were followed for 19 months with passive surveillance that detected cases seen in the Regional Health and Anti-epidemic Centre. Clinically suspect cases of typhoid fever were confirmed by blood culture. Over 19 months, there were 23 culture-confirmed cases of typhoid fever in the placebo group versus seven cases in the Vi group, yielding a point estimate of vaccine efficacy of 69% (CI, 28%– 87%; p=0.0038). Most of the cases occurred in school-aged children; 22 in the placebo group versus six in the Vi group (vaccine efficacy = 72%; CI, 32%–82%) (52).

Page 32: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines22

5.2.3 Evidence for cross protection against S. Paratyphi

Since neither S. Paratyphi A nor S. Paratyphi B expresses Vi capsular antigen, there is no biological basis for serum Vi antibodies elicited by Vi vaccine to provide cross-protection against these two serotypes. Not surprisingly, evidence from well-controlled field trials do not show any evidence of protection against disease caused by S. Paratyphi A or B (53). In theory, the Vi vaccine should offer protection against disease caused by S. Paratyphi C, which does express Vi capsular polysaccharide. However, this is a rare cause of enteric fever or other clinical forms of invasive disease, and there have been no data from field trials to document such protection, although it would be expected.

5.2.4 Indirect protection of non-vaccinated subjects

Evidence that Vi vaccine confers indirect protection comes from analysis of a Phase IV cluster randomized effectiveness trial in Kolkata, India in which 37 673 slum dweller children and adults got Vi vaccine (40 discrete geographic clusters) or hepatitis A vaccine (40 geographic clusters) (53). The clusters were stratified by ward and the number of residents who were age 18 years or younger (< 200 versus > 200 persons), and the number of residents who were older than 18 years of age (< 500 versus > 500 persons), resulting in eight strata. Within each stratum, one half of the clusters were randomly allocated to receive Vi vaccine and the other half hepatitis A control vaccine. The average rate of vaccine coverage was 61% for the Vi vaccine clusters and 60% for the hepatitis A vaccine clusters.

Typhoid fever was confirmed in 96 persons in the hepatitis A clusters versus 34 in the Vi vaccine clusters, providing a point estimate of vaccine effectiveness of 61% (CI, 4%–75%; p<0.001). In an adjusted statistical model, the Vi vaccine conferred significant indirect protection against confirmed typhoid fever among unvaccinated residents in the Vi vaccine cohorts, yielding a point estimate of 44% effectiveness (CI, 2%–69%; p=0.04) (53). The observation that considerable indirect protection of non-vaccinated persons against typhoid fever can be achieved with moderate levels of coverage with parenteral Vi vaccine is important from the public-health perspective.

Page 33: Salmonella enterica serovar Typhi (typhoid) vaccines

23

6.1 Ty21a

Formulation is important in the practicality and efficacy of Ty21a. Vaccine in enteric-coated capsules is highly practical for immunizing children above seven years of age (when virtually all children can readily ingest a capsule) and adults. However, in one large trial ~ 8 % of children < seven years of age were not able to swallow Ty21a in capsule formulation. By contrast, in this age group, all children could ingest a “vaccine cocktail” consisting of lyophilized vaccine and buffer powder reconstituted with 100 ml of water (49). This double sachet “liquid formulation” was shown to be highly practical for immunizing toddlers and preschool children in Asia, and led to high rates of seroconversion of serum IgG O antibody (187,188). Based on these data that documented the practicality and immunogenicity of Ty21a in toddlers and preschool children (187,188), it is assumed that such a formulation might also be amenable to use in infants. Unfortunately, although licensed by regulatory authorities in a number of countries, no manufacturer is currently producing a “liquid formulation” of Ty21a.

6.2 Vi

In pre-licensure studies, a lyophilized and a liquid formulation of Vi polysaccharide from one manufacturer were compared for clinical acceptability and immunogenicity (139). Only the liquid formulation was commercialized.

Two manufacturers produce combined hepatitis A-typhoid Vi combination vaccines to protect travellers from industrialized countries against both infections when they visit developing countries (189).

6. Vaccine formulation issues

Page 34: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines24

7.1 Ty21a

At the time the large-scale, randomized, controlled, pre-licensure field trials in Santiago, Chile were evaluating the efficacy of the enteric-coated capsule formulation of Ty21a, it was not clear if the primary mechanism of protection was mediated via antibodies (either intestinal SIgA or serum IgG) or via cell-mediated immune mechanisms. Some immunologic correlations, including the enumeration of IgA antibody secreting cells that make IgA antibody to Salmonella Typhi O antigen, and titres of serum IgG anti-O antibody, suggested that humoral responses might be important. If humoral immune responses were in fact critical, one would expect that extending the interval between the administration of three doses of enteric-coated capsule of Ty21a to 21 days between doses, rather than using the every other day schedule, would enhance the level of protection. By contrast, if cell-mediated immune mechanisms were more critical, it would be expected that administering the doses at the short interval (every other day) regimen might elicit greater protection. These two regimens were directly compared in the large-scale field trial in Area Occidente of Santiago, Chile where 21 598 schoolchildren 6–19 years of age were randomly allocated to receive three doses by the long interval regimen (21 days between doses), 22 170 schoolchildren to get three doses of enteric-coated capsules by the short interval regimen (48 hours between doses) and 21 906 schoolchildren to receive placebo (48). During the initial three years of follow-up, there were 34 cases of bacteriologically-confirmed typhoid fever in the long interval group, 23 cases in the short interval group and 68 cases in the placebo group. For the long interval regimen this translates to a point estimate of vaccine efficacy of 49% (CI, 24%–66%) versus a point estimate of vaccine efficacy of 67% (CI, 47%–79%) for the short interval vaccine group. Although the overlapping confidence intervals bracketing the point estimates of vaccine efficacy indicate that the levels of efficacy were not statistically different, there was a trend to suggest that the short interval regimen (which was logistically simpler and more practical) was somewhat more efficacious.

In the large-scale, randomized, placebo-controlled field trial of the enteric-coated capsule formulation of Ty21a in Area Norte of Santiago, Chile, the efficacy of a single dose and of two doses (one week apart) of Ty21a were compared over five years of follow-up (50). In this trial, 27 620 schoolchildren were allocated to receive two doses of Ty21a, 27 618 got a single dose and 27 305 received placebo. The 2-dose regimen conferred significant protection during the first and second years of follow-up, including 52% vaccine efficacy during year 1 (CI, 26%–69%; p<0.001) and 71% vaccine efficacy during year 2 (CI, 44%–85%; p<0.001) (50). The level of efficacy fell to non-significant levels during year 3 (22% efficacy), year 4 (19% efficacy) and year 5 (7% efficacy).

7. Vaccine schedule issues

Page 35: Salmonella enterica serovar Typhi (typhoid) vaccines

25

When analysed separately, a single-dose of Ty21a in the Area Norte, Santiago, Chile field trial did not confer statistically significant protection during year 1 (25% efficacy) or year 2 (35%), However, when data from the first two years of follow-up are pooled and analysed, the modest point estimate of vaccine efficacy of 28.8% (CI, 4%–47%) was statistically significant (p=0.033). Collectively, these data document that two doses and even just a single dose of Ty21a in enteric-coated capsules provide some measureable protection against typhoid fever.

In the large-scale effectiveness trial in Area Sur and Area Central of Santiago, Chile, 225 998 schoolchildren were randomly allocated to receive either two, three or four doses of vaccine (every other day interval between doses). A total of 216 692 children received at least one dose of vaccine and 189 819 schoolchildren (84%) received all doses of the randomly assigned scheme; there was no placebo group in this effectiveness trial. The incidence of bacteriologically-confirmed typhoid fever in the three vaccine groups was compared after three years of follow-up. The incidence of confirmed typhoid was lowest in the four-dose group, highest in the two-dose group and intermediate in the three-dose group of schoolchildren. In both a per protocol analysis and in an “intent to treat” analysis the group that got four doses had a significantly lower incidence of typhoid fever than the group that got three doses of Ty21a. Based on the additional protection derived from administration of a fourth dose of Ty21a, the US Food and Drug Administration, and Health Canada, licensed Ty21a as a four-dose vaccine regimen, whereas a three-dose regimen is licensed in other countries worldwide.

7.2 Vi

Once Phase II trials demonstrated that Vi polysaccharide does not elicit immunologic memory and that additional spaced doses of Vi do not boost serum Vi antibody levels above the levels stimulated by a single dose, subsequent field trials and effectiveness trials utilized only a single-dose regimen. However, a relevant question is the interval at which re-immunization with Vi should be undertaken to extend the period of protection once the antibodies from an earlier immunization have fallen or disappeared. Monitoring of serum Vi antibody has shown that Vi antibody levels in a large proportion of vaccinated subjects fall to putatively non-protective levels after two years, and are even lower by three years (141). Most efficacy and effectiveness trials of Vi vaccine have maintained follow-up for less than two years. However, one field trial has provided data for three years (143) and another for six years (190). Follow-up of subjects in those vaccine efficacy trials shows that at three years the level of protection falls (143,190) and evidence of efficacy is absent by the fourth year (190). Additional data relevant to the duration of efficacy of Vi and the question of when to institute re-immunization comes from an outbreak of typhoid fever among French military personnel who had been immunized with Vi vaccine (144). An outbreak investigation revealed that a significant risk factor for developing typhoid fever among the military personnel was having received Vi vaccine more than three years before the outbreak exposure (144); risk was also significantly higher in individuals who had received the vaccine more than two years before.

Page 36: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines26

Based on the evidence summarized above, one manufacturer of Vi vaccine recommends re-immunization after two years if there is continued exposure to S. Typhi. Based on the epidemiologic and serologic data at hand, it would appear that in endemic countries re-immunization of high-risk target populations (e.g. schoolchildren) should not be spaced more than three years apart. Little data are available on whether repeated re-immunizations with Vi polysaccharide can elicit hyporesponsiveness to the antigen, as has been reported with some other bacterial polysaccharide vaccines, such as meningococcal C and 23-valent pneumococcal polysaccharides (178,179,181).

Page 37: Salmonella enterica serovar Typhi (typhoid) vaccines

27

8.1 Ty21a

An effectiveness trial involving 225 998 schoolchildren was carried out in the Southern (Sur) and Central administrative areas of Santiago, Chile to compare directly the protective effects of two, three, or four doses of Ty21a vaccine in enteric-coated capsules, and to assess the logistical practicality of Ty21a as a public-health tool; 216 692 children received at least one dose of vaccine (134). This trial established the logistical practicality of school-based mass immunization of school-age children with an oral typhoid vaccine that involved the administration of multiple doses of vaccine (either two, three or four) within a 7-day period. One salient feature of this trial was the observation that ingestion of four spaced doses of vaccine resulted in a significantly lower incidence of typhoid than ingestion of three spaced doses (every other day schedule). Among children eight years of age and above there were no problems with ingestion of enteric-coated capsules of Ty21a vaccine. On the contrary, oral vaccination of classes went smoothly. Among six and seven year olds, ~ 5% had difficulty swallowing a capsule.

8.2 Vi

In Asia there have been multiple effectiveness trials of Vi polysaccharide vaccine prepared by different manufacturers (53,191,192,193). These effectiveness trials have been large-scale and several were performed in areas where the incidence of typhoid was quite high, such as in, Kolkata, India, Jakarta, Indonesia and Karachi, Pakistan. The age groups collectively ranged from pre-school children to the elderly. These large trials established unequivocally that both school-based immunization (192) as well as community mass immunization of adults and children (191,193) could proceed, with appropriate prior mobilization of the community.

One striking difference in effectiveness noted across two post-licensure, randomized, controlled clinical trials was the level of effectiveness recorded in preschool children. In a cluster randomized trial in Kolkata (53), significant protection was observed in children 2–4 years of age (53). Moreover, the protection seen in the 2–4 year olds (80%, 95% CI, 53%–91%) was higher (albeit not statistically significant) than that observed in 5–14 year olds (56%, 95% CI, 18%–77%) or older subjects (46%, 95% CI, 43%–79%). This was an unexpected result, since efficacy trials with other types of typhoid vaccines, such as parenteral killed whole-cell vaccines (124,125) and oral Ty21a (48), showed lower protection in younger children than in older children (48,124,125). In contrast to the Kolkata results, in an effectiveness trial carried out in Karachi, Pakistan, there was no evidence that Vi vaccine protected preschool children < 5 years of age, while it conferred significant protection on older subjects (Professor Zulfiqar Bhutta, Aga Khan University, personal communication).

8. Vaccine effectiveness

Page 38: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines28

9.1 New generation vaccines under development

9.1.1 Attenuated strains as single-dose oral vaccines

New strains of S. Typhi that have been genetically-engineered to contain precise attenuating mutations have been successfully tested in Phase I and Phase II clinical trials, and shown to be well tolerated and immunogenic after ingestion of just a single oral dose. These live oral vaccine candidates include strains: M01ZH09, a derivative of the virulent S. Typhi strain Ty2 with deletion mutations in aroC and ssaV (a component of the type III secretion system encoded by Salmonella pathogenicity island-2) (194,195,196); Ty800, a Ty2 derivative deleted in phoP/phoQ (197); CVD 908-htrA, derived from Ty2, with deletion mutations in aroC, aroD and htrA (166,198); and CVD 909, a further derivative of CVD 908-htrA that constitutively expresses Vi (199,200).

9.1.2 Parenteral Vi conjugate

Booster doses of purified Vi do not raise antibody titres over those elicited by a single dose of vaccine; i.e. immunologic memory does not occur. To remedy this, Vi polysaccharide has been conjugated to a carrier protein (recombinant exotoxin A of Pseudomonas aeruginosa) to increase its immunogenicity by conferring T-cell-dependent properties upon the antigen, including the induction of immunologic memory. In children and adults in endemic areas, booster doses of Vi conjugate vaccine clearly increase the titres of antibody over those elicited by a priming dose (54,56,201). A randomized, controlled field trial of Vi conjugate in children immunized at ages 2–4 years in the Mekong Delta of Viet Nam demonstrated 91.5% (CI, 77%–97%) vaccine efficacy over 27 months of active surveillance (56) and 82% efficacy (CI, 22%–99%) during an additional 19 months of follow-up that utilized a passive surveillance system (54). This vaccine has not progressed to licensure.

Other Vi conjugate vaccines under development covalently link the Vi polysaccharide to tetanus toxoid (one licensed vaccine (202) and one in Phase III trial), diphtheria toxoid (pre-clinical) and CRM197 (Phase II) (203).

9. Future prospects

Page 39: Salmonella enterica serovar Typhi (typhoid) vaccines

29

9.2 Immune response to the new generation typhoid vaccines

9.2.1 Single-dose live oral typhoid vaccine candidatesAs discussed above, several live attenuated S. Typhi strains have been developed using recombinant deoxyribonucleic acid (DNA) technology. Among the candidates that have been evaluated in Phase I and Phase II clinical trials are Ty800 (197), CVD 908-htrA (166,198), CVD 909 (200,200a) and M01ZH09 (195,196,204,205). Oral immunization of adult volunteers with these vaccine strains elicited gut-derived IgA ASC and serum IgG and IgA antibodies to S. Typhi lipopolysaccharide O antigen. In the clinical trials with CVD 908-htrA, CMI responses were also intensively studied. CVD 908-htrA elicited robust CMI responses including S. Typhi-specific CD4+ T cells with the capacity to produce Th1-type cytokines (i.e. IFN-γ and TNF-α in the absence of interleukin 4 [IL-4] and [IL-5]), as well as CD8+ cytotoxic T-cells that kill S. Typhi-infected target cells (167). Immunization with CVD 909 also elicited a wide array of CMI responses similar to those induced by CVD 908-htrA (206). Further studies of these responses revealed that oral immunization with attenuated S. Typhi strains elicits diverse S. Typhi-specific IFN-γ-secreting CD4+ and CD8+ T central memory (TCM ) and TEM subsets that express, or not, integrin α4/β7. Thus, these cells are able to migrate to the gut (if they express integrin α4/β7) or secondary lymphoid tissues (if they express L-selectin, CD62L) (207). These studies provide the first demonstration in a bacterial system in humans that CD3+ CD8+ TEMRA subsets are stimulated in response to oral vaccination. CD3+ CD8+ TEMRA subsets are widely regarded as the most active effector cells in viral systems (207). Taken together, these results provide strong evidence for the contention that memory/effector B- and T-cells recirculate in humans. However, the studies to characterize these cells are only just beginning. Lymphoproliferative responses and IFN-γ production were also observed in subjects immunized with M01ZH09 (196). In clinical studies conducted in the USA and Viet Nam, M01ZH09 was well tolerated and immunogenic in children 5–14 years of age, as well as in adults. Collectively, these results illustrate the wide range of effector responses that can be elicited by a single immunization with the new generation of attenuated S. Typhi vaccine strains in different age groups, all of which are believed to play important roles in protection.

9.2.2 Vi conjugateTo enhance the immunogenicity of the Vi polysaccharide vaccine, Vi was conjugated to the recombinant exoprotein (rEPA) from Pseudomonas aeruginosa and it was found to be safe and immunogenic in a series of clinical studies that included adults living in the USA and in endemic areas. Children of school and pre-school age from highly endemic areas who received the Vi conjugate vaccine had increased levels of Vi IgG antibodies compared with those that received the unconjugated Vi polysaccharide vaccine (56,201). The immunogenicity of the Vi conjugate was shown to be dosage-dependent, with higher doses of Vi-rEPA inducing higher levels of Vi IgG antibodies (208). A dose of 25 µg of Vi as Vi-rEPA was recommended for immunization of 2–5 year old children. This dose induced the strongest serum IgG responses from the three doses tested (5, 12.5 and 25 µg) and conferred protection for at least four years (54). The Vi antibodies elicited by the conjugate vaccine are long lasting, reflecting the long-lived protection. At a point 42 months after children 2–5 years of age had been vaccinated, the geometric mean antibody level was 3.66 EU (interquartile range, 2.2–5.8 EU) compared to a geometric mean of 0.80 EU in children of the same age who had received placebo 42 months earlier (interquartile range, 0.30–2.26 EU) (54). Preliminary data from ongoing long-term follow-up studies in adults suggest Vi IgG antibody levels may remain elevated (> 7-fold over baseline) for as long as 10 years after vaccination.

Page 40: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines30

In a study in Viet Nam (210), 301 infants received EPI vaccines alone or with Vi-rEPA or Haemophilus influenzae type b-tetanus toxoid conjugate (Hib-TT) at 2, 4 and 6 months of age and Vi-rEPA or Hib-TT alone at 12 months. Maternal, cord and infant sera were assayed for IgG anti-Vi and for IgG antibodies to Hib capsular polysaccharide and the diphtheria, tetanus and pertussis toxins at 7, 12 and 13 months. In the Vi-rEPA group, IgG anti-Vi geometric mean (GM) increased from the cord level of 0.66 to 17.4 enzyme-linked immunosorbent assay units (EU) at seven months, declined to 4.76 EU at 12 months and increased to 50.1 EU one month after the fourth dose, whereupon 95% of infants had levels of ≥3.5 EU (the proposed protective level). Anti-Vi GM did not increase in the controls. Infants with cord anti-Vi levels of <3.5 EU achieved significantly higher IgG anti-Vi levels than infants with cord anti-Vi levels of ≥3.5 EU. Antibody responses to the routine EPI vaccines were comparable in all groups. Vi-rEPA was well tolerated in young infants, elicited putative protective levels of anti-Vi and was compatible with several concomitantly administered EPI vaccines.

Page 41: Salmonella enterica serovar Typhi (typhoid) vaccines

31

1. Stuart BM, Pullen RL. Typhoid. Archives of Internal Medicine, 1946, 78:629–661.

2. Osler W. Typhoid fever. In: Osler W, ed. The principles and practice of medicine, New York, D Appleton, 1892:2–43.

3. Vollaard AM et al. Identification of typhoid fever and paratyphoid fever cases at presentation in outpatient clinics in Jakarta, Indonesia. Transactions of the Royal Society of Tropical Medicine and Hygiene, 2005, 99(6):440–450.

4. Maskey AP et al. Salmonella enterica serovar Paratyphi A and S. enterica serovar Typhi cause indistinguishable clinical syndromes in Kathmandu, Nepal. Clinical Infectious Diseases : an official publication of the Infectious Diseases Society of America, 2006, 42(9):1247–1253.

5. Karkey A et al. Kathmandu, Nepal: still an enteric fever capital of the world. Journal of Infection in Developing Countries, 2008, 2(6):461–465.

6. Ochiai RL et al. Salmonella paratyphi A rates, Asia. Emerging Infectious Diseases, 2005, 11(11):1764–1766.

7. Threlfall EJ et al. Alternatives to ciprofloxacin use for enteric Fever, United Kingdom. Emerging Infectious Diseases, 2008, 14(5):860–861.

8. Gupta SK et al. Laboratory-based surveillance of paratyphoid fever in the United States: travel and antimicrobial resistance. Clinical Infectious Diseases : an official publication of the Infectious Diseases Society of America, 2008, 46(11):1656–1663.

9. Fangtham M, Wilde H. Emergence of Salmonella paratyphi A as a major cause of enteric fever: need for early detection, preventive measures, and effective vaccines. Journal of Travel Medicine, 2008, 15(5):344–350.

10. Meltzer E, Sadik C, Schwartz E. Enteric fever in Israeli travelers: a nationwide study. Journal of Travel Medicine, 12(5):275–281.

11. Wolman A, Gorman A. The significance of waterborne typhoid fever outbreaks, Baltimore, Williams & Wilkins, 1931.

12. [Anonymous]. Typhoid in the large cities of the United States in 1919. JAMA : the journal of the American Medical Association, 1920, 672–675.

13. Levine MM, Taylor DN, Ferreccio C. Typhoid vaccines come of age. The Pediatric Infectious Disease Journal, 1989, 8:374–381.

14. Ochiai RL et al. A study of typhoid fever in five Asian countries: disease burden and implications for controls. Bulletin of the World Health Organization, 2008, 86(4):260–268.

References

Page 42: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines32

15. Lin FY et al. The epidemiology of typhoid fever in the Dong Thap Province, Mekong Delta region of Viet Nam. The American Journal of Tropical Medicine and Hygiene, 2000, 62(5):644–648.

16. Mermin JH et al. Typhoid fever in the United States, 1985–1994: changing risks of international travel and increasing antimicrobial resistance. Archives of Internal Medicine, 1998, 158(6):633–638.

17. Steinberg EB et al. Typhoid fever in travelers: who should be targeted for prevention? Clinical Infectious Diseases : an official publication of the Infectious Diseases Society of America, 39(2):186–191.

18. Lynch MF et al. Typhoid fever in the United States, 1999–2006. JAMA : the journal of the American Medical Association, 302(8):859–865.

19. Blaser MJ et al. Salmonella typhi: the laboratory as a reservoir of infection. The Journal of Infectious Diseases, 1980, 142:934–938.

20. Blaser MJ, Lofgren JP. Fatal salmonellosis originating in a clinical microbiology laboratory. Journal of Clinical Microbiology, 1981, 13:855–858.

21. Parry CM et al. Typhoid fever. The New England Journal of Medicine, 2002, 347(22):1770–1782.

22. Gupta A. Multidrug-resistant typhoid fever in children: epidemiology and therapeutic approach. Pediatric Infectious Disease, 1994, 13:124–140.

23. Rowe B, Ward LR, Threlfall EJ. Spread of multiresistant Salmonella typhi. Lancet, 1990, 336:1065–1066.

24. Bhutta ZA. Impact of age and drug resistance on mortality in typhoid fever. Archives of Disease in Childhood, 1996, 75:214–217.

25. Wain J et al. Quinolone-resistant Salmonella typhi in Viet Nam: molecular basis of resistance and clinical response to treatment. Clinical Infectious Diseases : an official publication of the Infectious Diseases Society of America, 1997, 25(6):1404–1410.

26. Gilman RH et al. Comparison of trimethoprim-sulfamethoxazole and amoxicillin in therapy of chloramphenicol-resistant and chloramphenicol-sensitive typhoid fever. The Journal of Infectious Diseases, 1975, 132:630–636.

27. Butler T et al. Chloramphenicol-resistant typhoid fever in Viet Nam associated with R factor. Lancet, 1973, 2:983–985.

28. Goldstein FW et al. Plasmid-mediated resistance to multiple antibiotics in Salmonella typhi. The Journal of Infectious Diseases, 1986, 153:261–266.

29. Brenner FW et al. Salmonella nomenclature. Journal of Clinical Microbiology, 2000, 38(7):2465–2467.

30. Frankel G et a l . Intragenic recombination in a f lagel l in gene: Characterization of the H1-j gene of Salmonella typhi. The EMBO Journal, 1989, 8:3149–3152.

31. Guinee PA et al. An unusual H antigen (Z66) in strains of Salmonella typhi. Annales de Microbiologie, 1981, 132(3):331–334.

32. Robbins J, Robbins J. Reexamination of the protective role of the capsular polysaccharide Vi antigen of Salmonella typhi. The Journal of Infectious Diseases, 1984, 150:436–449.

Page 43: Salmonella enterica serovar Typhi (typhoid) vaccines

33

33. Looney RJ, Steigbigel RT. Role of the Vi antigen of Salmonella typhi in residence to host defense in vitro. The Journal of Laboratory and Clinical Medicine, 1986, 108:506–516.

34. Holt KE et al. High-throughput sequencing provides insights into genome variation and evolution in Salmonella Typhi. Nature Genetics, 2008, 40(8):987–993.

35. Kidgell C et al. Salmonella typhi, the causative agent of typhoid fever, is approximately 50,000 years old. Infection, Genetics and Evolution : journal of Molecular Epidemiology and Evolutionary Genetics in Infectious Diseases, 2002, 2(1):39–45.

36. Hickman-Brenner FW, Farmer JJ, III. Bacteriophage types of Salmonella typhi in the United States from 1974 through 1981. Journal of Clinical Microbiology, 1983, 17(1):172–174.

37. Thong KL et al. Analysis of Salmonella typhi isolates from Southeast Asia by pulsed-field gel electrophoresis. Journal of Clinical Microbiology, 1995, 33(7):1938–1941.

38. Baker S et al. High-throughput genotyping of Salmonella Typhi allows geographical assignment of haplotypes and pathotypes within an urban district of Jakarta, Indonesia. Journal of Clinical Microbiology, 2008, 46:1741–1746.

39. Parkhill J et al. Complete genome sequence of a multiple drug resistant Salmonella enterica serovar Typhi CT18. Nature, 2001, 413(6858):848–852.

40. Deng W et al. Comparative genomics of Salmonella enterica serovar Typhi strains Ty2 and CT18. Journal of Bacteriology, 2003, 185(7):2330–2337.

41. Kidgell C et al. Characterisation and distribution of a cryptic Salmonella typhi plasmid pHCM2. Plasmid, 2002, 47(3):159–171.

42. Crump JA, Luby SP, Mintz ED. The global burden of typhoid fever. Bulletin of the World Health Organization, 2004, 82(5):346–353.

43. Gilman R et al. Relative efficacy of blood, urine, rectal swab, bone-marrow, and rose-spot cultures for recovery of Salmonella typhi in typhoid fever. Lancet, 1975, 1:1211–1213.

44. Avendano A et al. Duodenal string cultures; Practicality and sensitivity for diagnosing enteric fever in children. The Journal of Infectious Diseases, 1986, 53:359–362.

45. Ferreccio C et al. Benign bacteraemia caused by Salmonella typhi and paratyphi in children younger than 2 years. The Journal of Pediatrics, 1984, 104:899–901.

46. Lepage P et al. Community-acquired bacteraemia in African children. Lancet, 1987, 1(8548):1458–1461.

47. Sur D et al. The malaria and typhoid fever burden in the slums of Kolkata, India: data from a prospective community-based study. Transactions of the Royal Society of Tropical Medicine and Hygiene, 2006, 100:725–733.

48. Levine MM et al. Large-scale field trial of Ty21a live oral typhoid vaccine in enteric-coated capsule formulation. Lancet, 1987, 1:1049–1052.

49. Levine MM et al. Comparison of enteric-coated capsules and liquid formulation of Ty21a typhoid vaccine in randomized controlled field trial. Lancet, 1990, 336:891–894.

Page 44: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines34

50. Black RE et al. Efficacy of one or two doses of Ty21a Salmonella typhi vaccine in enteric-coated capsules in a controlled field trial. Chilean Typhoid Committee. Vaccine, 1990, 8:81–84.

51. Simanjuntak C et al. Oral immunization against typhoid fever in Indonesia with Ty21a vaccine. Lancet, 1991, 338:1055–1059.

52. Yang HH et al. Efficacy trial of Vi polysaccharide vaccine against typhoid fever in south-western China. Bulletin of the World Health Organization, 2001, 79(7):625–631.

53. Sur D et al. A cluster-randomized effectiveness trial of Vi typhoid vaccine in India. The New England Journal of Medicine, 2009, 361:335–334.

54. Mai NL et al. Persistent efficacy of Vi conjugate vaccine against typhoid fever in young children. The New England Journal of Medicine, 2003, 349(14):1390–1391.

55. Acharya VI et al. Prevention of typhoid fever in Nepal with the Vi capsular polysaccharide of Salmonella typhi. A preliminary report. The New England Journal of Medicine, 1987, 317:1101–1104.

56. Lin FYC et al. The efficacy of a Salmonella Typhi Vi conjugate vaccine in two-to-five-year-old children. The New England Journal of Medicine, 2001, 344:1263–1268.

57. Sinha A et al. Typhoid fever in children aged less than 5 years. Lancet, 1999, 354(9180):734–737.

58. Brooks WA et al. Bacteremic typhoid fever in children in an urban slum, Bangladesh. Emerging Infectious Diseases, 2005, 11(2):326–329.

59. Siddiqui FJ et al. Typhoid fever in children: some epidemiological considerations from Karachi, Pakistan. International Journal of Infectious Diseases : IJID : official publication of the International Society for Infectious Diseases, 2006, 10(3):215–222.

59. Owais A et al. Incidence of typhoid bacteremia in infants and young children in southern coastal Pakistan. The Pediatric Infectious Disease Journal, 2010, 29:1035–1039.

60. Mahle WT, Levine MM. Salmonella typhi infection in children younger than five years of age. The Pediatric Infectious Disease Journal, 1993, 12:627–631.

61. Berkley JA et al. Bacteremia among children admitted to a rural hospital in Kenya. The New England Journal of Medicine, 2005, 352(1):39–47.

62. Enwere G et al. Epidemiologic and clinical characteristics of community-acquired invasive bacterial infections in children aged 2–29 months in The Gambia. The Pediatric Infectious Disease Journal, 2006, 25(8):700–705.

63. Walsh AL et al. Bacteremia in febrile Malawian children: clinical and microbiologic features. The Pediatric Infectious Disease Journal, 2000, 19(4):312–318.

64. Levy H et al. A PCR method to identify Salmonella enterica serovars Typhi, Paratyphi A and Paratyphi B among Salmonella isolates from the blood of patients with clinical enteric fever. Journal of Clinical Microbiology, 2008, 46(5):1861–1866.

Page 45: Salmonella enterica serovar Typhi (typhoid) vaccines

35

65. Tennant SM et al. Multiplex PCR method to identify Salmonella enterica serovars Typhimurium, Enteritidis and Dublin among Salmonella isolated from the blood of febrile patients. PLoS Neglected Tropical Diseases, 2010, 4(2):e621.doi:10.1371/ journal.pntd.0000621.

66. Meltzer E et al. Epidemiology and clinical aspects of enteric fever in Israel. The American Journal of Tropical Medicine and Hygiene, 2006, 74(4):540–545.

67. Crump JA, Mintz ED. Global trends in typhoid and paratyphoid fever. Clinical Infectious Diseases : an official publication of the Infectious Diseases Society of America, 2010, 50(2):241–246.

68. Ekdahl K, de JB, Andersson Y. Risk of travel-associated typhoid and paratyphoid fevers in various regions. Journal of Travel Medicine, 2005, 12(4):197–204.

69. Angell SY, Cetron MS. Health disparities among travelers visiting friends and relatives abroad. Annals of Internal Medicine, 2005, 142(1):67–72.

70. Cooke FJ et al. Cases of typhoid fever imported into England, Scotland and Wales (2000–2003). Transactions of the Royal Society of Tropical Medicine and Hygiene, 2007, 101(4):398–404.

71. Lin FY et al. Restaurant-associated outbreak of typhoid fever in Maryland: identification of carrier facilitated by measurement of serum Vi antibodies. Journal of Clinical Microbiology, 1988, 26(6):1194–1197.

72. Yoon J, Segal-Maurer S, Rahal JJ. An outbreak of domestically acquired typhoid fever in Queens, NY. Archives of Internal Medicine, 2004, 164(5):565–567.

73. Feldman RE et al. Epidemiology of Salmonella typhi infection in a migrant labor camp in Dade County, Florida. The Journal of Infectious Diseases, 1974, 130:334–342.

74. Neumann AK, Dinbergs IK. Typhoid and old lace, a grandmother carrier. Clinical Pediatrics, 1973, 12(5):288–290.

75. Levine MM, Black RE, Lanata C. Precise estimation of the numbers of chronic carriers of Salmonella typhi in Santiago, Chile, an endemic area. The Journal of Infectious Diseases, 1982, 146:724–726.

76. Levine MM et al. Precise estimation of the numbers of chronic carriers of Salmonella typhi in Santiago, Chile, an endemic area. The Journal of Infectious Diseases, 1982, 146:724–726.

77. Khatri NS et al. Gallbladder carriage of Salmonella paratyphi A may be an important factor in the increasing incidence of this infection in South Asia. Annals of Internal Medicine, 2009, 150(8):567–568.

78. Holt KE et al. Multidrug-resistant Salmonella enterica serovar paratyphi A harbors IncHI1 plasmids similar to those found in serovar typhi. Journal of Bacteriology, 2007, 189(11):4257–4264.

79. Salas M, Angulo O, Villegus J. Patologia de la fiebre tifoida en los ninos [Pathology of typhoid fever in children]. Boletín médico del Hospital Infantil de México, 1960, 17:63–68.

80. Hornick RB et al. Typhoid fever; pathogenesis and immunologic control. The New England Journal of Medicine, 1970, 283:686–691, 739–746.

Page 46: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines36

81. Gaines S et al. Studies on infection and immunity in experimental typhoid fever. VII. The distribution of Salmonella typhi in chimpanzee tissue following oral challenge and the relationship between the numbers of bacilli and morphologic lesions. The Journal of Infectious Diseases, 1968, 118:293–306.

82. Carter P, Collins R. The route of enteric infection in normal mice. The Journal of Experimental Medicine, 1974, 139:1189–1203.

83. Kohbata S, Yokoyama H, Yabuchi E. Cytopathogenic effect of Salmonella typhi GIFU 10007 on M cells of murine ileal Peyer’s patches in ligated ileal loops: an ultrastructural study. Microbiology and Immunology, 1986, 30:1225–1237.

84. Takeuchi A. Electron microscope studies of experimental Salmonella infection. I. Penetrations into the intestinal epithelium by Salmonella typhimurium. The American Journal of Pathology, 1967, 50:109–136.

85. Kops SK et al. Migration of Salmonella typhi through intestinal epithelial monolayers: an in vitro study. Microbiology and Immunology, 1996, 40(11):799–811.

86. Gilman RH et al. Anatomical and immunological responses of rabbit gallbladders to bacterial infections. Infection and Immunity, 1982, 36(1):407–416.

87. Hoffman S et al. Duodenal string-capsule culture compared with bone-marrow, blood and rectal swab cultures for diagnosing typhoid and paratyphoid fever. The Journal of Infectious Diseases, 1984, 149:157–161.

88. Benavente L et al. Diagnosis of typhoid fever using a string capsule device. Transactions of the Royal Society of Tropical Medicine and Hygiene, 1984, 78:404–406.

89. Ames W, Robins M. Age and sex as factors in the development of the typhoid carrier state and a method of estimating carrier prelavence. American Journal of Public Health, 1943, 33:221–230.

90. Merselis JG Jr. et al. Quantitative bacteriology of the typhoid carrier state. The American Journal of Tropical Medicine and Hygiene, 1964, 13:425–429.

91. Saha MR et al. Emergence of Salmonella typhi Vi antigen-negative strains in an epidemic of multidrug-resistant typhoid fever cases in Calcutta, India. The National Medical Journal of India, 2000, 13(3):164.

92. Standfast AFB. Experiments with Vi-negative strains of Salmonella typhi. Bulletin of the World Health Organization, 1960, 23:47–52.

93. Arya SC, Agarwal N. A cluster-randomized effectiveness trial of Vi typhoid vaccine in India. The New England Journal of Medicine, 2009, 361(22):2191–2193.

94. Mehta G, Arya SC. Capsular Vi polysaccharide antigen in Salmonella enterica serovar typhi isolates. Journal of Clinical Microbiology, 2002, 40(3):1127–1128.

95. Baker S 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. Journal of Clinical Microbiology, 2005, 43(9):4418–4425.

96. Wain J et al. Vi antigen expression in Salmonella enterica serovar Typhi clinical isolates from Pakistan. Journal of Clinical Microbiology, 2005, 43(3):1158–1165.

Page 47: Salmonella enterica serovar Typhi (typhoid) vaccines

37

97. Ali A et al. Multiplex PCR for differential diagnosis of emerging typhoidal pathogens directly from blood samples. Epidemiology and Infection, 2009, 137(1):102–107.

98. Levine MM et al. Diagnostic value of the Widal test in areas endemic for typhoid fever. The American Journal of Tropical Medicine and Hygiene, 1978, 27:795–800.

99. Balakrishna-Sarma VN et al. Development of immune response during typhoid fever in man. Clinical and Experimental Immunology, 1977, 28:35–39.

100. Mabel TJ, Paniker CKJ. The role of cell-mediated immunity in typhoid. Asian Journal of Infectious Diseases, 1979, 3:69–75.

101. Nyerges G, Ferencz A, Funk O. Development of specific cellular immunoreactivity in typhoid fever. Acta Microbiologica Academiae Scientiarum Hungaricae, 1979, 26:321–324.

102. Rajagopalan P, Kumar R, Malaviva N. Immunological studies in typhoid fever. II. Cell-mediated immune responses and lymphocyte subpopulations in patients with typhoid fever. Clinical and Experimental Immunology, 1982, 47:269–274.

103. Sarasombath S et al. Systemic and intestinal immunities after natural typhoid infection. Journal of Clinical Microbiology, 1987, 15:1088–1093.

104. Thevanesam V et al. Cell-mediated and humoral immune responses in human typhoid fever. Tropical and Geographical Medicine, 1982, 34:13–17.

105. Ortiz V et al. Immunoblot detection of class-specific humoral immune response to outer membrane proteins isolated from Salmonella typhi in humans with typhoid fever. Journal of Clinical Microbiology, 1989, 27(7):1640–1645.

106. Losonsky GA et al. Development and evaluation of an enzyme-linked immunosorbent assay for serum Vi antibodies for detection of chronic Salmonella typhi carriers. Journal of Clinical Microbiology, 1987, 25:2266–2269.

107. Lanata CF et al. Vi serology in detection of chronic Salmonella typhi carriers in an endemic area. Lancet, 1983, 2:441–443.

108. Mirza NB et al. Typhim Vi vaccine against typhoid fever: a clinical trial in Kenya. East African Medical Journal, 1995, 72(3):162–164.

109. Nolan CM et al. Evaluation of a new assay for Vi antibody in chronic carriers of Salmonella typhi. Journal of Clinical Microbiology, 1980, 12:22–26.

110. Ferreccio C et al. Detección de portadores cronicos de S. typhi: metodo practico aplicado a manipuladores de alimentos del centro de Santiago [Detection of chronic S. Typhi carriers: a practical method applied to food handlers in downtown Santiago]. Revista Médica de Chile, 1990, 118:33–37.

111. Lanata CF et al. Detection of Salmonella typhi carriers in food handlers by Vi serology in Lima, Peru. Bulletin of the Pan American Health Organization, 1990, 24(2):177–182.

112. Engleberg NC et al. Identification of a carrier by using Vi enzyme-linked immunosorbent assay serology in an outbreak of typhoid fever on an Indian reservation. Journal of Clinical Microbiology, 1983, 18(6):1320–1322.

113. Pulickal AS et al. Kinetics of the natural, humoral immune response to Salmonella enterica serovar Typhi in Kathmandu, Nepal. Clinical and Vaccine Immunology : CVI, 2009, 16(10):1413–1419.

Page 48: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines38

114. Levine MM et al. Interventions to control endemic typhoid fever: Field studies in Santiago, Chile. Washington, DC, PAHO Copublication Series No. 1, 1986:37–53.

115. Murphy JR et al. Characteristics of humoral and cellular immunity to Salmonella typhi in residents of typhoid-endemic and typhoid-free regions. The Journal of Infectious Diseases, 1987, 156:1005–1009.

116. Murphy JR et al. Immunity to Salmonella typhi: considerations relevant to measurement of cellular immunity in typhoid-endemic regions. Clinical and Experimental Immunology, 1989, 75:228–233.

117. Typhoid vaccines: WHO position paper. Weekly Epidemiological Record, 2008, 83(6):49–59.

118. Levine MM. Typhoid fever vaccines. In: Plotkin SA, Orenstein WA, eds. Vaccines, 3rd ed. Philadelphia, WB Saunders, 1999:781–814.

119. Ivanoff B, Levine MM, Lambert PH. Vaccination against typhoid fever: present status. Bulletin of the World Health Organization , 1994, 72(6):957–971.

120. Levine MM et al. Progress in vaccines against typhoid fever. Reviews of Infectious Diseases, 1989, 11(Suppl. 3):S552–S567.

121. Yugoslav Typhoid Commission. A controlled field trial of the effectiveness of acetone-dried and inactivated and heat-phenol-inactivated typhoid vaccines in Yugoslavia. Bulletin of the World Health Organization, 1964, 30:623–630.

122. Hejfec LB et al. A controlled field trial and laboratory study of five typhoid vaccines in the USSR. Bulletin of the World Health Organization, 1966, 34:321–339.

123. Polish Typhoid Committee. Controlled field trial and laboratory studies on the effectiveness of typhoid vaccines in Poland 1961–64. Bulletin of the World Health Organization, 1966, 34:211–222.

124. Ashcroft MT et al. A seven-year field trial of two typhoid vaccines in Guiana. Lancet, 1967, 2:1056–1060.

125. Ashcroft MT, Morrison-Ritchie J, Nicholson CC. Controlled field trial in British Guyana schoolchildren of heat-killed-phenolized and acetone-killed lyophilized typhoid vaccines. American Journal of Hygiene, 1964, 79:196–206.

126. Levine MM. Use of vaccines for the prevention of typhoid fever. Indian Pediatrics, 2003, 40(11):1029–1034.

127. Ashcroft MT et al. Antibody responses to vaccination of British Guiana school children with heat-killed-phenolized and acetone-killed lyophiloized vaccines. American Journal of Hygiene, 1964, 80:221–228.

128. Edsall G et al. Laboratory tests of typhoid vaccines within a controlled field study. Bulletin of the World Health Organization, 1959, 20:1017–1032.

129. Wong KH, Feeley JC, Pittman M. Effect of Vi-degrading enzyme on potency of typhoid vaccines in mice. The Journal of Infectious Diseases, 1972, 125:360–366.

130. Germanier R, Furer E. Isolation and characterization of gal E mutant Ty21a of Salmonella typhi: a candidate strain for a live oral typhoid vaccine. The Journal of Infectious Diseases, 1975, 141:553–558.

Page 49: Salmonella enterica serovar Typhi (typhoid) vaccines

39

131. Kopecko DJ et al. Genetic stability of vaccine strain Salmonella Typhi Ty21a over 25 years. International Journal of Medical Microbiology : IJMM, 2009, 299(4):233–246.

132. Black RE et al. Immunogenicity of Ty21a attenuated Salmonella typhi given with sodium bicarbonate or in enteric-coated capsules. Developments in Biological Standardization, 1982, 53:9–14.

133. Wahdan MH et al. A controlled field trial of live Salmonella typhi strain Ty21a oral vaccine against typhoid: three year results. The Journal of Infectious Diseases, 1982, 145:292–296.

134. Ferreccio C et al. Comparative efficacy of two, three, or four doses of Ty21a live oral typhoid vaccine in enteric-coated capsules: a field trial in an endemic area. The Journal of Infectious Diseases, 1989, 159:766–769.

135. Levine MM et al. Duration of efficacy of ty21a, attenuated salmonella typhi live oral vaccine. Vaccine, 1999, 17(Suppl. 2):S22–S27.

136. Wong KH et al. Vi antigen from Salmonella typhosa and immunity against typhoid fever. I. Isolation and immunologic properties in animals. Infection and Immunity, 1974, 9:348–353.

137. Tacket CO et al. Safety and immunogenicity of two Salmonella typhi Vi capsular polysaccharide vaccines. The Journal of Infectious Diseases, 1986, 154:342–345.

138. Klugman K et al. Protective activity of Vi polysaccharide vaccine against typhoid fever. Lancet, 1987, 2:1165–1169.

139. Keitel WA et al. Clinical and serological responses following primary and booster immunization with Salmonella typhi Vi capsular polysaccharide vaccines. Vaccine, 1994, 12(3):195–199.

140. Tacket CO, Levine MM, Robbins JB. Persistence of antibody titres three years after vaccination with Vi polysaccharide vaccine against typhoid fever. Vaccine, 1988, 6:307–308.

141. Froeschle JE, Decker MD. Duration of Vi antibodies in participants vaccinated with Typhim Vi (Typhoid Vi polysaccharide vaccine) in an area not endemic for typhoid fever. Vaccine, 2009, 28:1451–1453.

142. Overbosch D et al. Combined typhoid fever and hepatitis A vaccine: comparison of immunogenicity and safety to concomitant monovalent vaccine over 3 years. Journal of Travel Medicine, 2005, 12(6):319–326.

143. Klugman KP et al. Immunogenicity, efficacy and serological correlate of protection of Salmonella typhi Vi capsular polysaccharide vaccine three years after immunization. Vaccine, 1996, 14(5):435–438.

144. Michel R et al. Outbreak of typhoid fever in vaccinated members of the French armed forces in the Ivory Coast. European Journal of Epidemiology, 2005, 20(7):635–642.

145. Yang HH et al. An outbreak of typhoid fever, Xing-An County, People’s Republic of China, 1999: estimation of the field effectiveness of Vi polysaccharide typhoid vaccine. The Journal of Infectious Diseases, 2001, 183(12):1775–1780.

146. Viret JF et al. Mucosal and systemic immune responses in humans after primary and booster immunizations with orally administered invasive and noninvasive live attenuated bacteria. Infection and Immunity, 1999, 67(7):3680–3685.

Page 50: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines40

147. Forrest BD et al. The human humoral immune response to Salmonella typhi Ty21a. The Journal of Infectious Diseases, 1991, 163:336–345.

148. D’Amelio R et al. Comparative analysis of immunological responses to oral (Ty21a) and parenteral (TAB) typhoid vaccines. Infection and Immunity, 1988, 56:2731–2735.

149. Cancellieri V, Fara GM. Demonstration of specific IgA in human feces after immunization with line Ty21a Salmonella typhi vaccine. The Journal of Infectious Diseases, 1985, 151:482–484.

150. Nisini R et al. Clinical and immunological response to typhoid vaccination with parenteral or oral vaccines in two groups of 30 recruits. Vaccine, 1993, 11(5):582–586.

151. Kantele A. Antibody-secreting cells in the evaluation of the immunogenicity of an oral vaccine. Vaccine, 1990, 8:321–326.

152. Kantele A, Makela PH. Different profiles of the human immune response to primary and secondary immunization with an oral Salmonella typhi Ty21a vaccine. Vaccine, 1991, 9:423–427.

153. Kantele A, Arvilommi H, Jokinen I. Specific immunoglobulin-secreting human blood cells after peroral vaccination against Salmonella typhi. The Journal of Infectious Diseases, 1986, 153:1126–1131.

154. Kantele A et al. Comparison of the human immune response to live oral, killed oral or killed parenteral Salmonella typhi Ty21a vaccines. Microbial Pathogenesis, 1991, 10:117–126.

155. Kantele A et al. Active immunity is seen as a reduction in the cell response to oral live vaccine. Vaccine, 1991, 9:428–431.

156. Kantele A et al. Homing potentials of circulating lymphocytes in humans depend on the site of activation: oral, but not parenteral, typhoid vaccination induces circulating antibody-secreting cells that all bear homing receptors directing them to the gut. Journal of Immunology (Baltimore, Md. : 1950), 1997, 158(2):574–579.

157. Kantele A et al. Differences in immune responses induced by oral and rectal immunizations with Salmonella typhi Ty21a: evidence for compartmentalization within the common mucosal immune system in humans. Infection and Immunity, 1998, 66(12):5630–5635.

158. Kantele A et al. Unique characteristics of the intestinal immune system as an inductive site after antigen reencounter. The Journal of Infectious Diseases, 2005, 191(2):312–317.

159. Salerno-Goncalves R, Pasetti MF, Sztein MB. Characterization of CD8(+) effector T cell responses in volunteers immunized with Salmonella enterica serovar Typhi strain Ty21a typhoid vaccine. Journal of Immunology (Baltimore, Md. : 1950), 2002, 169(4):2196–2203.

160. Sztein MB. Cell-mediated immunity and antibody responses elicited by attenuated Salmonella enterica Serovar Typhi strains used as live oral vaccines in humans. Clinical Infectious Diseases : an official publication of the Infectious Diseases Society of America, 2007, 45(Suppl. 1):S15–S19.

Page 51: Salmonella enterica serovar Typhi (typhoid) vaccines

41

161. Salerno-Goncalves R, Wahid R, Sztein MB. Immunization of volunteers with Salmonella enterica serovar Typhi strain Ty21a elicits the oligoclonal expansion of CD8+ T cells with predominant Vbeta repertoires. Infection and Immunity, 2005, 73(6):3521–3530.

162. Esser MT et al. Memory T cells and vaccines. Vaccine, 2003, 21(5–6):419–430.

163. Levine MM, Sztein MB. Vaccine development strategies for improving immunization: the role of modern immunology. Nature Immunology, 2004, 5(5):460–464.

164. Lundin BS, Johansson C, Svennerholm AM. Oral immunization with a Salmonella enterica serovar Typhi vaccine induces specific circulating mucosa-homing CD4(+) and CD8(+) T cells in humans. Infection and Immunity, 2002, 70(10):5622–5627.

165. Kilhamn J et al. T- and B-cell immune responses of patients who had undergone colectomies to oral administration of Salmonella enterica serovar Typhi Ty21a vaccine. Clinical and Diagnostic Laboratory Immunology, 2003, 10(3):426–430.

166. Tacket CO et al. Phase 2 clinical trial of attenuated Salmonella enterica serovar Typhi oral live vector vaccine CVD 908-htrA in U.S. volunteers. Infection and Immunity, 2000, 68(3):1196–1201.

167. Salerno-Goncalves R et al. Concomitant induction of CD4(+) and CD8(+) T cell responses in volunteers immunized with Salmonella enterica serovar Typhi strain CVD 908-htrA. Journal of Immunology (Baltimore, Md. : 1950), 2003, 170(5):2734–2741.

168. Sztein MB et al. Cytokine production patterns and lymphoproliferative responses in volunteers orally immunized with attenuated vaccine strains of Salmonella typhi. The Journal of Infectious Diseases, 1994, 170:1508–1517.

169. Tagliabue A et al. IgA-driven T-Cell-mediated antibacterial immunity in man after live oral Ty21a vaccine. Journal of Immunology (Baltimore, Md. : 1950), 1986, 137:1504–1510.

170. Tagliabue A et al. Cellular immunity against Salmonella typhi after live oral vaccines. Clinical and Experimental Immunology, 1985, 52:242–247.

171. Plotkin SA, Bouveret-Le Cam N. A new typhoid vaccine composed of the Vi capsular polysaccharide. Archives of Internal Medicine, 1995, 155:2293–2299.

172. Szu SC et al. Laboratory and preliminary clinical characterization of Vi capsular polysaccharide-protein conjugate vaccines. Infection and Immunity, 1994, 62:4440–4444.

173. Cordero-Yap L et al. Evaluation of a new Vi polysaccharide typhoid vaccine in children aged 2–5 years. BioDrugs : Clinical Immunotherapeutics, Biopharmaceuticals and Gene Therapy, 2001, 15(Suppl. 1):S27.

174. Pelser HH. Reactogenicity and immunogenicity of a single dose of typhoid Vi polysaccharide vaccine in children aged between 4 and 14 years. BioDrugs : Clinical Immunotherapeutics, Biopharmaceuticals and Gene Therapy, 2001, 15(Suppl. 1):S13–S19.

Page 52: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines42

175. Ramkissoon A, Jugnundan P. Reactogenicity and immunogenicity of a single dose of a typhoid Vi polysaccharide vaccine in adolescents. BioDrugs : Clinical Immunotherapeutics, Biopharmaceuticals and Gene Therapy, 2001, 15(Suppl. 1):S21–S26.

176. Hessel L et al. Experience with Salmonella typhi Vi capsular polysaccharide vaccine. European Journal of Clinical Microbiology & Infectious Diseases : official publication of the European Society of Clinical Microbiology, 1999, 18(9):609–620.

177. Pollard AJ, Perrett KP, Beverley PC. Maintaining protection against invasive bacteria with protein-polysaccharide conjugate vaccines. Nature Reviews. Immunology, 2009, 9(3):213–220.

178. Granoff DM et al. Induction of immunologic refractoriness in adults by meningococcal C polysaccharide vaccination. The Journal of Infectious Diseases, 1998, 178(3):870–874.

179. Richmond P et al. Meningococcal C polysaccharide vaccine induces immunologic hyporesponsiveness in adults that is overcome by meningococcal C conjugate vaccine. The Journal of Infectious Diseases, 2000, 181(2):761–764.

180. Torling J et al. Revaccination with the 23-valent pneumococcal polysaccharide vaccine in middle-aged and elderly persons previously treated for pneumonia. Vaccine, 2003, 22(1):96–103.

181. Russell FM et al. Hyporesponsiveness to re-challenge dose following pneumococcal polysaccharide vaccine at 12 months of age; a randomized controlled trial. Vaccine, 2010, 28:3341–3349.

182. Zhou WZ et al. Revaccination with locally-produced Vi typhoid polysaccharide vaccine among Chinese school-aged children: safety and immunogenicity findings. The Pediatric Infectious Disease Journal, 2007, 26(11):1001–1005.

183. Brugier JC et al. Isotypes of human vaccinal antibodies to the Vi capsular polysaccharide of Salmonella typhi. International Journal of Clinical & Laboratory Research, 1993, 23(1):38–41.

184. Gilman RH et al. Evaluation of a UDP-glucose-4-epimeraseless mutant of Salmonella typhi as a live oral vaccine. The Journal of Infectious Diseases, 1977, 136:717–723.

185. Wahdan MH et al. A controlled field trial of live oral typhoid vaccine Ty21a. Bulletin of the World Health Organization, 1980, 58:469–474.

186. Levine MM et al. Ty21a live oral typhoid vaccine and prevention of paratyphoid fever caused by Salmonella enterica Serovar Paratyphi B. Clinical Infectious Diseases : an official publication of the Infectious Diseases Society of America, 2007, 45(Suppl. 1):S24–S28.

187. Cryz SJ Jr. et al. Safety and immunogenicity of Salmonella typhi Ty21a vaccine in young Thai children. Infection and Immunity, 1993, 61:1149–1151.

188. Olanratmanee T et al. Safety and immunogenicity of Salmonella typhi Ty21a liquid formulation vaccine in 4- to 6-year old Thai children. The Journal of Infectious Diseases, 1992, 166:451–452.

189. Beeching NJ et al. Comparison of two combined vaccines against typhoid fever and hepatitis A in healthy adults. Vaccine, 2004, 23(1):29–35.

Page 53: Salmonella enterica serovar Typhi (typhoid) vaccines

43

190. Acosta CJ et al. Efficacy of a locally produced, Chinese Vi polysaccharide typhoid fever vaccine during six years of follow-up. Vaccine, 2005, 23(48–49):5618–5623.

191. Khan MI et al. Lessons and implications from a mass immunization campaign in squatter settlements of Karachi, Pakistan: an experience from a cluster-randomized double-blinded controlled trial [NCT00125047]. Trials, 2006, 7(1):17.

192. Agtini MD et al. Introducing Vi polysaccharide typhoid fever vaccine to primary school children in North Jakarta, Indonesia, via an existent school-based vaccination platform. Public Health, 2006, 120(11):1081–1087.

193. Yang J et al. A mass vaccination campaign targeting adults and children to prevent typhoid fever in Hechi; expanding the use of Vi polysaccharide vaccine in southeast China: a cluster-randomized trial. BMC Public Health, 2005, 5(1):49.

194. Hindle Z 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. Infection and Immunity, 2002, 70(7):3457–3467.

195. Kirkpatrick BD et al. Evaluation of Salmonella enterica serovar Typhi (Ty2 aroC-ssaV-) M01ZH09, with a defined mutation in the Salmonella pathogenicity island 2, as a live, oral typhoid vaccine in human volunteers. Vaccine, 2006, 24(2):116–123.

196. Kirkpatrick BD et al. The novel oral typhoid vaccine M01ZH09 is well tolerated and highly immunogenic in 2 vaccine presentations. The Journal of Infectious Diseases, 2005, 192(3):360–366.

197. Hohmann EL et al. phoP/phoQ-deleted Salmonella typhi (Ty800) is a safe and immunogenic single-dose typhoid fever vaccine in volunteers. The Journal of Infectious Diseases, 1996, 173:1408–1414.

198. Tacket CO et al. Safety of live oral Salmonella typhi vaccine strains with deletions in htrA and aroC aroD and immune response in humans. Infection and Immunity, 1997, 65:452–456.

199. Wang JY et al. Constitutive expression of the Vi polysaccharide capsular antigen in attenuated Salmonella enterica serovar Typhi oral vaccine strain CVD 909. Infection and Immunity, 2000, (68):4647–4652.

200. Tacket CO et al. Immune responses to an oral typhoid vaccine strain that is modified to constitutively express Vi capsular polysaccharide. The Journal of Infectious Diseases, 2004, 190(3):565–570.

201. Wahid R et al. Oral priming with Salmonella Typhi vaccine strain CVD 909 followed by parenteral boost with the S. Typhi Vi capsular polysaccharide vaccine induces CD27+IgD-S. Typhi-specific IgA and IgG B memory cells in humans. Clinical Immunology, 2011, 138(2):187–200.

202. Kossaczka Z et al. Safety and immunogenicity of Vi conjugate vaccines for typhoid fever in adults, teenagers, and 2- to 4-year-old children in Viet Nam. Infection and Immunity, 1999, 67(11):5806–5810.

203. Garg SP. Vi conjugate typhoid vaccine. Indian Pediatrics, 2009, 46(8):736–737.

Page 54: Salmonella enterica serovar Typhi (typhoid) vaccines

The Immunological basis for immunization series - Module 20: Salmonella enterica serovar Typhi (typhoid) vaccines44

204. Podda A et al. Conjugate vaccines for enteric fever: proceedings of a meeting organized in New Delhi, India in 2009. Journal of Infection in Developing Countries, 2010, 4(6):404–411.

205. Kirkpatrick BD et al. Comparison of the antibodies in lymphocyte supernatant and antibody-secreting cell assays for measuring intestinal mucosal immune response to a novel oral typhoid vaccine (M01ZH09). Clinical and Diagnostic Laboratory Immunology, 2005, 12(9):1127–1129.

206. Lyon CE et al. In a randomized, double-blinded, placebo-controlled trial, the single oral dose typhoid vaccine, M01ZH09, is safe and immunogenic at doses up to 1.7x10(10) colony-forming units. Vaccine, 2010, 28:3602–3608.

207. Wahid R et al. Cell-mediated immune responses in humans after immunization with one or two doses of oral live attenuated typhoid vaccine CVD 909. Vaccine, 2007, 25(8):1416–1425.

208. Wahid R et al. Generation of specific effector and memory T cells with gut- and secondary lymphoid tissue-homing potential by oral attenuated CVD 909 typhoid vaccine in humans. Mucosal Immunology, 2008, 1(5):389–398.

209. Canh dG et al. Effect of dosage on immunogenicity of a Vi conjugate vaccine injected twice into 2- to 5-year-old Vietnamese children. Infection and Immunity, 2004, 72(11):6586–6588.

210. Fraser A et al. Vaccines for preventing typhoid fever (review). Cochrane Database of Systematic Reviews, 2007, (3): CD001261.DOI:10.1002/14651858. CD001261. pub2.

211. Thiem VD et al. The Vi conjugate typhoid vaccine is safe, elicits protective levels of IgG anti-Vi, and is compatible with routine infant vaccines. Clinical and Vaccine Immunology, 2011, 18(5):730–735.

Page 55: Salmonella enterica serovar Typhi (typhoid) vaccines

The World Health Organization has provided technical support to its Member States in the field of vaccine-preventable diseases since 1975. The office carrying out this function at WHO headquarters is the Department of Immunization, Vaccines and Biologicals (IVB).

IVB’s mission is the achievement of a world in which all people at risk are protected against vaccine-preventable diseases. The Department covers a range of activities including research and development, standard-setting, vaccine regulation and quality, vaccine supply and immunization financing, and immunization system strengthening.

These activities are carried out by three technical units: the Initiative for Vaccine Research; the Quality, Safety and Standards team; and the Expanded Programme on Immunization.

The Initiative for Vaccine Research guides, facilitates and provides a vision for worldwide vaccine and immunization technology research and development efforts. It focuses on current and emerging diseases of global public health importance, including pandemic influenza. Its main activities cover: i ) research and development of key candidate vaccines; ii ) implementation research to promote evidence-based decision-making on the early introduction of new vaccines; and iii ) promotion of the development, evaluation and future availability of HIV, tuberculosis and malaria vaccines.

The Quality, Safety and Standards team focuses on supporting the use of vaccines, other biological products and immunization-related equipment that meet current inter-national norms and standards of quality and safety. Activities cover: i ) setting norms and standards and establishing reference preparation materials; ii ) ensuring the use of quality vaccines and immunization equipment through prequalification activities and strengthening national regulatory authorities; and iii ) monitoring, assessing and responding to immunization safety issues of global concern.

The Expanded Programme on Immunization focuses on maximizing access to high quality immunization services, accelerating disease control and linking to other health interventions that can be delivered during immunization contacts. Activities cover: i ) immunization systems strengthening, including expansion of immunization services beyond the infant age group; ii ) accelerated control of measles and maternal and neonatal tetanus; iii ) introduction of new and underutilized vaccines; iv ) vaccine supply and immunization financing; and v ) disease surveillance and immunization coverage monitoring for tracking global progress.

The Director’s Office directs the work of these units through oversight of immunization programme policy, planning, coordination and management. It also mobilizes resources and carries out communication, advocacy and media-related work.

Family and Community Health

World Health Organization 20, Avenue Appia

CH-1211 Geneva 27 Switzerland

E-mail: [email protected] Web site: http://www.who.int/immunization/en/

Department of Immunization, Vaccines and Biologicals

Immunization, Vaccines and Biologicals

ISBN 978 92 4 150261 0