4
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The Time is Now to Control Typhoid

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The death of Prince Albert, married to Queen Victoria of the United Kingdom, in 1861 from typhoid profoundly impacted the Monarchy. The queen mourned for the rest of her life, some 40 years, highlighting the individual and familial impact of typhoid in a very public way [1]. The diagnosis was made by the prince’s physician, Sir William Jenner [2], who managed many typhoid patients and meticulously documented the prince’s symptoms, including the appearance of rose spots. As a result of municipal clean water and sewage works, typhoid has essentially disappeared from the wealthy nations of the world, with almost all cases in Europe and North America now attributed to travel to endemic regions. However, there are still thought to be nearly 12 million cases and more than 128 000 deaths each year associated with Salmonella serotype Typhi [3]. Cases occur largely in resource-poor regions of the world, sub-Saharan Africa, and South/Southeast Asia, where there is limited access to clean water and inadequate sanitation [4, 5]. In these communities, the disease continues to have the same profound impact on families as was experienced by Queen Victoria. Fortunately, with the availability of antimicrobial therapy, the mortality from typhoid has fallen from the high levels of the preantibiotic era. Recently, however, extensively drug-resistant (XDR) S. typhi variants have emerged in India, Bangladesh, the Philippines, Iraq, and Guatemala, and, importantly, have caused a large outbreak in Pakistan [6, 7], further threatening the health of these populations. Since 2008, the World Health Organization (WHO) has advocated vaccine control of typhoid [8], but uptake has been slow due to lack of a suitable vaccine for children aged <2 years. Of course, typhoid can be controlled by use of clean water and improved sanitation and hygiene, which would stop the spread of typhoid and other water- and foodborne enteric pathogens. However, we cannot wait for safe water and proper sanitation to be available for every child; too many will suffer from enteric fever if we do not act. It is time to control typhoid now. The Typhoid Vaccine Acceleration Consortium (TyVAC) [9], funded by the Bill & Melinda Gates Foundation, launched in 2017 as a partnership of the Center for Vaccine Development and Global Health at the University of Maryland School of Medicine, the Oxford Vaccine Group at the University of Oxford, and PATH, an international nonprofit organization. TyVAC’s mission is to protect children who suffer the greatest burden by accelerating the use of typhoid conjugate vaccine (TCV) in Africa and Asia. As a major component, the consortium is evaluating Typbar-TCV, the leading vaccine candidate, to provide data to support introduction of the vaccine in those regions suffering a substantial burden of disease. The studies were designed to be complementary to each other and to other efforts, including data available from the vaccine manufacturer and partners [10]. This supplement highlights some of the current and future work of TyVAC and its associates. The approach is similar to that used to help drive global introductions of other vaccines, as summarized by Jamka et al [11]. With the March 2018 WHO-revised typhoid position paper and the current Gavi, the Vaccine Alliance, funding window for TCV introduction, the supplement provides key data on vaccine efficacy and immunogenicity in sub-Saharan Africa and Southeast Asia. The supplement includes methods papers for the 4 TyVAC sites and articles on health economics, modeling, demand forecasting, global incidence, public engagement and lessons learned, antimicrobial resistance, decision-making, data management, and accelerating TCV introduction. The supplement includes a systematic review of data on the global burden of typhoid [12] and a series of important articles on new burden data from Myanmar, Democratic Republic of Congo, Bangladesh, and Tanzania [13–16]. As identified in these articles, and others published recently [17, 18], typhoid is far more widespread than might be appreciated from the microbiological literature. Surveillance for the disease requires microbiological culture facilities since the clinical syndrome is largely indistinguishable from other illnesses that present with fever, making syndromic surveillance unhelpful. Unfortunately, the regions with the highest rates of typhoid often have the least developed laboratory facilities, emphasizing the importance of these studies in supporting vaccine deployment and control of the disease. Lack of country- and regional-level surveillance data is one of the main challenges for decision-makers. Vaccine efficacy of 54%–87% was recently demonstrated using a controlled human infection model of typhoid [19], which strongly supports the likely efficacy of Typbar-TCV, manufactured by Bharat Biotech. The study was run using human adult volunteers in Oxford, United Kingdom, who had not lived in an endemic area but received 10 000 bacteria as the infection challenge after neutralization of gastric acid. It is unclear how efficacy in the model relates to performance of this TCV in children in an endemic region where there are currently no field efficacy data. Liu et al [20] discuss the design and analysis of seroefficacy studies for TCVs. TCV was licensed and recommended by WHO based on immunogenicity data and a human controlled infection model in nonendemic adults, but the lack of field data at this time undermines confidence at the country level of the potential impact. Given limitations on health budgets, many countries will need more information before deciding to introduce a vaccine, particularly data are needed on impact, how programs might be structured, and how the vaccine performs in different transmission settings. As part of the TyVAC program, we are undertaking individually randomized clinical trials in Malawi and Nepal and a cluster-randomized trial in Bangladesh. The protocols for these studies are summarized in this supplement, in addition to an immunogenicity trial protocol from Burkina Faso [21–24]. In the efficacy trials, more than 85 000 children from age 9 months to age 15 years were vaccinated during late 2017 through the first half of 2018. Given that duration of immunity is critically important to country-level decisions on vaccine introduction, these children will be followed for at least 2 years using enhanced passive microbiological surveillance (blood cultures), with vaccine efficacy measures available by 2020. The control vaccine in these studies is either a capsular group A meningococcal vaccine (Nepal and Malawi), as this is the main agent causing meningococcal disease in these regions, or Japanese encephalitis vaccine in Bangladesh, since this disease is important in rural areas of that country. Given the importance of typhoid in the population, control-arm children in Nepal and Bangladesh will receive TCV at the end of the study. In Malawi, the epidemiology and burden are less certain; therefore, the decision on whether or not to provide TCV for children in the control group will be made in concert with Malawian health officials once data are available. Other typhoid vaccines were not considered for control vaccines because they are not licensed for the youngest children, do not confer long-term protection, and, therefore, are less suitable for routine country introduction. Another important outstanding question is the potential role of TCV in reducing transmission of typhoid in the field and thus inducing herd protection, as was recently indicated as possible in studies using the human challenge model [25]. The TyVAC field trials will directly address this through the cluster-randomized trial in Bangladesh. Taken together, the data from the TyVAC trials will provide valuable information to inform vaccine deployment in Africa and Asia, and we anticipate reassurance on field safety will be important for national immunization technical advisory groups (NITAGs) and health ministers, when considering introduction of new TCVs. The process of obtaining funding for introductions, especially through international donors, is slow. The TyVAC trials will provide important data ahead of possible introductions in most countries [26]. Important components of vaccine field studies that are rarely described in the scientific literature are logistical elements of study delivery and the role and implementation of public engagement. Two articles in this supplement deal with these issues and provide a high-level view that we anticipate will be useful for deployment of clinical studies in the future [27, 28]. As mentioned above, antimicrobial resistance is a major threat to the health of populations in which Salmonella infections are prevalent as these bacteria readily pick up resistance genes. Multidrug resistance (MDR, defined as resistance to the first-line antibiotics ampicillin, cotrimoxazole, and chloramphenicol) was recognized several decades ago and became widespread among the agents of enteric fever. However, there has been a decline in resistance over the past 10 years, especially in South Asia, as antibiotic pressure was reduced after these drugs became redundant in typhoid treatment [29]. More recent use of fluoroquinolones has driven very widespread acquisition of resistance to this class of antibiotic; currently severe cases often must be treated with cephalosporins and milder cases with azithromycin. A recent outbreak of XDR typhoid (defined as MDR plus resistance to fluoroquinolones and cephalosporins) has been identified in Pakistan [7], considerably reducing treatment options. In this supplement, we provide insight into antimicrobial resistance (AMR) from genomic analyses of typhoid strains [30] and country-specific data on AMR in the enteric fever burden studies. Introduction of new TCVs will impact AMR infections in the same way as susceptible typhoid, and our modeling article summarizes predicted vaccine impact [31]. In October 2017, TyVAC provided evidence for WHO’s Strategic Advisory Group of Experts (SAGE) on Immunization that recommended the use of new TCVs. Also, WHO supplied information for Gavi, the Vaccine Alliance, to support Gavi’s planning for TCV financing, with $85 million provided for vaccine deployment as outlined by Jamka et al [11]. An important component of country decision-making for new vaccines, especially for those graduating from Gavi support for new vaccine programs in the next 5 years, is the economics of vaccine introduction. In this supplement, we include a review of typhoid vaccine economic studies and summarize plans for further work of the TyVAC consortium [32]. For manufacturers and vaccine financiers (Gavi), an accurate demand forecast is important to ensure supply meets demand and funds are available to purchase products. Reducing uncertainty is important for country-level decisions, as highlighted by supply shortages for other vaccines in recent years, which have caused difficulties for national programs. The demand-forecasting article describes the model built to estimate TCV demand [33]. Finally, in this supplement, we discuss the importance of communications about typhoid, which must involve international, national, and local-level stakeholders. This ensures that SAGE recommendations are translated into appropriate decisions by the NITAG, with government support, and are clear and understood by local public health immunization officials and frontline staff so that new programs are introduced rapidly and with clarity for the benefit of the population [34]. The articles in this supplement show the development of high-quality evidence by TyVAC to inform decision-making on TCV introduction. Through the data produced and the advocacy of TyVAC, we are tasked with improving health among some of the most vulnerable populations in the world through introduction of TCVs where they are most needed to drive substantial and clinically meaningful reductions in typhoid burden. There should be no more families to mourn like Queen Victoria did. It is time to control typhoid.

          Related collections

          Most cited references25

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Emergence of an Extensively Drug-Resistant Salmonella enterica Serovar Typhi Clone Harboring a Promiscuous Plasmid Encoding Resistance to Fluoroquinolones and Third-Generation Cephalosporins

          ABSTRACT Antibiotic resistance is a major problem in Salmonella enterica serovar Typhi, the causative agent of typhoid. Multidrug-resistant (MDR) isolates are prevalent in parts of Asia and Africa and are often associated with the dominant H58 haplotype. Reduced susceptibility to fluoroquinolones is also widespread, and sporadic cases of resistance to third-generation cephalosporins or azithromycin have also been reported. Here, we report the first large-scale emergence and spread of a novel S. Typhi clone harboring resistance to three first-line drugs (chloramphenicol, ampicillin, and trimethoprim-sulfamethoxazole) as well as fluoroquinolones and third-generation cephalosporins in Sindh, Pakistan, which we classify as extensively drug resistant (XDR). Over 300 XDR typhoid cases have emerged in Sindh, Pakistan, since November 2016. Additionally, a single case of travel-associated XDR typhoid has recently been identified in the United Kingdom. Whole-genome sequencing of over 80 of the XDR isolates revealed remarkable genetic clonality and sequence conservation, identified a large number of resistance determinants, and showed that these isolates were of haplotype H58. The XDR S. Typhi clone encodes a chromosomally located resistance region and harbors a plasmid encoding additional resistance elements, including the bla CTX-M-15 extended-spectrum β-lactamase, and carrying the qnrS fluoroquinolone resistance gene. This antibiotic resistance-associated IncY plasmid exhibited high sequence identity to plasmids found in other enteric bacteria isolated from widely distributed geographic locations. This study highlights three concerning problems: the receding antibiotic arsenal for typhoid treatment, the ability of S. Typhi to transform from MDR to XDR in a single step by acquisition of a plasmid, and the ability of XDR clones to spread globally.
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            Typhoid vaccines: WHO position paper.

            (2008)
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Laboratory and molecular surveillance of paediatric typhoidal Salmonella in Nepal: Antimicrobial resistance and implications for vaccine policy

              Background Children are substantially affected by enteric fever in most settings with a high burden of the disease, including Nepal. However pathogen population structure and transmission dynamics are poorly delineated in young children, the proposed target group for immunization programs. Here we present whole genome sequencing and antimicrobial susceptibility data on 198 S. Typhi and 66 S. Paratyphi A isolated from children aged 2 months to 15 years of age during blood culture surveillance at Patan Hospital, Nepal, 2008–2016. Principal findings S. Typhi was the dominant agent and comprised several distinct genotypes, dominated by 4.3.1 (H58). The heterogeneity of genotypes in children under five was reduced compared to data from 2005–2006, attributable to ongoing clonal expansion of H58. Most isolates (86%) were non-susceptible to fluoroquinolones, associated mainly with S. Typhi H58 lineage II and S. Paratyphi A harbouring mutations in the quinolone resistance-determining region (QRDR); non-susceptible strains from these groups accounted for 50% and 25% of all isolates. Multi-drug resistance (MDR) was rare (3.5% of S. Typhi, 0 S. Paratyphi A) and restricted to chromosomal insertions of resistance genes in H58 lineage I strains. Temporal analyses revealed a shift in dominance from H58 Lineage I to H58 Lineage II, with the latter being significantly more common after 2010. Comparison to global data sets showed the local S. Typhi and S. Paratyphi A strains had close genetic relatives in other South Asian countries, indicating regional strain circulation. Multiple imports from India of ciprofloxacin-resistant H58 lineage II strains were identified, but these were rare and showed no evidence of clonal replacement of local S. Typhi. Significance These data indicate that enteric fever in Nepal continues to be a major public health issue with ongoing inter- and intra-country transmission, and highlights the need for regional coordination of intervention strategies. The absence of a S. Paratyphi A vaccine is cause for concern, given its prevalence as a fluoroquinolone resistant enteric fever agent in this setting.
                Bookmark

                Author and article information

                Journal
                Clin Infect Dis
                Clin. Infect. Dis
                cid
                Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America
                Oxford University Press (US )
                1058-4838
                1537-6591
                15 March 2019
                07 March 2019
                07 March 2019
                : 68
                : Suppl 2 , Typhoid fever and the introduction of the typhoid conjugate vaccine in Africa and Asia
                : S47-S49
                Affiliations
                [1 ]Oxford Vaccine Group, University of Oxford, and the National Institute for Health Research Oxford Biomedical Research Centre, United Kingdom
                [2 ]Center for Vaccine Innovation and Access, PATH, Seattle, Washington
                [3 ]Center for Vaccine Development and Global Health at the University of Maryland School of Medicine, Baltimore, MD
                Author notes
                Correspondence: K. M. Neuzil, Center for Vaccine Development and Global Health at the University of Maryland School of Medicine, 685 West Baltimore St. HSF1-Room 480, Baltimore, MD 21201 ( kneuzil@ 123456som.umaryland.edu ).
                Article
                ciy1115
                10.1093/cid/ciy1115
                6405270
                30845327
                ce15763d-a48d-4e38-88bf-737aeccae84a
                © The Author(s) 2019. Published by Oxford University Press for the Infectious Diseases Society of America.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                Page count
                Pages: 3
                Funding
                Funded by: Bill and Melinda Gates Foundation 10.13039/100000865
                Award ID: OPP1151153
                Categories
                Supplement Articles

                Infectious disease & Microbiology
                tyvac,typhoid,vaccine
                Infectious disease & Microbiology
                tyvac, typhoid, vaccine

                Comments

                Comment on this article