1
|
Nguyen AT, McSorley SJ. Fighting the enemy within: Systemic immune defense against mucosal Salmonella infection. Immunol Lett 2024; 270:106930. [PMID: 39343314 DOI: 10.1016/j.imlet.2024.106930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024]
Abstract
Salmonella infection remains a persistent global health threat, as different serovars induce a range of clinical disease, depending upon bacterial virulence and host susceptibility. While some Salmonella serovars induce gastroenteritis in healthy individuals, others can cause more serious systemic enteric fever or invasive nontyphoidal Salmonellosis. The rise of antibiotic resistance, coupled with the absence of effective vaccines for most serovars, perpetuates the spread of Salmonella in endemic regions. A detailed mechanistic understanding of immunity to Salmonella infections has been aided by the availability of mouse models that have served as a valuable tool for understanding host-pathogen interactions under controlled laboratory conditions. These mouse studies have delineated the processes by which early inflammation is triggered after infection, how adaptive immunity is initiated in lymphoid tissues, and the contribution of lymphocyte memory responses to resistance. While recent progress has been made in vaccine development for some causes of enteric fever, deeper understanding of Salmonella-specific immune memory might allow the formation of new vaccines for all serovars. This review will provide a summary of our understanding of vaccination and protective immunity to Salmonella with a focus on recent developments in T cell memory formation.
Collapse
Affiliation(s)
- Alana T Nguyen
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Stephen J McSorley
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
2
|
Batool R, Qamar ZH, Salam RA, Yousafzai MT, Ashorn P, Qamar FN. Efficacy of typhoid vaccines against culture-confirmed Salmonella Typhi in typhoid endemic countries: a systematic review and meta-analysis. Lancet Glob Health 2024; 12:e589-e598. [PMID: 38485426 DOI: 10.1016/s2214-109x(23)00606-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 03/19/2024]
Abstract
BACKGROUND Typhoid is a serious public health threat in many low-income and middle-income countries. Several vaccines for typhoid have been recommended by WHO for typhoid prevention in endemic countries. This study aimed to review the efficacy of typhoid vaccines against culture-confirmed Salmonella enterica serovar Typhi. METHODS We searched the Cochrane Central Register of Controlled Trials, MEDLINE, and Embase for studies published in English between Jan 1, 1986 and Nov 2, 2023. We included randomised controlled trials (RCTs) comparing typhoid vaccines with a placebo or another vaccine. This meta-analysis evaluated the efficacy and safety of several typhoid vaccines, including live attenuated oral Ty21a vaccine, Vi capsular polysaccharide (Vi-PS), Vi polysaccharide conjugated to recombinant Pseudomonas aeruginosa exotoxin A vaccine (Vi-rEPA), and Vi-tetanus toxoid conjugate vaccine (TCV). The certainty of evidence for key outcomes was evaluated using Grading of Recommendations, Assessment, Development, and Evaluations methodology. The outcome of interest was typhoid fever confirmed by the isolation of Salmonella enterica serovar Typhi in blood and adverse events following immunisation. This study is registered with PROSPERO (CRD42021241043). FINDINGS We included 14 RCTs assessing four different vaccines (Ty21a: four trials; Vi-PS: five trials; Vi-rEPA: one trial; TCV: four trials) involving 585 253 participants. All trials were conducted in typhoid endemic countries and the age of participants ranged from 6 months to 50 years. The pooled efficacy against typhoid fever was 45% (95% CI 33-55%; four trials; 247 649 participants; I2 59%; moderate certainty) for Ty21a and 58% (44-69%; five trials; 214 456 participants; I2 34%; moderate certainty) for polysaccharide Vi-PS. The cumulative efficacy of two doses of Vi-rEPA vaccine at 2 years was 91% (88-96%; one trial; 12 008 participants; moderate certainty). The pooled efficacy of a single shot of TCV at 2 years post-immunisation was 83% (77-87%; four trials; 111 130 participants; I2 0%; moderate certainty). All vaccines were safe, with no serious adverse effects reported in the trials. INTERPRETATION The existing data from included trials provide promising results regarding the efficacy and safety of the four recommended typhoid vaccines. TCV and Vi-rEPA were found to have the highest efficacy at 2 years post-immunisation. However, follow-up data for Vi-rEPA are scarce and only TCV is pre-qualified by WHO. Therefore, roll-out of TCV into routine immunisation programmes in typhoid endemic settings is highly recommended. FUNDING There was no funding source for this study.
Collapse
Affiliation(s)
- Rabab Batool
- Department of Epidemiology and Biostatistics, Aga Khan University Hospital, Karachi, Pakistan; Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Pakistan; Centre for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Zoya Haq Qamar
- Liaquat National Hospital and Medical College, Karachi, Pakistan
| | | | - Mohammad Tahir Yousafzai
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Pakistan; The Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Per Ashorn
- Centre for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Farah Naz Qamar
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Pakistan.
| |
Collapse
|
3
|
Webster E, Palanco Lopez P, Kirchhelle C. Shifting targets: typhoid's transformation from an environmental to a vaccine-preventable disease, 1940-2019. THE LANCET. INFECTIOUS DISEASES 2024; 24:e232-e244. [PMID: 37995738 DOI: 10.1016/s1473-3099(23)00500-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 11/25/2023]
Abstract
160 years after the discovery of its waterborne transmission and 120 years after the development of the first-generation of vaccines, typhoid fever remains a major health threat globally. In this Historical Review, we use WHO's Institutional Repository for Information Sharing to examine changes in typhoid control policy from January, 1940, to December, 2019. We used a mixed-methods approach in the analysis of infection control priorities, combining semi-inductive thematic coding with historical analysis to show major thematic shifts in typhoid control policy, away from water, sanitation, and hygiene (WASH)-based control towards vaccine-based interventions concurrent with declining attention to the disease. Documentary analysis shows that, although international planners never officially disavowed WASH and low-income countries persistently lobbied for WASH, vaccines emerged as a permanent stopgap while meaningful support of sustained WASH strengthening lost momentum-with serious, long-term ramifications for typhoid control.
Collapse
Affiliation(s)
- Emily Webster
- Department of Philosophy, Durham University, Durham, UK; Department of Philosophy, University of Johannesburg, Johannesburg, South Africa
| | | | - Claas Kirchhelle
- School of History, University College Dublin, Dublin, Ireland; Oxford Vaccine Group, University of Oxford, Oxford, UK.
| |
Collapse
|
4
|
Zhu H, Chelysheva I, Cross DL, Blackwell L, Jin C, Gibani MM, Jones E, Hill J, Trück J, Kelly DF, Blohmke CJ, Pollard AJ, O’Connor D. Molecular correlates of vaccine-induced protection against typhoid fever. J Clin Invest 2023; 133:e169676. [PMID: 37402153 PMCID: PMC10425215 DOI: 10.1172/jci169676] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUNDTyphoid fever is caused by the Gram-negative bacterium Salmonella enterica serovar Typhi and poses a substantial public health burden worldwide. Vaccines have been developed based on the surface Vi-capsular polysaccharide of S. Typhi; these include a plain-polysaccharide-based vaccine, ViPS, and a glycoconjugate vaccine, ViTT. To understand immune responses to these vaccines and their vaccine-induced immunological protection, molecular signatures were analyzed using bioinformatic approaches.METHODSBulk RNA-Seq data were generated from blood samples obtained from adult human volunteers enrolled in a vaccine trial, who were then challenged with S. Typhi in a controlled human infection model (CHIM). These data were used to conduct differential gene expression analyses, gene set and modular analyses, B cell repertoire analyses, and time-course analyses at various post-vaccination and post-challenge time points between participants receiving ViTT, ViPS, or a control meningococcal vaccine.RESULTSTranscriptomic responses revealed strong differential molecular signatures between the 2 typhoid vaccines, mostly driven by the upregulation in humoral immune signatures, including selective usage of immunoglobulin heavy chain variable region (IGHV) genes and more polarized clonal expansions. We describe several molecular correlates of protection against S. Typhi infection, including clusters of B cell receptor (BCR) clonotypes associated with protection, with known binders of Vi-polysaccharide among these.CONCLUSIONThe study reports a series of contemporary analyses that reveal the transcriptomic signatures after vaccination and infectious challenge, while identifying molecular correlates of protection that may inform future vaccine design and assessment.TRIAL REGISTRATIONClinicalTrials.gov NCT02324751.
Collapse
Affiliation(s)
- Henderson Zhu
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre and Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Irina Chelysheva
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre and Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Deborah L. Cross
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre and Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Luke Blackwell
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre and Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Celina Jin
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre and Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Malick M. Gibani
- Department of Infectious Disease, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Elizabeth Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre and Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Jennifer Hill
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre and Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Johannes Trück
- Division of Immunology, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Dominic F. Kelly
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre and Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Christoph J. Blohmke
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre and Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre and Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Daniel O’Connor
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre and Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
5
|
Saha T, Arisoyin AE, Bollu B, Ashok T, Babu A, Issani A, Jhaveri S, Avanthika C. Enteric Fever: Diagnostic Challenges and the Importance of Early Intervention. Cureus 2023; 15:e41831. [PMID: 37575696 PMCID: PMC10423039 DOI: 10.7759/cureus.41831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Enteric fever is a systemic infection caused by highly virulent Salmonella enterica serovars: Typhi and Paratyphi. Diagnosis of enteric fever is challenging due to a wide variety of clinical features which overlap with other febrile illnesses. The current diagnostic methods are limited because of the suboptimal sensitivity of conventional tests like blood culture in detecting organisms and the invasive nature of bone marrow culture. It emphasizes the need to develop improved and more reliable diagnostic modalities. The rising rates of multidrug-resistant Salmonella strains call for an accurate understanding of the current management of the disease. Proper public health measures and large-scale immunization programs will help reduce the burden of the disease. A comprehensive surveillance system can help detect the chronic carrier state and is crucial in understanding antibiotic susceptibility patterns. We conducted an all-language literature search on Medline, Cochrane, Embase, and Google Scholar till May 2022. The following search words and medical subject headings (MeSH) were used: "enteric fever," "Salmonella Typhi," "multidrug-resistant Salmonella," chronic carrier state," "Salmonella detection, "and "typhoid vaccine." We reviewed the literature on clinical features, pathophysiology, new diagnostic tests, and interventions to prevent the disease. This article explores enteric fever and its various clinical features and addresses the emerging threat of multidrug resistance. It focuses on novel methods for diagnosis and prevention strategies, including vaccines and the use of surveillance systems employed across different parts of the world.
Collapse
Affiliation(s)
- Tias Saha
- Internal Medicine, Samorita General Hospital, Faridpur, BGD
- Internal Medicine, Diabetic Association Medical College, Faridpur, BGD
| | | | - Bhaswanth Bollu
- Emergency Medicine, All India Institute of Medical Sciences, New Delhi, IND
| | - Tejaswini Ashok
- Internal Medicine, Jagadguru Sri Shivarathreeshwara Medical College, Mysore, IND
| | - Athira Babu
- Pediatrics, Saudi German Hospital, Dubai, ARE
| | - Ali Issani
- Emergency Medicine, Aga Khan University, Karachi, PAK
| | - Sharan Jhaveri
- Internal Medicine, Nathiba Hargovandas Lakhmichand Municipal Medical College, Ahmedabad, IND
| | - Chaithanya Avanthika
- Pediatrics, Icahn School of Medicine at Mount Sinai, Elmhurst Hospital Center, New York, USA
- Medicine and Surgery, Karnataka Institute of Medical Sciences, Hubli, IND
| |
Collapse
|
6
|
Naini A, Bartetzko MP, Sanapala SR, Broecker F, Wirtz V, Lisboa MP, Parameswarappa SG, Knopp D, Przygodda J, Hakelberg M, Pan R, Patel A, Chorro L, Illenberger A, Ponce C, Kodali S, Lypowy J, Anderson AS, Donald RGK, von Bonin A, Pereira CL. Semisynthetic Glycoconjugate Vaccine Candidates against Escherichia coli O25B Induce Functional IgG Antibodies in Mice. JACS AU 2022; 2:2135-2151. [PMID: 36186572 PMCID: PMC9516715 DOI: 10.1021/jacsau.2c00401] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 06/01/2023]
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) is a major health concern due to emerging antibiotic resistance. Along with O1A, O2, and O6A, E. coli O25B is a major serotype within the ExPEC group, which expresses a unique O-antigen. Clinical studies with a glycoconjugate vaccine of the above-mentioned O-types revealed O25B as the least immunogenic component, inducing relatively weak IgG titers. To evaluate the immunological properties of semisynthetic glycoconjugate vaccine candidates against E. coli O25B, we here report the chemical synthesis of an initial set of five O25B glycan antigens differing in length, from one to three repeat units, and frameshifts of the repeat unit. The oligosaccharide antigens were conjugated to the carrier protein CRM197. The resulting semisynthetic glycoconjugates induced functional IgG antibodies in mice with opsonophagocytic activity against E. coli O25B. Three of the oligosaccharide-CRM197 conjugates elicited functional IgGs in the same order of magnitude as a conventional CRM197 glycoconjugate prepared with native O25B O-antigen and therefore represent promising vaccine candidates for further investigation. Binding studies with two monoclonal antibodies (mAbs) revealed nanomolar anti-O25B IgG responses with nanomolar K D values and with varying binding epitopes. The immunogenicity and mAb binding data now allow for the rational design of additional synthetic antigens for future preclinical studies, with expected further improvements in the functional antibody responses. Moreover, acetylation of a rhamnose residue was shown to be likely dispensable for immunogenicity, as a deacylated antigen was able to elicit strong functional IgG responses. Our findings strongly support the feasibility of a semisynthetic glycoconjugate vaccine against E. coli O25B.
Collapse
Affiliation(s)
- Arun Naini
- Vaxxilon
Deutschland GmbH, Part of Idorsia Pharmaceuticals Ltd., Magnusstr. 11, 12489 Berlin, Germany
| | - Max Peter Bartetzko
- Vaxxilon
Deutschland GmbH, Part of Idorsia Pharmaceuticals Ltd., Magnusstr. 11, 12489 Berlin, Germany
| | - Someswara Rao Sanapala
- Vaxxilon
Deutschland GmbH, Part of Idorsia Pharmaceuticals Ltd., Magnusstr. 11, 12489 Berlin, Germany
| | - Felix Broecker
- Vaxxilon
Deutschland GmbH, Part of Idorsia Pharmaceuticals Ltd., Magnusstr. 11, 12489 Berlin, Germany
| | - Victoria Wirtz
- Vaxxilon
Deutschland GmbH, Part of Idorsia Pharmaceuticals Ltd., Magnusstr. 11, 12489 Berlin, Germany
| | - Marilda P. Lisboa
- Vaxxilon
Deutschland GmbH, Part of Idorsia Pharmaceuticals Ltd., Magnusstr. 11, 12489 Berlin, Germany
| | | | - Daniel Knopp
- Vaxxilon
Deutschland GmbH, Part of Idorsia Pharmaceuticals Ltd., Magnusstr. 11, 12489 Berlin, Germany
| | - Jessica Przygodda
- Vaxxilon
Deutschland GmbH, Part of Idorsia Pharmaceuticals Ltd., Magnusstr. 11, 12489 Berlin, Germany
| | - Matthias Hakelberg
- Vaxxilon
Deutschland GmbH, Part of Idorsia Pharmaceuticals Ltd., Magnusstr. 11, 12489 Berlin, Germany
| | - Rosalind Pan
- Pfizer
Vaccine Research and Development, Pearl River, New York 10965, United States
| | - Axay Patel
- Pfizer
Vaccine Research and Development, Pearl River, New York 10965, United States
| | - Laurent Chorro
- Pfizer
Vaccine Research and Development, Pearl River, New York 10965, United States
| | - Arthur Illenberger
- Pfizer
Vaccine Research and Development, Pearl River, New York 10965, United States
| | - Christopher Ponce
- Pfizer
Vaccine Research and Development, Pearl River, New York 10965, United States
| | - Srinivas Kodali
- Pfizer
Vaccine Research and Development, Pearl River, New York 10965, United States
| | - Jacqueline Lypowy
- Pfizer
Vaccine Research and Development, Pearl River, New York 10965, United States
| | | | - Robert G. K. Donald
- Pfizer
Vaccine Research and Development, Pearl River, New York 10965, United States
| | - Arne von Bonin
- Vaxxilon
Deutschland GmbH, Part of Idorsia Pharmaceuticals Ltd., Magnusstr. 11, 12489 Berlin, Germany
| | - Claney L. Pereira
- Vaxxilon
Deutschland GmbH, Part of Idorsia Pharmaceuticals Ltd., Magnusstr. 11, 12489 Berlin, Germany
| |
Collapse
|
7
|
Liston SD, Ovchinnikova OG, Kimber MS, Whitfield C. A dedicated C-6 β-hydroxyacyltransferase required for biosynthesis of the glycolipid anchor for Vi antigen capsule in typhoidal Salmonella. J Biol Chem 2022; 298:102520. [PMID: 36152747 DOI: 10.1016/j.jbc.2022.102520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Vi antigen is an extracellular polysaccharide produced by Salmonella enterica Typhi, Citrobacter freundii, and some soil bacteria belonging to the Burkholderiales. In Salmonella Typhi, Vi-antigen capsule protects the bacterium against host defenses, and the glycan is used in a current glycoconjugate vaccine to protect against typhoid. Vi antigen is a glycolipid assembled in the cytoplasm and translocated to the cell surface by an export complex driven by an ABC transporter. In Salmonella Typhi, efficient export and cell-surface retention of the capsule layer depend on a reducing terminal acylated-HexNAc moiety. Although the precise structure and biosynthesis of the acylated terminus has not been resolved, it distinguishes Vi antigen from other known glycolipid substrates for bacterial ABC transporters. The genetic locus for Vi antigen-biosynthesis encodes a single acyltransferase candidate (VexE), which is implicated in the acylation process. Here, we determined the structure of the VexE in vitro reaction product by mass spectrometry and nuclear magnetic resonance spectroscopy, to reveal that VexE catalyzes β-hydroxyacyl-ACP dependent acylation of the activated sugar precursor, uridine-5'-diphospho-N-acetylglucosamine (UDP-GlcNAc), at C-6 to form UDP-6-O-[β-hydroxymyristoyl]-α-d-GlcNAc. VexE belongs to the lysophosphatidyl acyltransferase (LPLAT) family, and comparison of an Alphafold VexE model to solved LPLAT structures, together with modeling enzyme:substrate complexes, led us to predict an enzyme mechanism. This study provides new insight into Vi terminal structure, offers a new model substrate to investigate the mechanism of glycolipid ABC transporters, and adds biochemical understanding for a novel reaction used in synthesis of an important bacterial virulence factor.
Collapse
Affiliation(s)
- S D Liston
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - O G Ovchinnikova
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - M S Kimber
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - C Whitfield
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
8
|
Sztein MB, Booth JS. Controlled human infectious models, a path forward in uncovering immunological correlates of protection: Lessons from enteric fevers studies. Front Microbiol 2022; 13:983403. [PMID: 36204615 PMCID: PMC9530043 DOI: 10.3389/fmicb.2022.983403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Enteric infectious diseases account for more than a billion disease episodes yearly worldwide resulting in approximately 2 million deaths, with children under 5 years old and the elderly being disproportionally affected. Enteric pathogens comprise viruses, parasites, and bacteria; the latter including pathogens such as Salmonella [typhoidal (TS) and non-typhoidal (nTS)], cholera, Shigella and multiple pathotypes of Escherichia coli (E. coli). In addition, multi-drug resistant and extensively drug-resistant (XDR) strains (e.g., S. Typhi H58 strain) of enteric bacteria are emerging; thus, renewed efforts to tackle enteric diseases are required. Many of these entero-pathogens could be controlled by oral or parenteral vaccines; however, development of new, effective vaccines has been hampered by lack of known immunological correlates of protection (CoP) and limited knowledge of the factors contributing to protective responses. To fully comprehend the human response to enteric infections, an invaluable tool that has recently re-emerged is the use of controlled human infection models (CHIMs) in which participants are challenged with virulent wild-type (wt) organisms. CHIMs have the potential to uncover immune mechanisms and identify CoP to enteric pathogens, as well as to evaluate the efficacy of therapeutics and vaccines in humans. CHIMs have been used to provide invaluable insights in the pathogenesis, host-pathogen interaction and evaluation of vaccines. Recently, several Oxford typhoid CHIM studies have been performed to assess the role of multiple cell types (B cells, CD8+ T, Tregs, MAIT, Monocytes and DC) during S. Typhi infection. One of the key messages that emerged from these studies is that baseline antigen-specific responses are important in that they can correlate with clinical outcomes. Additionally, volunteers who develop typhoid disease (TD) exhibit higher levels and more activated cell types (e.g., DC and monocytes) which are nevertheless defective in discrete signaling pathways. Future critical aspects of this research will involve the study of immune responses to enteric infections at the site of entry, i.e., the intestinal mucosa. This review will describe our current knowledge of immunity to enteric fevers caused byS. Typhi and S. Paratyphi A, with emphasis on the contributions of CHIMs to uncover the complex immunological responses to these organisms and provide insights into the determinants of protective immunity.
Collapse
Affiliation(s)
- Marcelo B. Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- *Correspondence: Marcelo B. Sztein,
| | - Jayaum S. Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Jayaum S. Booth,
| |
Collapse
|
9
|
Facility-based disease surveillance and Bayesian hierarchical modeling to estimate endemic typhoid fever incidence, Kilimanjaro Region, Tanzania, 2007–2018. PLoS Negl Trop Dis 2022; 16:e0010516. [PMID: 35788572 PMCID: PMC9286265 DOI: 10.1371/journal.pntd.0010516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/15/2022] [Accepted: 05/17/2022] [Indexed: 11/25/2022] Open
Abstract
Growing evidence suggests considerable variation in endemic typhoid fever incidence at some locations over time, yet few settings have multi-year incidence estimates to inform typhoid control measures. We sought to describe a decade of typhoid fever incidence in the Kilimanjaro Region of Tanzania. Cases of blood culture confirmed typhoid were identified among febrile patients at two sentinel hospitals during three study periods: 2007–08, 2011–14, and 2016–18. To account for under-ascertainment at sentinel facilities, we derived adjustment multipliers from healthcare utilization surveys done in the hospital catchment area. Incidence estimates and credible intervals (CrI) were derived using a Bayesian hierarchical incidence model that incorporated uncertainty of our observed typhoid fever prevalence, of healthcare seeking adjustment multipliers, and of blood culture diagnostic sensitivity. Among 3,556 total participants, 50 typhoid fever cases were identified. Of typhoid cases, 26 (52%) were male and the median (range) age was 22 (<1–60) years; 4 (8%) were aged <5 years and 10 (20%) were aged 5 to 14 years. Annual typhoid fever incidence was estimated as 61.5 (95% CrI 14.9–181.9), 6.5 (95% CrI 1.4–20.4), and 4.0 (95% CrI 0.6–13.9) per 100,000 persons in 2007–08, 2011–14, and 2016–18, respectively. There were no deaths among typhoid cases. We estimated moderate typhoid incidence (≥10 per 100 000) in 2007–08 and low (<10 per 100 000) incidence during later surveillance periods, but with overlapping credible intervals across study periods. Although consistent with falling typhoid incidence, we interpret this as showing substantial variation over the study periods. Given potential variation, multi-year surveillance may be warranted in locations making decisions about typhoid conjugate vaccine introduction and other control measures. There is evidence that typhoid fever incidence may vary over time, but there are few longitudinal studies estimating incidence. This is especially true in Sub-Saharan Africa, where recent estimates show wide variation in incidence across different settings, but very limited longitudinal descriptions from those settings. Incidence estimates were generated using facility-based surveillance data from three study periods that was adjusted for health-seeking behavior established through healthcare utilization surveys performed in the catchment area. In addition to coupling facility-based surveillance data with healthcare utilization data, we utilized a Bayesian statistical methodology in order to estimate incidence and characterize uncertainty around the estimates. Our results demonstrate moderate typhoid incidence in 2007–08 and low incidence during 2012–14 and 2016–18, but with overlapping credible intervals across study periods. Our data are consistent with evidence that endemic typhoid may vary substantially over time. Given potential variation, multi-year surveillance may be warranted in locations making decisions about typhoid conjugate vaccine introduction and other control measures.
Collapse
|
10
|
Wear SS, Sande C, Ovchinnikova OG, Preston A, Whitfield C. Investigation of core machinery for biosynthesis of Vi antigen capsular polysaccharides in Gram-negative bacteria. J Biol Chem 2021; 298:101486. [PMID: 34896394 PMCID: PMC8760489 DOI: 10.1016/j.jbc.2021.101486] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 11/18/2022] Open
Abstract
Salmonella enterica serovar Typhi causes typhoid fever. It possesses a Vi antigen capsular polysaccharide coat that is important for virulence and is the basis of a current glycoconjugate vaccine. Vi antigen is also produced by environmental Bordetella isolates, while mammal-adapted Bordetella species (such as Bordetella bronchiseptica) produce a capsule of undetermined structure that cross-reacts with antibodies recognizing Vi antigen. The Vi antigen backbone is composed of poly-α-(1→4)-linked N-acetylgalactosaminuronic acid, modified with O-acetyl residues that are necessary for vaccine efficacy. Despite its biological and biotechnological importance, some central aspects of Vi antigen production are poorly understood. Here we demonstrate that TviE and TviD, two proteins encoded in the viaB (Vi antigen production) locus, interact and are the Vi antigen polymerase and O-acetyltransferase, respectively. Structural modeling and site-directed mutagenesis reveal that TviE is a GT4-family glycosyltransferase. While TviD has no identifiable homologs beyond Vi antigen systems in other bacteria, structural modeling suggests that it belongs to the large SGNH hydrolase family, which contains other O-acetyltransferases. Although TviD possesses an atypical catalytic triad, its O-acetyltransferase function was verified by antibody reactivity and 13C NMR data for tviD-mutant polysaccharide. The B. bronchiseptica genetic locus predicts a mode of synthesis distinct from classical S. enterica Vi antigen production, but which still involves TviD and TviE homologs that are both active in a reconstituted S. Typhi system. These findings provide new insight into Vi antigen production and foundational information for the glycoengineering of Vi antigen production in heterologous bacteria.
Collapse
Affiliation(s)
- Samantha S Wear
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Caitlin Sande
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Olga G Ovchinnikova
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Andrew Preston
- Milner Centre for Evolution and Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, UK
| | - Chris Whitfield
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
11
|
Patel PD, Patel P, Liang Y, Meiring JE, Misiri T, Mwakiseghile F, Tracy JK, Masesa C, Msuku H, Banda D, Mbewe M, Henrion M, Adetunji F, Simiyu K, Rotrosen E, Birkhold M, Nampota N, Nyirenda OM, Kotloff K, Gmeiner M, Dube Q, Kawalazira G, Laurens MB, Heyderman RS, Gordon MA, Neuzil KM. Safety and Efficacy of a Typhoid Conjugate Vaccine in Malawian Children. N Engl J Med 2021; 385:1104-1115. [PMID: 34525285 PMCID: PMC8202713 DOI: 10.1056/nejmoa2035916] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Typhoid fever caused by multidrug-resistant H58 Salmonella Typhi is an increasing public health threat in sub-Saharan Africa. METHODS We conducted a phase 3, double-blind trial in Blantyre, Malawi, to assess the efficacy of Vi polysaccharide typhoid conjugate vaccine (Vi-TCV). We randomly assigned children who were between 9 months and 12 years of age, in a 1:1 ratio, to receive a single dose of Vi-TCV or meningococcal capsular group A conjugate (MenA) vaccine. The primary outcome was typhoid fever confirmed by blood culture. We report vaccine efficacy and safety outcomes after 18 to 24 months of follow-up. RESULTS The intention-to-treat analysis included 28,130 children, of whom 14,069 were assigned to receive Vi-TCV and 14,061 were assigned to receive the MenA vaccine. Blood culture-confirmed typhoid fever occurred in 12 children in the Vi-TCV group (46.9 cases per 100,000 person-years) and in 62 children in the MenA group (243.2 cases per 100,000 person-years). Overall, the efficacy of Vi-TCV was 80.7% (95% confidence interval [CI], 64.2 to 89.6) in the intention-to-treat analysis and 83.7% (95% CI, 68.1 to 91.6) in the per-protocol analysis. In total, 130 serious adverse events occurred in the first 6 months after vaccination (52 in the Vi-TCV group and 78 in the MenA group), including 6 deaths (all in the MenA group). No serious adverse events were considered by the investigators to be related to vaccination. CONCLUSIONS Among Malawian children 9 months to 12 years of age, administration of Vi-TCV resulted in a lower incidence of blood culture-confirmed typhoid fever than the MenA vaccine. (Funded by the Bill and Melinda Gates Foundation; ClinicalTrials.gov number, NCT03299426.).
Collapse
Affiliation(s)
- Priyanka D Patel
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Pratiksha Patel
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Yuanyuan Liang
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - James E Meiring
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Theresa Misiri
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Felistas Mwakiseghile
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - J Kathleen Tracy
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Clemens Masesa
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Harrison Msuku
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - David Banda
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Maurice Mbewe
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Marc Henrion
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Fiyinfolu Adetunji
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Kenneth Simiyu
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Elizabeth Rotrosen
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Megan Birkhold
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Nginache Nampota
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Osward M Nyirenda
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Karen Kotloff
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Markus Gmeiner
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Queen Dube
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Gift Kawalazira
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Matthew B Laurens
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Robert S Heyderman
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Melita A Gordon
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| | - Kathleen M Neuzil
- From the Malawi-Liverpool-Wellcome Program (P.D.P., P.P., J.E.M., T.M., F.M., C.M., H.M., D.B., M.M., M.H., M.G., M.A.G.), the Blantyre Malaria Project (N.N., O.M.N.), the Department of Paediatrics, Queen Elizabeth Central Hospital (Q.D.), the District Health Office, Blantyre District Council (G.K.), and Kamuzu University of Health Sciences (M.A.G.) - all in Blantyre, Malawi; the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore (Y.L., J.K.T., F.A., K.S., E.R., M.B., K.K., M.B.L., K.M.N.); and Oxford Vaccine Group, the Department of Paediatrics, Oxford University, Oxford (J.E.M.), Liverpool School of Tropical Medicine (C.M., M.H., M.G.), and the Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool (M.A.G.), and the Division of Infection and Immunity, University College London, London (R.S.H.) - all in the United Kingdom
| |
Collapse
|
12
|
Haque S, Swami P, Khan A. S. Typhi derived vaccines and a proposal for outer membrane vesicles (OMVs) as potential vaccine for typhoid fever. Microb Pathog 2021; 158:105082. [PMID: 34265371 DOI: 10.1016/j.micpath.2021.105082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 12/22/2022]
Abstract
Typhoid fever is a serious systemic infection caused by Salmonella Typhi (S. Typhi), spread by the feco-oral route and closely associated with poor food hygiene and inadequate sanitation. Nearly 93% of S. Typhi strains have acquired antibiotic resistance against most antibiotics. Vaccination is the only promising way to prevent typhoid fever. This review covers the nature and composition of S. Typhi, pathogenecity and mode of infection, epidemiology, and nature of drug resistance. Several components (Vi-polysaccharides, O-antigens, flagellar antigens, full length OMPs, and short peptides from OMPs) of S. Typhi have been utilized for vaccine design for protection against typhoid fever. Vaccine delivery systems also contribute to efficacy of the vaccines. In this study, we propose to develop S. Typhi derived OMVs as vaccine for protection against typhoid fevers.
Collapse
Affiliation(s)
- Shabirul Haque
- Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA.
| | - Pooja Swami
- Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA.
| | - Azhar Khan
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal, Pradesh, India.
| |
Collapse
|
13
|
Ghaderi E, Zahraei SM, Moradi G, Goodarzi E, Norouzinejad A, Mohsenpour B, Naemi H, Khazaei Z. Geographical distribution of Typhoid using Geographic Information System (GIS) during 2009-2014 in Iran. Med J Islam Repub Iran 2021; 35:35. [PMID: 34211937 PMCID: PMC8236083 DOI: 10.47176/mjiri.35.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Indexed: 11/09/2022] Open
Abstract
Background: Salmonella induced infections remain one of the most important health problems worldwide. The purpose of this study is to investigate the incidence and geographical distribution of typhoid using GIS and to predict its incidence in Iran in 2021.
Methods: This study is a descriptive analytical study. Information on pertussis was obtained from the Center for Communicable Diseases Control during 2009-2015. In the next step, ArcGIS 9.3 was used to prepare geographic maps of the disease incidence and frequency Therefore, using the Raster Calculator tool, the disease prediction map was drawn.
Results: The results showed that the highest incidence of typhoid during 2009-2014 was in Kermanshah, Lorestan, Hamadan, Kurdistan, and Ilam provinces. The incidence of typhoid in Iran increased during 2009-2010. The annual incidence of typhoid decreased from 0.85 per 100,000 in 2010 to 0.5 in 2014. Based on the modeling results for Iran, Kermanshah, Lorestan, Kurdistan, Ilam and Hamadan provinces with 92.17%, 46.56%, 31.74%, 25.62% and 22.96% of their areas (Km2) are at high risk for typhoid in the coming years, respectively.
Conclusion: Considering that the provinces of Kermanshah, Lorestan, Kurdistan, Ilam, and Hamadan are at risk of typhoid incidence in the coming years in Iran, and given that salmonella infections have a direct relationship with the individual’s health status and individual’s environmental health and socioeconomic status, improving the health status and disease control in carriers as well as improving the socio-economic status of the population living in these areas can prevent the disease in the years to come.
Collapse
Affiliation(s)
- Ebrahim Ghaderi
- Zoonoses Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Seyed Mohsen Zahraei
- Center for Communicable Diseases Control, Ministry of Health and Medical Education, Tehran, Iran
| | - Ghobad Moradi
- Social Determinants of Health Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Elham Goodarzi
- Social Determinants of Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Abbas Norouzinejad
- Deputy for Administrative Affairs The Center for Communicable Disease Control, Ministry of Health and Medical Education, Tehran, Iran
| | - Behzad Mohsenpour
- Department of Infectious Disease, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Hasan Naemi
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Zaher Khazaei
- Department of Public Health,School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| |
Collapse
|
14
|
Masuet-Aumatell C, Atouguia J. Typhoid fever infection - Antibiotic resistance and vaccination strategies: A narrative review. Travel Med Infect Dis 2020; 40:101946. [PMID: 33301931 DOI: 10.1016/j.tmaid.2020.101946] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
Typhoid fever is a bacterial infection caused by the Gram-negative bacterium Salmonella enterica subspecies enterica serovar Typhi (S. Typhi), prevalent in many low- and middle-income countries. In high-income territories, typhoid fever is predominantly travel-related, consequent to travel in typhoid-endemic regions; however, data show that the level of typhoid vaccination in travellers is low. Successful management of typhoid fever using antibiotics is becoming increasingly difficult due to drug resistance; emerging resistance has spread geographically due to factors such as increasing travel connectivity, affecting those in endemic regions and travellers alike. This review provides an overview of: the epidemiology and diagnosis of typhoid fever; the emergence of drug-resistant typhoid strains in the endemic setting; drug resistance observed in travellers; vaccines currently available to prevent typhoid fever; vaccine recommendations for people living in typhoid-endemic regions; strategies for the introduction of typhoid vaccines and stakeholders in vaccination programmes; and travel recommendations for a selection of destinations with a medium or high incidence of typhoid fever.
Collapse
Affiliation(s)
- Cristina Masuet-Aumatell
- Preventive Medicine Department, Bellvitge Biomedical Research Institute (IDIBELL), University Hospital of Bellvitge, Faculty of Medicine, University of Barcelona, Feixa Llarga s/n, L'Hospitalet de Llobregat, 08907, Barcelona, Catalonia, Spain.
| | - Jorge Atouguia
- Instituto Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junquiera, 100, Lisbon, Portugal.
| |
Collapse
|
15
|
Kim JH, Im J, Parajulee P, Holm M, Cruz Espinoza LM, Poudyal N, Mogeni OD, Marks F. A Systematic Review of Typhoid Fever Occurrence in Africa. Clin Infect Dis 2020; 69:S492-S498. [PMID: 31665777 PMCID: PMC6821235 DOI: 10.1093/cid/ciz525] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Our current understanding of the burden and distribution of typhoid fever in Africa relies on extrapolation of data from a small number of population-based incidence rate estimates. However, many other records on the occurrence of typhoid fever are available, and those records contain information that may enrich our understanding of the epidemiology of the disease as well as secular trends in reporting by country and over time. METHODS We conducted a systematic review of typhoid fever occurrence in Africa, published in PubMed, Embase, and ProMED (Program for Monitoring Emerging Diseases). RESULTS At least one episode of culture-confirmed typhoid fever was reported in 42 of 57 African countries during 1900-2018. The number of reports on typhoid fever has increased over time in Africa and was highly heterogeneous between countries and over time. Outbreaks of typhoid fever were reported in 15 countries, with their frequency and size increasing over time. CONCLUSIONS Efforts should be made to leverage existing typhoid data, for example, by incorporating them into models for estimating the burden and distribution of typhoid fever.
Collapse
Affiliation(s)
- Jong-Hoon Kim
- Public Health, Access, and Vaccine Epidemiology (PAVE) Unit, International Vaccine Institute, Seoul, Korea
| | - Justin Im
- Public Health, Access, and Vaccine Epidemiology (PAVE) Unit, International Vaccine Institute, Seoul, Korea
| | - Prerana Parajulee
- Public Health, Access, and Vaccine Epidemiology (PAVE) Unit, International Vaccine Institute, Seoul, Korea
| | - Marianne Holm
- Public Health, Access, and Vaccine Epidemiology (PAVE) Unit, International Vaccine Institute, Seoul, Korea
| | - Ligia Maria Cruz Espinoza
- Public Health, Access, and Vaccine Epidemiology (PAVE) Unit, International Vaccine Institute, Seoul, Korea
| | - Nimesh Poudyal
- Public Health, Access, and Vaccine Epidemiology (PAVE) Unit, International Vaccine Institute, Seoul, Korea
| | - Ondari D Mogeni
- Public Health, Access, and Vaccine Epidemiology (PAVE) Unit, International Vaccine Institute, Seoul, Korea
| | - Florian Marks
- Public Health, Access, and Vaccine Epidemiology (PAVE) Unit, International Vaccine Institute, Seoul, Korea.,Department of Medicine, University of Cambridge, United Kingdom
| |
Collapse
|
16
|
Ali M, Sur D, Kanungo S, Qadri F, Kim DR, Islam T, Im J, Ahmmed F, Chon Y, Khan AI, Zaman K, Marks F, Dutta S, Bhattacharya SK, Clemens JD. Re-evaluating herd protection by Vi typhoid vaccine in a cluster randomized trial. Int Health 2020; 12:36-42. [PMID: 31608962 PMCID: PMC6964216 DOI: 10.1093/inthealth/ihz069] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/04/2019] [Accepted: 07/11/2019] [Indexed: 01/14/2023] Open
Abstract
Background In a cluster randomized trial (CRT) of a Vi polysaccharide vaccine against typhoid in the slums of Kolkata we found evidence of vaccine herd protection. However, transmission of typhoid into clusters from the outside likely occurred in this densely populated setting, which could have diminished our estimates of vaccine herd protection. Methods Eighty clusters (40 in each arm) were randomised to receive a single dose of either Vi or inactivated hepatitis A vaccine. We analysed protection for the entire cluster and for subclusters consisting of residents of the innermost households. Results During 2 y of follow-up, total protection was 61% (95% CI 41 to 75), overall protection was 57% (95% CI 37 to 71) and indirect protection was 44% (95% CI 2 to 69). Analyses of the innermost 75% and 50% of households of the clusters showed similar findings. However, in the innermost 25% of households of the clusters, total protection was 82% (95% CI 48 to 94) and overall protection was 66% (95% CI 27 to 84). There was not a sufficient sample size to demonstrate such a trend for indirect protection in these innermost households. Conclusions The findings suggest that analyses of the entire cluster may have led to underestimation of herd protection against typhoid by Vi vaccine and that restriction of the analyses to the inner subclusters may have led to a more accurate estimation of vaccine herd effects.
Collapse
Affiliation(s)
- Mohammad Ali
- Johns Hopkins Bloomberg School of Public Health, 615, N Wolfe Street, Baltimore, MD-21205, USA
| | - Dipika Sur
- National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme XM, Beliaghata, Kolkata, India
| | - Suman Kanungo
- National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme XM, Beliaghata, Kolkata, India
| | - Firdausi Qadri
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka 1212, Bangladesh
| | - Deok Ryun Kim
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Taufiqul Islam
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka 1212, Bangladesh
| | - Justin Im
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Faisal Ahmmed
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka 1212, Bangladesh
| | - Yun Chon
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Ashraful Islam Khan
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka 1212, Bangladesh
| | - K Zaman
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka 1212, Bangladesh
| | - Florian Marks
- International Vaccine Institute, SNU Research Park, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea.,Department of Medicine, University of Cambridge, Cambridge, UK
| | - Shanta Dutta
- National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme XM, Beliaghata, Kolkata, India
| | - Sujit K Bhattacharya
- National Institute of Cholera and Enteric Diseases, P-33 CIT Road, Scheme XM, Beliaghata, Kolkata, India
| | - John D Clemens
- International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka 1212, Bangladesh.,UCLA Fielding School of Public Health, Los Angeles, CA 90095-1772, USA
| |
Collapse
|
17
|
O'Reilly PJ, Pant D, Shakya M, Basnyat B, Pollard AJ. Progress in the overall understanding of typhoid fever: implications for vaccine development. Expert Rev Vaccines 2020; 19:367-382. [PMID: 32238006 DOI: 10.1080/14760584.2020.1750375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Typhoid fever continues to have a substantial impact on human health, especially in Asia and sub-Saharan Africa. Access to safe water, and adequate sanitation and hygiene remain the cornerstone of prevention, but these are not widely available in many impoverished settings. The emergence of antibiotic resistance affects typhoid treatment and adds urgency to typhoid control efforts. Vaccines provide opportunities to prevent and control typhoid fever in endemic settings. AREAS COVERED Literature search was performed looking for evidence concerning the global burden of typhoid and strategies for the prevention and treatment of typhoid fever. Cost of illness, available typhoid and paratyphoid vaccines and cost-effectiveness were also reviewed. The objective was to provide a critical overview of typhoid fever, in order to assess the current understanding and potential future directions for typhoid treatment and control. EXPERT COMMENTARY Our understanding of typhoid burden and methods of prevention has grown over recent years. However, typhoid fever still has a significant impact on health in low and middle-income countries. Introduction of typhoid conjugate vaccines to the immunization schedule is expected to make a major contribution to control of typhoid fever in endemic countries, although vaccination alone is unlikely to eliminate the disease.
Collapse
Affiliation(s)
- Peter J O'Reilly
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre , Oxford, UK
| | - Dikshya Pant
- Department of Paediatrics, Patan Academy of Health Sciences, Patan Hospital , Kathmandu, Nepal
| | - Mila Shakya
- Oxford University Clinical Research Unit, Patan Academy of Health Sciences , Kathmandu, Nepal
| | - Buddha Basnyat
- Oxford University Clinical Research Unit, Patan Academy of Health Sciences , Kathmandu, Nepal
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre , Oxford, UK
| |
Collapse
|
18
|
Aljohani S, Hussein WM, Toth I, Simerska P. Carbohydrates in Vaccine Development. Curr Drug Deliv 2020; 16:609-617. [PMID: 31267872 DOI: 10.2174/1567201816666190702153612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/25/2019] [Accepted: 05/29/2019] [Indexed: 02/03/2023]
Abstract
Despite advances in the development of new vaccines, there are still some diseases with no vaccine solutions. Therefore, further efforts are required to more comprehensively discern the different antigenic components of these microorganisms on a molecular level. This review summarizes advancement in the development of new carbohydrate-based vaccines. Following traditional vaccine counterparts, the carbohydrate-based vaccines introduced a new approach in fighting infectious diseases. Carbohydrates have played various roles in the development of carbohydrate-based vaccines, which are described in this review, including carbohydrates acting as antigens, carriers or targeting moieties. Carbohydrate-based vaccines against infectious diseases, such as group A streptococcus, meningococcal meningitis and human immunodeficiency virus, are also discussed. A number of carbohydrate- based vaccines, such as Pneumovax 23, Menveo and Pentacel, have been successfully marketed in the past few years and there is a promising standpoint for many more to come in the near future.
Collapse
Affiliation(s)
- Salwa Aljohani
- The University of Queensland, School of Chemistry and Molecular Biosciences, Cooper Road, St. Lucia QLD 4072, Australia
| | - Waleed M Hussein
- The University of Queensland, School of Chemistry and Molecular Biosciences, Cooper Road, St. Lucia QLD 4072, Australia
| | - Istvan Toth
- The University of Queensland, School of Chemistry and Molecular Biosciences, Cooper Road, St. Lucia QLD 4072, Australia.,The University of Queensland, School of Pharmacy, Pharmacy Australia Centre of Excellence, Cornwall Street, Woolloongabba, QLD 4072, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Pavla Simerska
- The University of Queensland, School of Chemistry and Molecular Biosciences, Cooper Road, St. Lucia QLD 4072, Australia
| |
Collapse
|
19
|
Thakur R, Pathania P, Kaur N, Joshi V, Kondepudi KK, Suri CR, Rishi P. Prophylactic potential of cytolethal distending toxin B (CdtB) subunit of typhoid toxin against Typhoid fever. Sci Rep 2019; 9:18404. [PMID: 31804525 PMCID: PMC6895121 DOI: 10.1038/s41598-019-54690-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/18/2019] [Indexed: 02/04/2023] Open
Abstract
Typhoid fever caused by Salmonella enterica serovar Typhi (S.Typhi) continues to be a major problem, especially in developing countries. Due to the rapid emergence of multi-drug-resistant (MDR) strains, which limits the efficacy of conventional antibiotics as well as problems associated with the existing vaccines, efforts are being made to develop effective prophylactic agents. CdtB subunit of typhoid toxin was selected for assessing its vaccine potential due to its high conservation throughout the Typhi strains. In-vitro assessment of DNase activity of cloned and purified CdtB protein showed a significant decrease in the band intensity of DNA. The measure of metabolic activity and morphological alterations assessed using different cell lines in the presence of CdtB protein showed no significant signs of toxicity. These observations were further strengthened by cell cycle analysis, assessed by flow cytometry. Keeping these observations in mind, the immunoprotective potential of CdtB was assessed using S.Typhi induced mouse peritonitis model. A significant titer of IgG antibodies (>128000) against CdtB protein was recorded in the immunized mice by enzyme-linked immunosorbent assay (ELISA), which was also validated by immunoblotting. Active immunization with the protein protected 75% mice against a lethal dose of S.Typhi Ty2. The data indicated a significant (up to 5 log) reduction in the bacterial load in the spleen and liver of immunized-infected mice compared to control (unimmunized-infected) mice which might have resulted in the modulation of histoarchitecture of spleen and liver and the levels of cytokines (IL-6, TNF-α and IL-10) production; thereby indicating the effectiveness of the subunit. The observations deduced from the study give the proof of concept of immunogenic potential of protein. However, further studies involving the immunoreactivity of CdtB with the statistically significant number of sera samples obtained from the human patients would be helpful in establishing the relevance of CdtB protein in humans and for making the strategies to develop it as an effective vaccine candidate.
Collapse
Affiliation(s)
- Reena Thakur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Preeti Pathania
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Navneet Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Vattan Joshi
- Department of Microbiology, Panjab University, Chandigarh, India
| | | | | | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
20
|
Jeon HJ, Pak GD, Im J, Owusu-Dabo E, Adu-Sarkodie Y, Gassama Sow A, Bassiahi Soura A, Gasmelseed N, Keddy KH, Bjerregaard-Andersen M, Konings F, Aseffa A, Crump JA, Chon Y, Breiman RF, Park SE, Cruz Espinoza LM, Seo HJ, May J, Meyer CG, Andrews JR, Panzner U, von Kalckreuth V, Wierzba TF, Rakotozandrindrainy R, Dougan G, Levine MM, Hombach J, Kim JH, Clemens JD, Baker S, Marks F. Determining the Best Immunization Strategy for Protecting African Children Against Invasive Salmonella Disease. Clin Infect Dis 2019; 67:1824-1830. [PMID: 29746615 PMCID: PMC6260167 DOI: 10.1093/cid/ciy386] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/27/2018] [Indexed: 12/26/2022] Open
Abstract
Background The World Health Organization recently prequalified a typhoid conjugate vaccine (TCV), recommending its use in persons ≥6 months to 45 years residing in typhoid fever (TF)-endemic areas. We now need to consider how TCVs can have the greatest impact in the most vulnerable populations. Methods The Typhoid Fever Surveillance in Africa Program (TSAP) was a blood culture-based surveillance of febrile patients from defined populations presenting at healthcare facilities in 10 African countries. TF and invasive non-typhoidal Salmonella (iNTS) disease incidences were estimated for 0-10 year-olds in one-year age increments. Results Salmonella Typhi and iNTS were the most frequently isolated pathogens; 135 and 94 cases were identified, respectively. Analysis from three countries was excluded (incomplete person-years of observation (PYO) data). Thirty-seven of 123 TF cases (30.1%) and 71/90 iNTS disease cases (78.9%) occurred in children aged <5 years. No TF and 8/90 iNTS infections (8.9%) were observed in infants aged <9 months. The TF incidences (/100 000 PYO) for children aged <1 year and 1 to <2 years were 5 and 39, respectively; the highest incidence was 304 per 100 000 PYO in 4 to <5 year-olds. The iNTS disease incidence in the defined age groups ranged between 81 and 233 per 100 000 PYO, highest in 1 to <2 year-olds. TF and iNTS disease incidences were higher in West Africa. Conclusions High burden of TF detected in young children strengthens the need for TCV introduction. Given the concurrent iNTS disease burden, development of a trivalent vaccine against S. Typhi, S. Typhimurium, and S. Enteritidis may be timely in this region.
Collapse
Affiliation(s)
- Hyon Jin Jeon
- International Vaccine Institute, Seoul, Republic of Korea
| | - Gi Deok Pak
- International Vaccine Institute, Seoul, Republic of Korea
| | - Justin Im
- International Vaccine Institute, Seoul, Republic of Korea
| | - Ellis Owusu-Dabo
- Kumasi Center for Collaborative Research in Tropical Medicine, Kumasi, Ghana.,Departments of Global and International Health, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Yaw Adu-Sarkodie
- Departments of Clinical Microbiology, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Amy Gassama Sow
- Institute Pasteur de Dakar.,Université Cheikh Anta Diop de Dakar, Senegal
| | | | - Nagla Gasmelseed
- Faculty of Medicine, University of Gezira, Wad Medani, Sudan.,Faculty of Science, University of Hafr Al Batin, Saudi Arabia
| | - Karen H Keddy
- National Institute for Communicable Diseases, Johannesburg, South Africa.,Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Morten Bjerregaard-Andersen
- Bandim Health Project, Bissau, Guinea-Bissau.,Research Center for Vitamins and Vaccines, Copenhagen, Denmark
| | - Frank Konings
- International Vaccine Institute, Seoul, Republic of Korea
| | - Abraham Aseffa
- Armauer Hansen Research Institute, ALERT Campus, Addis Ababa, Ethiopia
| | - John A Crump
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania.,Division of Infectious Diseases and International Health, Duke University Medical Center.,Duke Global Health Institute, Duke University, Durham, North Carolina.,Centre for International Health, University of Otago, Dunedin, New Zealand
| | - Yun Chon
- International Vaccine Institute, Seoul, Republic of Korea
| | | | - Se Eun Park
- International Vaccine Institute, Seoul, Republic of Korea.,Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | | | - Hye Jin Seo
- International Vaccine Institute, Seoul, Republic of Korea
| | - Jürgen May
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
| | - Christian G Meyer
- Institute of Tropical Medicine, Eberhard-Karls University Tübingen, Germany.,Duy Tan University, Da Nang, Vietnam
| | - Jason R Andrews
- Division of Infectious Diseases and Geographic Medicine, Stanford University, California
| | - Ursula Panzner
- International Vaccine Institute, Seoul, Republic of Korea
| | | | | | | | - Gordon Dougan
- Department of Medicine, University of Cambridge, United Kingdom
| | - Myron M Levine
- Department of Medicine, University of Maryland School of Medicine, Baltimore
| | | | - Jerome H Kim
- International Vaccine Institute, Seoul, Republic of Korea
| | - John D Clemens
- International Centre for Diarrheal Disease Research, Bangladesh, Dhaka.,Fielding School of Public Health, University of California, Los Angeles.,Korea University School of Medicine, Seoul, Republic of Korea
| | - Stephen Baker
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam.,Department of Medicine, University of Cambridge, United Kingdom
| | - Florian Marks
- International Vaccine Institute, Seoul, Republic of Korea.,Department of Medicine, University of Cambridge, United Kingdom
| |
Collapse
|
21
|
Micoli F, Del Bino L, Alfini R, Carboni F, Romano MR, Adamo R. Glycoconjugate vaccines: current approaches towards faster vaccine design. Expert Rev Vaccines 2019; 18:881-895. [PMID: 31475596 DOI: 10.1080/14760584.2019.1657012] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Over the last decades, glycoconjugate vaccines have been proven to be a successful strategy to prevent infectious diseases. Many diseases remain to be controlled, especially in developing countries, and emerging antibiotic-resistant bacteria present an alarming public-health threat. The increasing complexity of future vaccines, and the need to accelerate development processes have triggered the development of faster approaches to glycoconjugate vaccines design. Areas covered: This review provides an overview of recent progress in glycoconjugation technologies toward faster vaccine design. Expert opinion: Among the different emerging approaches, glycoengineering has the potential to combine glycan assembly and conjugation to carrier systems (such as proteins or outer membrane vesicles) in one step, resulting in a simplified manufacturing process and fewer analytical controls. Chemical and enzymatic strategies, and their automation can facilitate glycoepitope identification for vaccine design. Other approaches, such as the liposomal encapsulation of polysaccharides, potentially enable fast and easy combination of numerous antigens in the same formulation. Additional progress is envisaged in the near future, and some of these systems still need to be further validated in humans. In parallel, new strategies are needed to accelerate the vaccine development process, including the associated clinical trials, up to vaccine release onto the market.
Collapse
Affiliation(s)
- Francesca Micoli
- Technology Platform, GSK Vaccines Institute for Global Health s.r.l , Siena , Italy
| | | | - Renzo Alfini
- Technology Platform, GSK Vaccines Institute for Global Health s.r.l , Siena , Italy
| | | | | | | |
Collapse
|
22
|
Abstract
BACKGROUND Contemporary incidence estimates of typhoid fever are needed to guide policy decisions and control measures and to improve future epidemiological studies. METHODS We systematically reviewed 3 databases (Ovid Medline, PubMed, and Scopus) without restriction on age, country, language, or time for studies reporting the incidence of blood culture-confirmed typhoid fever. Outbreak, travel-associated, and passive government surveillance reports were excluded. We performed a meta-analysis using a random-effects model to calculate estimates of pooled incidence, stratifying by studies that reported the incidence of typhoid fever and those that estimated incidence by using multipliers. RESULTS Thirty-three studies were included in the analysis. There were 26 study sites from 16 countries reporting typhoid cases from population-based incidence studies, and 17 sites in 9 countries used multipliers to account for underascertainment in sentinel surveillance data. We identified Africa and Asia as regions with studies showing high typhoid incidence while noting considerable variation of typhoid incidence in time and place, including in consecutive years at the same location. Overall, more recent studies reported lower typhoid incidence compared to years prior to 2000. We identified variation in the criteria for collecting a blood culture, and among multiplier studies we identified a lack of a standardization for the types of multipliers being used to estimate incidence. CONCLUSIONS Typhoid fever incidence remains high at many sites. Additional and more accurate typhoid incidence studies are needed to support country decisions about typhoid conjugate vaccine adoption. Standardization of multiplier types applied in multiplier studies is recommended.
Collapse
Affiliation(s)
| | - Chuen Yen Hong
- Centre for International Health, University of Otago, New Zealand
| | - John A Crump
- Centre for International Health, University of Otago, New Zealand
| |
Collapse
|
23
|
Keddy KH, Smith AM, Sooka A, Tau NP, Ngomane HMP, Radhakrishnan A, Als D, Benson FG. The Burden of Typhoid Fever in South Africa: The Potential Impact of Selected Interventions. Am J Trop Med Hyg 2018; 99:55-63. [PMID: 30047360 PMCID: PMC6128358 DOI: 10.4269/ajtmh.18-0182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Typhoid fever is notifiable in South Africa but clinical notification is notoriously poor. South Africa has an estimated annual incidence rate of 0.1 cases per 100,000 population of culture-confirmed typhoid fever, decreased from 17 cases per 100,000 population in the 1980s. This work was undertaken to identify the reasons for this decrease and identify potential weaknesses that may result in an increase of observed cases. Culture-confirmed cases, with additional demographic and clinical data have been collected from selected sentinel sites since 2003. Data on contextual factors (gross domestic product [GDP], sanitation, female education, and childhood diarrhea mortality) were collected. National incidence rates of culture-confirmed typhoid fever have remained constant for the past 13 years, with the exception of an outbreak in 2005: incidence was 0.4 per 100,000 population. Paratyphoid fever remains a rare disease. Antimicrobial susceptibility data suggest resistance to ciprofloxacin and azithromycin is emerging. The South African population increased from 27.5 million in 1980 to 55.0 million in 2015: urbanization increased from 50% to 65%, GDP increased from United States Dollar (USD) $2,910 to USD $6,167, access to sanitation improved from 64.4% to 70.0% in the urban population and 26.4% to 60.5% in rural areas. Female literacy levels improved from 74.8% to 92.6% over the period. Improved socioeconomic circumstances in South Africa have been temporally associated with decreasing incidence rates of typhoid fever over a 35-year period. Ongoing challenges remain including potential for large outbreaks, a large immigrant population, and emerging antimicrobial resistance. Continued active surveillance is mandatory.
Collapse
Affiliation(s)
- Karen H Keddy
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Centre for Enteric Diseases, National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Anthony M Smith
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Centre for Enteric Diseases, National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Arvinda Sooka
- Centre for Enteric Diseases, National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Nomsa P Tau
- Centre for Enteric Diseases, National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Hlengiwe M P Ngomane
- Centre for Enteric Diseases, National Institute for Communicable Diseases, Johannesburg, South Africa
| | | | - Daina Als
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Canada
| | - Frew G Benson
- Gauteng Provincial Health Department, Johannesburg, South Africa
| |
Collapse
|
24
|
Meiring JE, Patel P, Patel P, Gordon MA. Typhoid conjugate vaccines: making vaccine history in Africa. Expert Rev Vaccines 2018; 17:673-676. [PMID: 29972655 DOI: 10.1080/14760584.2018.1496825] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- James E Meiring
- a Oxford Vaccine Group, Department of Paediatrics , Oxford University , Oxford , UK.,b Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine , Blantyre , Malawi
| | - Pratiksha Patel
- b Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine , Blantyre , Malawi
| | - Priyanka Patel
- b Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine , Blantyre , Malawi
| | - Melita A Gordon
- b Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine , Blantyre , Malawi
| |
Collapse
|
25
|
Abstract
BACKGROUND Typhoid fever and paratyphoid fever continue to be important causes of illness and death, particularly among children and adolescents in south-central and southeast Asia. Two typhoid vaccines are widely available, Ty21a (oral) and Vi polysaccharide (parenteral). Newer typhoid conjugate vaccines are at varying stages of development and use. The World Health Organization has recently recommended a Vi tetanus toxoid (Vi-TT) conjugate vaccine, Typbar-TCV, as the preferred vaccine for all ages. OBJECTIVES To assess the effects of vaccines for preventing typhoid fever. SEARCH METHODS In February 2018, we searched the Cochrane Infectious Diseases Group Specialized Register, CENTRAL, MEDLINE, Embase, LILACS, and mRCT. We also searched the reference lists of all included trials. SELECTION CRITERIA Randomized and quasi-randomized controlled trials (RCTs) comparing typhoid fever vaccines with other typhoid fever vaccines or with an inactive agent (placebo or vaccine for a different disease) in adults and children. Human challenge studies were not eligible. DATA COLLECTION AND ANALYSIS Two review authors independently applied inclusion criteria and extracted data, and assessed the certainty of the evidence using the GRADE approach. We computed vaccine efficacy per year of follow-up and cumulative three-year efficacy, stratifying for vaccine type and dose. The outcome addressed was typhoid fever, defined as isolation of Salmonella enterica serovar Typhi in blood. We calculated risk ratios (RRs) and efficacy (1 - RR as a percentage) with 95% confidence intervals (CIs). MAIN RESULTS In total, 18 RCTs contributed to the quantitative analysis in this review: 13 evaluated efficacy (Ty21a: 5 trials; Vi polysaccharide: 6 trials; Vi-rEPA: 1 trial; Vi-TT: 1 trial), and 9 reported on adverse events. All trials but one took place in typhoid-endemic countries. There was no information on vaccination in adults aged over 55 years of age, pregnant women, or travellers. Only one trial included data on children under two years of age.Ty21a vaccine (oral vaccine, three doses)A three-dose schedule of Ty21a vaccine probably prevents around half of typhoid cases during the first three years after vaccination (cumulative efficacy 2.5 to 3 years: 50%, 95% CI 35% to 61%, 4 trials, 235,239 participants, moderate-certainty evidence). These data include patients aged 3 to 44 years.Compared with placebo, this vaccine probably does not cause more vomiting, diarrhoea, nausea or abdominal pain (2 trials, 2066 participants; moderate-certainty evidence), headache, or rash (1 trial, 1190 participants; moderate-certainty evidence); however, fever (2 trials, 2066 participants; moderate-certainty evidence) is probably more common following vaccination.Vi polysaccharide vaccine (injection, one dose)A single dose of Vi polysaccharide vaccine prevents around two-thirds of typhoid cases in the first year after vaccination (year 1: 69%, 95% CI 63% to 74%; 3 trials, 99,979 participants; high-certainty evidence). In year 2, trial results were more variable, with the vaccine probably preventing between 45% and 69% of typhoid cases (year 2: 59%, 95% CI 45% to 69%; 4 trials, 194,969 participants; moderate-certainty evidence). These data included participants aged 2 to 55 years of age.The three-year cumulative efficacy of the vaccine may be around 55% (95% CI 30% to 70%; 11,384 participants, 1 trial; low-certainty evidence). These data came from a single trial conducted in South Africa in the 1980s in participants aged 5 to 15 years.Compared with placebo, this vaccine probably did not increase the incidence of fever (3 trials, 132,261 participants; moderate-certainty evidence) or erythema (3 trials, 132,261 participants; low-certainty evidence); however, swelling (3 trials, 1767 participants; moderate-certainty evidence) and pain at the injection site (1 trial, 667 participants; moderate-certainty evidence) were more common in the vaccine group.Vi-rEPA vaccine (two doses)Administration of two doses of the Vi-rEPA vaccine probably prevents between 50% and 96% of typhoid cases during the first two years after vaccination (year 1: 94%, 95% CI 75% to 99%; year 2: 87%, 95% CI 56% to 96%, 1 trial, 12,008 participants; moderate-certainty evidence). These data came from a single trial with children two to five years of age conducted in Vietnam.Compared with placebo, both the first and the second dose of this vaccine increased the risk of fever (1 trial, 12,008 and 11,091 participants, low-certainty evidence) and the second dose increase the incidence of swelling at the injection site (one trial, 11,091 participants, moderate-certainty evidence).Vi-TT vaccine (two doses)We are uncertain of the efficacy of administration of two doses of Vi-TT (PedaTyph) in typhoid cases in children during the first year after vaccination (year 1: 94%, 95% CI -1% to 100%, 1 trial, 1625 participants; very low-certainty evidence). These data come from a single cluster-randomized trial in children aged six months to 12 years and conducted in India. For single dose Vi-TT (Typbar-TCV), we found no efficacy trials evaluating the vaccine with natural exposure.There were no reported serious adverse effects in RCTs of any of the vaccines studied. AUTHORS' CONCLUSIONS The licensed Ty21a and Vi polysaccharide vaccines are efficacious in adults and children older than two years in endemic countries. The Vi-rEPA vaccine is just as efficacious, although data is only available for children. The new Vi-TT vaccine (PedaTyph) requires further evaluation to determine if it provides protection against typhoid fever. At the time of writing, there were only efficacy data from a human challenge setting in adults on the Vi-TT vaccine (Tybar), which clearly justify the ongoing field trials to evaluate vaccine efficacy.
Collapse
Affiliation(s)
- Rachael Milligan
- Liverpool School of Tropical MedicineCochrane Infectious Diseases GroupPembroke PlaceLiverpoolUKL3 5QA
| | - Mical Paul
- Rambam Health Care CampusDivision of Infectious DiseasesHa‐aliya 8 StHaifaIsrael33705
| | - Marty Richardson
- Liverpool School of Tropical MedicineCochrane Infectious Diseases GroupPembroke PlaceLiverpoolUKL3 5QA
| | - Ami Neuberger
- Rambam Health Care Campus and The Ruth and Bruce Rappaport Faculty of Medicine, Technion – Israel Institute of TechnologyDivision of Infectious DiseasesTel AvivIsrael
| |
Collapse
|
26
|
Parker AR, Bradley C, Harding S, Sánchez-Ramón S, Jolles S, Kiani-Alikhan S. Measurement and interpretation of Salmonella typhi Vi IgG antibodies for the assessment of adaptive immunity. J Immunol Methods 2018; 459:1-10. [PMID: 29800575 DOI: 10.1016/j.jim.2018.05.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/26/2018] [Accepted: 05/21/2018] [Indexed: 01/15/2023]
Abstract
Response to polysaccharide vaccination can be an invaluable tool for assessing functionality of the adaptive immune system. Measurement of antibodies raised in response to Pneumovax®23 is the current gold standard test, but there are significant challenges and constraints in both the measurement and interpretation of the response. An alternative polysaccharide vaccine approach (Salmonella typhi Vi capsule (ViCPS)) has been suggested. In the present article, we review current evidence for the measurement of ViCPS antibodies in the diagnosis of primary and secondary antibody deficiencies. In particular, we review emerging data suggesting their interpretation in combination with the response to Pneumovax®23 and comment upon the utility of these vaccines to assess humoral immune responses while receiving immunoglobulin replacement therapy (IGRT).
Collapse
Affiliation(s)
| | | | | | - Silvia Sánchez-Ramón
- Department of Clinical Immunology Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
| | - Sorena Kiani-Alikhan
- Department of Immunology, Barts and The London National Health Service Trust, London, UK
| |
Collapse
|
27
|
Hiyoshi H, Tiffany CR, Bronner DN, Bäumler AJ. Typhoidal Salmonella serovars: ecological opportunity and the evolution of a new pathovar. FEMS Microbiol Rev 2018; 42:527-541. [DOI: 10.1093/femsre/fuy024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/19/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
- Hirotaka Hiyoshi
- Department of Medial Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Connor R Tiffany
- Department of Medial Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Denise N Bronner
- Department of Medial Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medial Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
28
|
Zuckerman JN, Hatz C, Kantele A. Review of current typhoid fever vaccines, cross-protection against paratyphoid fever, and the European guidelines. Expert Rev Vaccines 2018; 16:1029-1043. [PMID: 28856924 DOI: 10.1080/14760584.2017.1374861] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Typhoid and paratyphoid fever remain a global health problem, which - in non-endemic countries - are mainly seen in travelers, particularly in VFRs (visiting friends and relatives), with occasional local outbreaks occurring. A rise in anti-microbial resistance emphasizes the role of preventive measures, especially vaccinations against typhoid and paratyphoid fever for travelers visiting endemic countries. Areas covered: This state-of-the-art review recapitulates the epidemiology and mechanisms of disease of typhoid and paratyphoid fever, depicts the perspective of non-endemic countries and travelers (VFRs), and collectively presents current European recommendations for typhoid fever vaccination. We provide a brief overview of available (and developmental) vaccines in Europe, present current data on cross-protection to S. Paratyphi, and aim to provide a background for typhoid vaccine decision-making in travelers. Expert commentary: European recommendations are not harmonized. Experts must assess vaccination of travelers based on current country-specific recommendations. Travel health practitioners should be aware of the issues surrounding vaccination of travelers and be motivated to increase awareness of typhoid and paratyphoid fever risks.
Collapse
Affiliation(s)
- Jane N Zuckerman
- a Department of Infection and Immunity , Royal Free London Travel Health and Immunisation Clinic , London , UK
| | - Christoph Hatz
- b Department of Medicine and Diagnostics , Swiss Tropical and Public Health Institute , Basel , Switzerland.,c Epidemiology, Biostatistics and Prevention Institute , University of Zurich , Zurich , Switzerland
| | - Anu Kantele
- d Department of Clinical Medicine , University of Helsinki , Helsinki , Finland.,e Inflammation Center, Division of Infectious Diseases , Helsinki University Hospital , Helsinki , Finland.,f Unit of Infectious Diseases , Karolinska Institutet , Stockholm , Sweden
| |
Collapse
|
29
|
Evans C, Bateman E, Steven R, Ponsford M, Cullinane A, Shenton C, Duthie G, Conlon C, Jolles S, Huissoon AP, Longhurst HJ, Rahman T, Scott C, Wallis G, Harding S, Parker AR, Ferry BL. Measurement of Typhi Vi antibodies can be used to assess adaptive immunity in patients with immunodeficiency. Clin Exp Immunol 2018; 192:292-301. [PMID: 29377063 PMCID: PMC5980364 DOI: 10.1111/cei.13105] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2017] [Indexed: 02/06/2023] Open
Abstract
Vaccine‐specific antibody responses are essential in the diagnosis of antibody deficiencies. Responses to Pneumovax II are used to assess the response to polysaccharide antigens, but interpretation may be complicated. Typhim Vi®, a polysaccharide vaccine for Salmonella typhoid fever, may be an additional option for assessing humoral responses in patients suspected of having an immunodeficiency. Here we report a UK multi‐centre study describing the analytical and clinical performance of a Typhi Vi immunoglobulin (Ig)G enzyme‐linked immunosorbent assay (ELISA) calibrated to an affinity‐purified Typhi Vi IgG preparation. Intra‐ and interassay imprecision was low and the assay was linear, between 7·4 and 574 U/ml (slope = 0·99–1·00; R2 > 0·99); 71% of blood donors had undetectable Typhi Vi IgG antibody concentrations. Of those with antibody concentrations > 7·4 U/ml, the concentration range was 7·7–167 U/ml. In antibody‐deficient patients receiving antibody replacement therapy the median Typhi Vi IgG antibody concentrations were < 25 U/ml. In vaccinated normal healthy volunteers, the median concentration post‐vaccination was 107 U/ml (range 31–542 U/ml). Eight of eight patients (100%) had post‐vaccination concentration increases of at least threefold and six of eight (75%) of at least 10‐fold. In an antibody‐deficient population (n = 23), only 30% had post‐vaccination concentration increases of at least threefold and 10% of at least 10‐fold. The antibody responses to Pneumovax II and Typhim Vi® correlated. We conclude that IgG responses to Typhim Vi® vaccination can be measured using the VaccZyme Salmonella typhi Vi IgG ELISA, and that measurement of these antibodies maybe a useful additional test to accompany Pneumovax II responses for the assessment of antibody deficiencies.
Collapse
Affiliation(s)
- C Evans
- Clinical Immunology Laboratory, Oxford University Hospitals Foundation Trust, Oxford, UK
| | - E Bateman
- Clinical Immunology Laboratory, Oxford University Hospitals Foundation Trust, Oxford, UK
| | - R Steven
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
| | - M Ponsford
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
| | - A Cullinane
- Clinical Immunology Laboratory, Oxford University Hospitals Foundation Trust, Oxford, UK
| | - C Shenton
- Clinical Immunology Laboratory, Oxford University Hospitals Foundation Trust, Oxford, UK
| | - G Duthie
- Infectious Disease Department, Oxford University Hospitals Foundation Trust, Oxford, UK
| | - C Conlon
- Infectious Disease Department, Oxford University Hospitals Foundation Trust, Oxford, UK
| | - S Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
| | - A P Huissoon
- West Midlands Primary Immunodeficiency Centre, Birmingham Heartlands Hospital, Birmingham, UK
| | - H J Longhurst
- Department of Immunology, Barts Health NHS Trust, London, UK
| | - T Rahman
- Department of Immunology, Barts Health NHS Trust, London, UK
| | - C Scott
- Department of Immunology, Barts Health NHS Trust, London, UK
| | - G Wallis
- Binding Site Group Limited, Birmingham, UK
| | - S Harding
- Binding Site Group Limited, Birmingham, UK
| | - A R Parker
- Binding Site Group Limited, Birmingham, UK
| | - B L Ferry
- Clinical Immunology Laboratory, Oxford University Hospitals Foundation Trust, Oxford, UK
| |
Collapse
|
30
|
Juel HB, Thomaides-Brears HB, Darton TC, Jones C, Jones E, Shrestha S, Sie R, Eustace A, Galal U, Kurupati P, Van TT, Thieu NTV, Baker S, Blohmke CJ, Pollard AJ. Salmonella Typhi Bactericidal Antibodies Reduce Disease Severity but Do Not Protect against Typhoid Fever in a Controlled Human Infection Model. Front Immunol 2018; 8:1916. [PMID: 29387052 PMCID: PMC5776093 DOI: 10.3389/fimmu.2017.01916] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 12/14/2017] [Indexed: 01/12/2023] Open
Abstract
Effective vaccines against Salmonella Typhi, a major cause of febrile illness in tropical regions, can have a significant effect as a disease control measure. Earlier work has shown that immunization with either of two Salmonella Typhi vaccines, licensed Ty21a or candidate M01ZH09, did not provide full immunity in a controlled human infection model. Here, we describe the human humoral immune responses to these oral vaccines and their functional role in protection after challenge with S. Typhi. Serum, obtained from healthy volunteers before and after vaccination with Ty21a or M01ZH09 or placebo and before and after oral challenge with wild-type S. Typhi, was assessed for bactericidal activity. Single-dose vaccination with M01ZH09 induced an increase in serum bactericidal antibodies (p = 0.001) while three doses of Ty21a did not. No association between bactericidal activity and protection against typhoid after challenge was seen in either vaccine arm. Bactericidal activity after vaccination correlated significantly with delayed disease onset (p = 0.013), lower bacterial burden (p = 0.006), and decreased disease severity scores (p = 0.021). Depletion of antibodies directed against lipopolysaccharide significantly reduced bactericidal activity (p = 0.009). We conclude that antibodies induced after ingestion of oral live-attenuated typhoid vaccines or after challenge with wild-type S. Typhi exhibit bactericidal activity. This bactericidal activity is mediated by anti-O:LPS antibodies and significantly reduces clinical symptoms but does not provide sterile immunity. This directs future vaccine studies toward other antigens or mechanisms of protection against typhoid.
Collapse
Affiliation(s)
- Helene B Juel
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, The NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom.,Statens Serum Institut, Copenhagen, Denmark
| | - Helena B Thomaides-Brears
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, The NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Thomas C Darton
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, The NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom.,Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Claire Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, The NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Elizabeth Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, The NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Sonu Shrestha
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, The NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Rebecca Sie
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, The NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Andrew Eustace
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, The NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Ushma Galal
- Nuffield Department of Primary Care Health Sciences, Clinical Trials Unit, University of Oxford, Oxford, United Kingdom
| | - Prathiba Kurupati
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Tan T Van
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Nga T V Thieu
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Stephen Baker
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,The Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christoph J Blohmke
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, The NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, The NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
31
|
Zhang GL, Wei MM, Song C, Ma YF, Zheng XJ, Xiong DC, Ye XS. Chemical synthesis and biological evaluation of penta- to octa- saccharide fragments of Vi polysaccharide fromSalmonella typhi. Org Chem Front 2018. [DOI: 10.1039/c8qo00471d] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The penta- to octa-saccharide fragments of Vi polysaccharide were synthesized efficiently, and the hexasaccharide might be the minimum epitope of Vi antigen based on ELISA analysis.
Collapse
Affiliation(s)
- Gao-Lan Zhang
- State Key Laboratory of Natural and Biomimetic Drugs
- School of Pharmaceutical Sciences
- Peking University
- Beijing 100191
- China
| | - Meng-Man Wei
- State Key Laboratory of Natural and Biomimetic Drugs
- School of Pharmaceutical Sciences
- Peking University
- Beijing 100191
- China
| | - Chengcheng Song
- School of Life Sciences
- Northeast Normal University
- Changchun 130024
- China
| | - Yu-Feng Ma
- State Key Laboratory of Natural and Biomimetic Drugs
- School of Pharmaceutical Sciences
- Peking University
- Beijing 100191
- China
| | - Xiu-Jing Zheng
- State Key Laboratory of Natural and Biomimetic Drugs
- School of Pharmaceutical Sciences
- Peking University
- Beijing 100191
- China
| | - De-Cai Xiong
- State Key Laboratory of Natural and Biomimetic Drugs
- School of Pharmaceutical Sciences
- Peking University
- Beijing 100191
- China
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs
- School of Pharmaceutical Sciences
- Peking University
- Beijing 100191
- China
| |
Collapse
|
32
|
Lee SJ, Benoun J, Sheridan BS, Fogassy Z, Pham O, Pham QM, Puddington L, McSorley SJ. Dual Immunization with SseB/Flagellin Provides Enhanced Protection against Salmonella Infection Mediated by Circulating Memory Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:1353-1361. [PMID: 28710253 PMCID: PMC5548602 DOI: 10.4049/jimmunol.1601357] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 06/19/2017] [Indexed: 01/09/2023]
Abstract
The development of a subunit Salmonella vaccine has been hindered by the absence of detailed information about antigenic targets of protective Salmonella-specific T and B cells. Recent studies have identified SseB as a modestly protective Ag in susceptible C57BL/6 mice, but the mechanism of protective immunity remains undefined. In this article, we report that simply combining Salmonella SseB with flagellin substantially enhances protective immunity, allowing immunized C57BL/6 mice to survive for up to 30 d following challenge with virulent bacteria. Surprisingly, the enhancing effect of flagellin did not require flagellin Ag targeting during secondary responses or recognition of flagellin by TLR5. Although coimmunization with flagellin did not affect SseB-specific Ab responses, it modestly boosted CD4 responses. In addition, protective immunity was effectively transferred in circulation to parabionts of immunized mice, demonstrating that tissue-resident memory is not required for vaccine-induced protection. Finally, protective immunity required host expression of IFN-γR but was independent of induced NO synthase expression. Taken together, these data indicate that Salmonella flagellin has unique adjuvant properties that improve SseB-mediated protective immunity provided by circulating memory.
Collapse
Affiliation(s)
- Seung-Joo Lee
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| | - Joseph Benoun
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| | - Brian S Sheridan
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Zachary Fogassy
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| | - Oanh Pham
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| | - Quynh-Mai Pham
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Lynn Puddington
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Stephen J McSorley
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616;
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| |
Collapse
|
33
|
Zhang GL, Yang L, Zhu J, Wei M, Yan W, Xiong DC, Ye XS. Synthesis and Antigenic Evaluation of Oligosaccharide Mimics of Vi Antigen from Salmonella typhi. Chemistry 2017. [DOI: 10.1002/chem.201702114] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Gao-Lan Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences; Peking University; Xue Yuan Road No. 38 Beijing 100191 China
| | - Lin Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences; Peking University; Xue Yuan Road No. 38 Beijing 100191 China
| | - Jingjing Zhu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences; Peking University; Xue Yuan Road No. 38 Beijing 100191 China
| | - Mengman Wei
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences; Peking University; Xue Yuan Road No. 38 Beijing 100191 China
| | - Wanjun Yan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences; Peking University; Xue Yuan Road No. 38 Beijing 100191 China
| | - De-Cai Xiong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences; Peking University; Xue Yuan Road No. 38 Beijing 100191 China
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences; Peking University; Xue Yuan Road No. 38 Beijing 100191 China
| |
Collapse
|
34
|
Schaballie H, Bosch B, Schrijvers R, Proesmans M, De Boeck K, Boon MN, Vermeulen F, Lorent N, Dillaerts D, Frans G, Moens L, Derdelinckx I, Peetermans W, Kantsø B, Jørgensen CS, Emonds MP, Bossuyt X, Meyts I. Fifth Percentile Cutoff Values for Antipneumococcal Polysaccharide and Anti- Salmonella typhi Vi IgG Describe a Normal Polysaccharide Response. Front Immunol 2017; 8:546. [PMID: 28553290 PMCID: PMC5427071 DOI: 10.3389/fimmu.2017.00546] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/24/2017] [Indexed: 11/21/2022] Open
Abstract
Background Serotype-specific antibody responses to unconjugated pneumococcal polysaccharide vaccine (PPV) evaluated by a World Health Organization (WHO)-standardized enzyme-linked immunosorbent assay (ELISA) are the gold standard for diagnosis of specific polysaccharide antibody deficiency (SAD). The American Academy of Allergy, Asthma and Immunology (AAAAI) has proposed guidelines to interpret the PPV response measured by ELISA, but these are based on limited evidence. Additionally, ELISA is costly and labor-intensive. Measurement of antibody response to Salmonella typhi (S. typhi) Vi vaccine and serum allohemagglutinins (AHA) have been suggested as alternatives. However, there are no large cohort studies and cutoff values are lacking. Objective To establish cutoff values for antipneumococcal polysaccharide antibody response, anti-S. typhi Vi antibody, and AHA. Methods One hundred healthy subjects (10–55 years) were vaccinated with PPV and S. typhi Vi vaccine. Blood samples were obtained prior to and 3–4 weeks after vaccination. Polysaccharide responses to 3 serotypes were measured by WHO ELISA and to 12 serotypes by an in-house bead-based multiplex assay. Anti-S. typhi Vi IgG were measured with a commercial ELISA kit. AHA were measured by agglutination method. Results Applying AAAAI criteria, 30% of healthy subjects had a SAD. Using serotype-specific fifth percentile (p5) cutoff values for postvaccination IgG and fold increase pre- over postvaccination, only 4% of subjects had SAD. One-sided 95% prediction intervals for anti-S. typhi Vi postvaccination IgG (≥11.2 U/ml) and fold increase (≥2) were established. Eight percent had a response to S. typhi Vi vaccine below these cutoffs. AHA titer p5 cutoffs were ½ for anti-B and ¼ for anti-A. Conclusion We establish reference cutoff values for interpretation of PPV response measured by bead-based assay, cutoff values for S. typhi Vi vaccine responses, and normal values for AHA. For the first time, the intraindividual consistency of all three methods is studied in a large cohort.
Collapse
Affiliation(s)
- Heidi Schaballie
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Barbara Bosch
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium.,St. Giles Laboratory of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Rik Schrijvers
- Department Microbiology and Immunology, KU Leuven - University of Leuven, Leuven, Belgium
| | - Marijke Proesmans
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Kris De Boeck
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Mieke Nelly Boon
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | | | - Natalie Lorent
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Doreen Dillaerts
- Department Microbiology and Immunology, KU Leuven - University of Leuven, Leuven, Belgium
| | - Glynis Frans
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Leen Moens
- Department Microbiology and Immunology, KU Leuven - University of Leuven, Leuven, Belgium
| | - Inge Derdelinckx
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Willy Peetermans
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Bjørn Kantsø
- Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | | | - Marie-Paule Emonds
- Histocompatibility and Immunogenetic Laboratory, Red Cross Flanders, Mechelen, Belgium
| | - Xavier Bossuyt
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Isabelle Meyts
- Department Microbiology and Immunology, KU Leuven - University of Leuven, Leuven, Belgium
| |
Collapse
|
35
|
Xiong K, Zhu C, Chen Z, Zheng C, Tan Y, Rao X, Cong Y. Vi Capsular Polysaccharide Produced by Recombinant Salmonella enterica Serovar Paratyphi A Confers Immunoprotection against Infection by Salmonella enterica Serovar Typhi. Front Cell Infect Microbiol 2017; 7:135. [PMID: 28484685 PMCID: PMC5401900 DOI: 10.3389/fcimb.2017.00135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/31/2017] [Indexed: 11/28/2022] Open
Abstract
Enteric fever is predominantly caused by Salmonella enterica serovar Typhi and Salmonella enterica serovar Paratyphi A, and accounts for an annual global incidence of 26.9 millions. In recent years, the rate of S. Paratyphi A infection has progressively increased. Currently licensed vaccines for typhoid fever, live Ty21a vaccine, Vi subunit vaccine, and Vi-conjugate vaccine, confer inadequate cross immunoprotection against enteric fever caused by S. Paratyphi A. Therefore, development of bivalent vaccines against enteric fever is urgently required. The immunogenic Vi capsular polysaccharide is characteristically produced in S. Typhi, but it is absent in S. Paratyphi A. We propose that engineering synthesis of Vi in S. Paratyphi A live-attenuated vaccine may expand its protection range to cover S. Typhi. In this study, we cloned the viaB locus, which contains 10 genes responsible for Vi biosynthesis, and integrated into the chromosome of S. Paratyphi A CMCC 50093. Two virulence loci, htrA and phoPQ, were subsequently deleted to achieve a Vi-producing attenuated vaccine candidate. Our data showed that, despite more than 200 passages, the viaB locus was stably maintained in the chromosome of S. Paratyphi A and produced the Vi polysaccharide. Nasal immunization of the vaccine candidate stimulated high levels of Vi-specific and S. Paratyphi A-specific antibodies in mice sera as well as total sIgA in intestinal contents, and showed significant protection against wild-type challenge of S. Paratyphi A or S. Typhi. Our study show that the Vi-producing attenuated S. Paratyphi A is a promising bivalent vaccine candidate for the prevention of enteric fever.
Collapse
Affiliation(s)
- Kun Xiong
- Department of Microbiology, Third Military Medical UniversityChongqing, China
| | - Chunyue Zhu
- Outpatient Department of 95851 Unit of PLANanjing, China
| | - Zhijin Chen
- Department of Microbiology, Third Military Medical UniversityChongqing, China
| | - Chunping Zheng
- Department of Microbiology, Third Military Medical UniversityChongqing, China
| | - Yong Tan
- Department of Microbiology, Third Military Medical UniversityChongqing, China
| | - Xiancai Rao
- Department of Microbiology, Third Military Medical UniversityChongqing, China
| | - Yanguang Cong
- Department of Microbiology, Third Military Medical UniversityChongqing, China
| |
Collapse
|
36
|
Khan MI, Franco-Paredes C, Sahastrabuddhe S, Ochiai RL, Mogasale V, Gessner BD. Barriers to typhoid fever vaccine access in endemic countries. Res Rep Trop Med 2017; 8:37-44. [PMID: 30050343 PMCID: PMC6034652 DOI: 10.2147/rrtm.s97309] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Typhoid vaccines have been available as a means of disease control and prevention since 1896; however, their use as a routine tool for disease prevention in endemic settings has been hampered because of: 1) insufficient data on disease burden particularly regarding the lack of health care access in the poorest communities affected by typhoid; 2) limitations of the typhoid vaccine, such as shorter duration of protection, moderate efficacy in young children, and no efficacy for infants; 3) inadequate evidence on potential economic benefits when used for a larger population; 4) neglect in favor of alternative interventions that require massive infrastructure; 5) no financial support or commitment regarding vaccine delivery cost; 6) ambivalence about whether to invest in water and sanitation hygiene versus the vaccine; and 7) clarity on global policy for country adoption. If current typhoid-protein conjugate vaccines live up to their promise of higher efficacy, longer duration of protection, and efficacy in young children, typhoid vaccine use will be a critical component of short- and medium-term disease control strategies. Typhoid control could be accelerated if the global framework includes plans for accelerated introduction of the conjugate typhoid vaccine in developing countries.
Collapse
Affiliation(s)
- M Imran Khan
- Center of Excellence in Women and Child Health, The Aga Khan University, Karachi, Pakistan,
| | - Carlos Franco-Paredes
- Hospital Infantil de México, Federico Gómez, México DF., Mexico.,Phoebe Putney Memorial Hospital, Albany, GA, USA
| | | | | | | | | |
Collapse
|
37
|
Dyson ZA, Thanh DP, Bodhidatta L, Mason CJ, Srijan A, Rabaa MA, Vinh PV, Thanh TH, Thwaites GE, Baker S, Holt KE. Whole Genome Sequence Analysis of Salmonella Typhi Isolated in Thailand before and after the Introduction of a National Immunization Program. PLoS Negl Trop Dis 2017; 11:e0005274. [PMID: 28060810 PMCID: PMC5245908 DOI: 10.1371/journal.pntd.0005274] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/19/2017] [Accepted: 12/20/2016] [Indexed: 12/31/2022] Open
Abstract
Vaccines against Salmonella Typhi, the causative agent of typhoid fever, are commonly used by travellers, however, there are few examples of national immunization programs in endemic areas. There is therefore a paucity of data on the impact of typhoid immunization programs on localised populations of S. Typhi. Here we have used whole genome sequencing (WGS) to characterise 44 historical bacterial isolates collected before and after a national typhoid immunization program that was implemented in Thailand in 1977 in response to a large outbreak; the program was highly effective in reducing typhoid case numbers. Thai isolates were highly diverse, including 10 distinct phylogenetic lineages or genotypes. Novel prophage and plasmids were also detected, including examples that were previously only reported in Shigella sonnei and Escherichia coli. The majority of S. Typhi genotypes observed prior to the immunization program were not observed following it. Post-vaccine era isolates were more closely related to S. Typhi isolated from neighbouring countries than to earlier Thai isolates, providing no evidence for the local persistence of endemic S. Typhi following the national immunization program. Rather, later cases of typhoid appeared to be caused by the occasional importation of common genotypes from neighbouring Vietnam, Laos, and Cambodia. These data show the value of WGS in understanding the impacts of vaccination on pathogen populations and provide support for the proposal that large-scale typhoid immunization programs in endemic areas could result in lasting local disease elimination, although larger prospective studies are needed to test this directly. Typhoid fever is a systemic infection caused by the bacterium Salmonella Typhi. Typhoid fever is associated with inadequate hygiene in low-income settings and a lack of sanitation infrastructure. A sustained outbreak of typhoid fever occurred in Thailand in the 1970s, which peaked in 1975–1976. In response to this typhoid fever outbreak the government of Thailand initiated an immunization program, which resulted in a dramatic reduction in the number of typhoid cases in Thailand. To better understand the population of S. Typhi circulating in Thailand at this time, as well as the impact of the immunization program on the pathogen population, we sequenced the genomes of 44 S. Typhi obtained from hospitals in Thailand before and after the immunization program. The genome sequences showed that isolates of S. Typhi bacteria isolated from post-immunization era typhoid cases were likely imported from neighbouring countries, rather than strains that have persisted in Thailand throughout the immunization period. Our work provides the first historical insights into S. Typhi in Thailand during the 1970s, and provides a model for the impact of immunization on S. Typhi populations.
Collapse
Affiliation(s)
- Zoe A. Dyson
- Centre for Systems Genomics, University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
- * E-mail: (KEH); (ZAD)
| | - Duy Pham Thanh
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Ladaporn Bodhidatta
- Department of Enteric Diseases, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Carl Jeffries Mason
- Department of Enteric Diseases, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Apichai Srijan
- Department of Enteric Diseases, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Maia A. Rabaa
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Oxford University, Oxford, United Kingdom
| | - Phat Voong Vinh
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Tuyen Ha Thanh
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Guy E. Thwaites
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Oxford University, Oxford, United Kingdom
| | - Stephen Baker
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Oxford University, Oxford, United Kingdom
- The London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Kathryn E. Holt
- Centre for Systems Genomics, University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
- * E-mail: (KEH); (ZAD)
| |
Collapse
|
38
|
Hu X, Chen Z, Xiong K, Wang J, Rao X, Cong Y. Vi capsular polysaccharide: Synthesis, virulence, and application. Crit Rev Microbiol 2016; 43:440-452. [PMID: 27869515 DOI: 10.1080/1040841x.2016.1249335] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Vi capsular polysaccharide, a linear homopolymer of α-1,4-linked N-acetylgalactosaminuronate, is characteristically produced by Salmonella enterica serovar Typhi. The Vi capsule covers the surface of the producing bacteria and serves as an virulence factor via inhibition of complement-mediated killing and promoting resistance against phagocytosis. Furthermore, Vi also represents a predominant protective antigen and plays a key role in the development of vaccines against typhoid fever. Herein, we reviewed the latest advances associated with the Vi polysaccharide, from its synthesis and transport within bacterial cells, mechanisms involved in virulence, immunological characteristics, and applications in vaccine, as well as its purification and detection methods.
Collapse
Affiliation(s)
- Xiaomei Hu
- a Department of Microbiology , Third Military Medical University , Chongqing , China
| | - Zhijin Chen
- a Department of Microbiology , Third Military Medical University , Chongqing , China
| | - Kun Xiong
- a Department of Microbiology , Third Military Medical University , Chongqing , China
| | - Jing Wang
- a Department of Microbiology , Third Military Medical University , Chongqing , China
| | - Xiancai Rao
- a Department of Microbiology , Third Military Medical University , Chongqing , China
| | - Yanguang Cong
- a Department of Microbiology , Third Military Medical University , Chongqing , China
| |
Collapse
|
39
|
Glycolipid substrates for ABC transporters required for the assembly of bacterial cell-envelope and cell-surface glycoconjugates. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:1394-1403. [PMID: 27793707 DOI: 10.1016/j.bbalip.2016.10.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 01/07/2023]
Abstract
Glycoconjugates, molecules that contain sugar components, are major components of the cell envelopes of bacteria and cover much of their exposed surfaces. These molecules are involved in interactions with the surrounding environment and, in pathogens, play critical roles in the interplay with the host immune system. Despite the remarkable diversity in glycoconjugate structures, most are assembled by glycosyltransferases that act on lipid acceptors at the cytosolic membrane. The resulting glycolipids are then transported to the cell surface in processes that frequently begin with ATP-binding cassette transporters. This review summarizes current understanding of the structure and biosynthesis of glycolipid substrates and the structure and functions of their transporters. This article is part of a Special Issue entitled: Bacterial Lipids edited by Russell E. Bishop.
Collapse
|
40
|
Zhao X, Chen Z, Gu G, Guo Z. Recent advances in the research of bacterial glucuronosyltransferases. J Carbohydr Chem 2016. [DOI: 10.1080/07328303.2016.1205597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
41
|
Sánchez-Ramón S, de Gracia J, García-Alonso AM, Rodríguez Molina JJ, Melero J, de Andrés A, García Ruiz de Morales JM, Ferreira A, Ocejo-Vinyals JG, Cid JJ, García Martínez JM, Lasheras T, Vargas ML, Gil-Herrera J, García Rodríguez MC, Castañer JL, González Granado LI, Allende LM, Soler-Palacin P, Herráiz L, López Hoyos M, Bellón JM, Silva G, Gurbindo DM, Carbone J, Rodríguez-Sáinz C, Matamoros N, Parker AR, Fernández-Cruz E. Multicenter study for the evaluation of the antibody response against salmonella typhi Vi vaccination (EMPATHY) for the diagnosis of Anti-polysaccharide antibody production deficiency in patients with primary immunodeficiency. Clin Immunol 2016; 169:80-84. [PMID: 27236002 DOI: 10.1016/j.clim.2016.05.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 02/07/2016] [Accepted: 05/21/2016] [Indexed: 11/28/2022]
Affiliation(s)
- S Sánchez-Ramón
- Clinical Immunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| | - J de Gracia
- Pneumology, Hospital Universitari Vall d'Hebrón, Barcelona, Spain
| | - A M García-Alonso
- Immunology, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - J J Rodríguez Molina
- Clinical Immunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - J Melero
- Immunology, Hospital Infanta Cristina, Badajoz, Spain
| | - A de Andrés
- Immunology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | - A Ferreira
- Immunology, Hospital Universitario La Paz, Madrid, Spain
| | | | - J J Cid
- Immunology, Hospital Juan Canalejo, La Coruña, Spain
| | | | - T Lasheras
- Pneumology, Hospital Universitari Vall d'Hebrón, Barcelona, Spain
| | - M L Vargas
- Immunology, Hospital Infanta Cristina, Badajoz, Spain
| | - J Gil-Herrera
- Clinical Immunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | - J L Castañer
- Immunology, Hospital Universitario La Paz, Madrid, Spain
| | - L I González Granado
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Pediatrics, Hospital 12 de Octubre, Madrid, Spain
| | - L M Allende
- Immunology, Hospital 12 de octubre, Madrid, Spain
| | - P Soler-Palacin
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - L Herráiz
- Clinical Immunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - M López Hoyos
- Immunology, Hospital Marqués de Valdecilla, Santander, Spain
| | - J M Bellón
- Statistics, Hospital General Universitario Gregorio Marañón, Spain
| | - G Silva
- Immunology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - D M Gurbindo
- Immunopediatrics, Hospital Maternoinfantil de O'Donnell, Madrid, Spain
| | - J Carbone
- Clinical Immunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - C Rodríguez-Sáinz
- Clinical Immunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - N Matamoros
- Immunology, Hospital Son Espases, Palma de Mallorca, Spain
| | - A R Parker
- The Binding Site Group Ltd, Birmingham, UK
| | - E Fernández-Cruz
- Clinical Immunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | |
Collapse
|
42
|
Blohmke CJ, Darton TC, Jones C, Suarez NM, Waddington CS, Angus B, Zhou L, Hill J, Clare S, Kane L, Mukhopadhyay S, Schreiber F, Duque-Correa MA, Wright JC, Roumeliotis TI, Yu L, Choudhary JS, Mejias A, Ramilo O, Shanyinde M, Sztein MB, Kingsley RA, Lockhart S, Levine MM, Lynn DJ, Dougan G, Pollard AJ. Interferon-driven alterations of the host's amino acid metabolism in the pathogenesis of typhoid fever. J Exp Med 2016; 213:1061-77. [PMID: 27217537 PMCID: PMC4886356 DOI: 10.1084/jem.20151025] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 04/08/2016] [Indexed: 12/30/2022] Open
Abstract
Enteric fever, caused by Salmonella enterica serovar Typhi, is an important public health problem in resource-limited settings and, despite decades of research, human responses to the infection are poorly understood. In 41 healthy adults experimentally infected with wild-type S. Typhi, we detected significant cytokine responses within 12 h of bacterial ingestion. These early responses did not correlate with subsequent clinical disease outcomes and likely indicate initial host-pathogen interactions in the gut mucosa. In participants developing enteric fever after oral infection, marked transcriptional and cytokine responses during acute disease reflected dominant type I/II interferon signatures, which were significantly associated with bacteremia. Using a murine and macrophage infection model, we validated the pivotal role of this response in the expression of proteins of the host tryptophan metabolism during Salmonella infection. Corresponding alterations in tryptophan catabolites with immunomodulatory properties in serum of participants with typhoid fever confirmed the activity of this pathway, and implicate a central role of host tryptophan metabolism in the pathogenesis of typhoid fever.
Collapse
Affiliation(s)
- Christoph J. Blohmke
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, England, UK
| | - Thomas C. Darton
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, England, UK
| | - Claire Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, England, UK
| | - Nicolas M. Suarez
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Claire S. Waddington
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, England, UK
| | - Brian Angus
- Nuffield Department of Medicine, University of Oxford, OX1 2JD, England, UK
| | - Liqing Zhou
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, England, UK
| | - Jennifer Hill
- Microbial Pathogenesis Group, The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, England, UK
| | - Simon Clare
- Microbial Pathogenesis Group, The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, England, UK
| | - Leanne Kane
- Microbial Pathogenesis Group, The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, England, UK
| | - Subhankar Mukhopadhyay
- Microbial Pathogenesis Group, The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, England, UK
| | - Fernanda Schreiber
- Microbial Pathogenesis Group, The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, England, UK
| | - Maria A. Duque-Correa
- Microbial Pathogenesis Group, The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, England, UK
| | - James C. Wright
- Proteomic Mass Spectrometry, The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, England, UK
| | | | - Lu Yu
- Proteomic Mass Spectrometry, The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, England, UK
| | - Jyoti S. Choudhary
- Proteomic Mass Spectrometry, The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, England, UK
| | - Asuncion Mejias
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Octavio Ramilo
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Milensu Shanyinde
- Nuffield Department of Primary Care Health Sciences, University of Oxford, OX1 2JD, England, UK
| | - Marcelo B. Sztein
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Robert A. Kingsley
- Microbial Pathogenesis Group, The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, England, UK
| | - Stephen Lockhart
- Emergent Product Development UK, Emergent BioSolutions, Wokingham RG41 5TU, England, UK
| | - Myron M. Levine
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201
| | - David J. Lynn
- EMBL Australia Group, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia,School of Medicine, Flinders University, Bedford Park, SA 5042, Australia
| | - Gordon Dougan
- Microbial Pathogenesis Group, The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, England, UK
| | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, England, UK
| |
Collapse
|
43
|
Unique lipid anchor attaches Vi antigen capsule to the surface of Salmonella enterica serovar Typhi. Proc Natl Acad Sci U S A 2016; 113:6719-24. [PMID: 27226298 DOI: 10.1073/pnas.1524665113] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Polysaccharide capsules are surface structures that are critical for the virulence of many Gram-negative pathogenic bacteria. Salmonella enterica serovar Typhi is the etiological agent of typhoid fever. It produces a capsular polysaccharide known as "Vi antigen," which is composed of nonstoichiometrically O-acetylated α-1,4-linked N-acetylgalactosaminuronic acid residues. This glycan is a component of currently available vaccines. The genetic locus for Vi antigen production is also present in soil bacteria belonging to the genus Achromobacter Vi antigen assembly follows a widespread general strategy with a characteristic glycan export step involving an ATP-binding cassette transporter. However, Vi antigen producers lack the enzymes that build the conserved terminal glycolipid characterizing other capsules using this method. Achromobacter species possess a Vi antigen-specific depolymerase enzyme missing in S enterica Typhi, and we exploited this enzyme to isolate acylated Vi antigen termini. Mass spectrometry analysis revealed a reducing terminal N-acetylhexosamine residue modified with two β-hydroxyl acyl chains. This terminal structure resembles one half of lipid A, the hydrophobic portion of bacterial lipopolysaccharides. The VexE protein encoded in the Vi antigen biosynthesis locus shares similarity with LpxL, an acyltransferase from lipid A biosynthesis. In the absence of VexE, Vi antigen is produced, but its physical properties are altered, its export is impaired, and a Vi capsule structure is not assembled on the cell surface. The structure of the lipidated terminus dictates a unique assembly mechanism and has potential implications in pathogenesis and vaccine production.
Collapse
|
44
|
Entirely Carbohydrate-Based Vaccines: An Emerging Field for Specific and Selective Immune Responses. Vaccines (Basel) 2016; 4:vaccines4020019. [PMID: 27213458 PMCID: PMC4931636 DOI: 10.3390/vaccines4020019] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 12/16/2022] Open
Abstract
Carbohydrates are regarded as promising targets for vaccine development against infectious disease because cell surface glycans on many infectious agents are attributed to playing an important role in pathogenesis. In addition, oncogenic transformation of normal cells, in many cases, is associated with aberrant glycosylation of the cell surface glycan generating tumor associated carbohydrate antigens (TACAs). Technological advances in glycobiology have added a new dimension to immunotherapy when considering carbohydrates as key targets in developing safe and effective vaccines to combat cancer, bacterial infections, viral infections, etc. Many consider effective vaccines induce T-cell dependent immunity with satisfactory levels of immunological memory that preclude recurrence. Unfortunately, carbohydrates alone are poorly immunogenic as they do not bind strongly to the MHCII complex and thus fail to elicit T-cell immunity. To increase immunogenicity, carbohydrates have been conjugated to carrier proteins, which sometimes can impede carbohydrate specific immunity as peptide-based immune responses can negate antibodies directed at the targeted carbohydrate antigens. To overcome many challenges in using carbohydrate-based vaccine design and development approaches targeting cancer and other diseases, zwitterionic polysaccharides (ZPSs), isolated from the capsule of commensal anaerobic bacteria, will be discussed as promising carriers of carbohydrate antigens to achieve desired immunological responses.
Collapse
|
45
|
Principi N, Esposito S. Preventing invasive salmonellosis in children through vaccination. Expert Rev Vaccines 2016; 15:897-905. [PMID: 27140662 DOI: 10.1080/14760584.2016.1183484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Vaccination is an important strategy to control endemic enteric fever (EF) and to interrupt transmission during outbreaks. The main aim of this paper is to discuss the efficacy of available EF vaccines in children and to highlight novel vaccination possibilities against EF and non-typhoid invasive salmonelloses. AREAS COVERED Two types of typhoid vaccines are presently available in the industrialized world. One of these vaccines is administered parenterally and is based on the virulence-associated (Vi) capsular polysaccaride of Salmonella typhi. The second vaccine is based on a live attenuated strain of the pathogen and is given orally. In addition, a Vi-tetanus toxoid conjugated vaccine is currently licensed in India; however, it is not available anywhere else. Expert commentary: Unfortunately, only typhoid fever is addressed by the currently licensed typhoid vaccines. Moreover, they are unsuitable for infants and remain a possible aid for reducing the risk of EF only in older subjects. They should be used in developing countries with endemic EF. New vaccines able to confer long-term protection to subjects in the first years of life and those with immature immune systems could significantly reduce incidence rates of EF in younger children. Vi-conjugate preparations are promising solutions in this regard.
Collapse
Affiliation(s)
- Nicola Principi
- a Paediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation , Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Susanna Esposito
- a Paediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation , Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| |
Collapse
|
46
|
Ramachandran G, Tennant SM, Boyd MA, Wang JY, Tulapurkar ME, Pasetti MF, Levine MM, Simon R. Functional Activity of Antibodies Directed towards Flagellin Proteins of Non-Typhoidal Salmonella. PLoS One 2016; 11:e0151875. [PMID: 26998925 PMCID: PMC4801366 DOI: 10.1371/journal.pone.0151875] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/04/2016] [Indexed: 11/23/2022] Open
Abstract
Non-typhoidal Salmonella (NTS) serovars Typhimurium and Enteritidis are major causes of invasive bacterial infections in children under 5 years old in sub-Saharan Africa, with case fatality rates of ~20%. There are no licensed NTS vaccines for humans. Vaccines that induce antibodies against a Salmonella Typhi surface antigen, Vi polysaccharide, significantly protect humans against typhoid fever, establishing that immune responses to Salmonella surface antigens can be protective. Flagella proteins, abundant surface antigens in Salmonella serovars that cause human disease, are also powerful immunogens, but the functional capacity of elicited anti-flagellar antibodies and their role in facilitating bacterial clearance has been unclear. We examined the ability of anti-flagellar antibodies to mediate microbial killing by immune system components in-vitro and assessed their role in protecting mice against invasive Salmonella infection. Polyclonal (hyperimmune sera) and monoclonal antibodies raised against phase 1 flagellin proteins of S. Enteritidis and S. Typhimurium facilitated bacterial uptake and killing of the homologous serovar pathogen by phagocytes. Polyclonal anti-flagellar antibodies accompanied by complement also achieved direct bacterial killing. Serum bactericidal activity was restricted to Salmonella serovars expressing the same flagellin used as immunogen. Notably, individual anti-flagellin monoclonal antibodies with complement were not bactericidal, but this biological activity was restored when different monoclonal anti-flagellin antibodies were combined. Passive transfer immunization with a monoclonal IgG antibody specific for phase 1 flagellin from S. Typhimurium protected mice against lethal challenge with a representative African invasive S. Typhimurium strain. These findings have relevance for the use of flagellin proteins in NTS vaccines, and confirm the role of anti-flagellin antibodies as mediators of protective immunity.
Collapse
Affiliation(s)
- Girish Ramachandran
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Sharon M. Tennant
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Mary A. Boyd
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jin Y. Wang
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Mohan E. Tulapurkar
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Division of Pulmonary and Critical Care, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Marcela F. Pasetti
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Myron M. Levine
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Raphael Simon
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
47
|
Trabbic KR, Bourgault JP, Shi M, Clark M, Andreana PR. Immunological evaluation of the entirely carbohydrate-based Thomsen-Friedenreich – PS B conjugate. Org Biomol Chem 2016; 14:3350-5. [DOI: 10.1039/c6ob00176a] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PS B, a CD4+T-cell stimulating zwitterionic polysaccharide fromB. fragilis, was conjugated with aminooxy TF tumor antigen. Immunization revealed Ab specificity to TF. FACS revealed Ab binding to MCF-7 but not HCT-116 cells.
Collapse
Affiliation(s)
- Kevin R. Trabbic
- Department of Chemistry and Biochemistry
- University of Toledo
- Toledo
- USA
| | | | - Mengchao Shi
- Department of Chemistry and Biochemistry
- University of Toledo
- Toledo
- USA
| | - Matthew Clark
- Department of Chemistry and Biochemistry
- University of Toledo
- Toledo
- USA
| | - Peter R. Andreana
- Department of Chemistry and Biochemistry
- University of Toledo
- Toledo
- USA
| |
Collapse
|
48
|
Kinnear CL, Strugnell RA. Vaccination Method Affects Immune Response and Bacterial Growth but Not Protection in the Salmonella Typhimurium Animal Model of Typhoid. PLoS One 2015; 10:e0141356. [PMID: 26509599 PMCID: PMC4625024 DOI: 10.1371/journal.pone.0141356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 10/07/2015] [Indexed: 01/22/2023] Open
Abstract
Understanding immune responses elicited by vaccines, together with immune responses required for protection, is fundamental to designing effective vaccines and immunisation programs. This study examines the effects of the route of administration of a live attenuated vaccine on its interactions with, and stimulation of, the murine immune system as well as its ability to increase survival and provide protection from colonisation by a virulent challenge strain. We assess the effect of administration method using the murine model for typhoid, where animals are infected with S. Typhimurium. Mice were vaccinated either intravenously or orally with the same live attenuated S. Typhimurium strain and data were collected on vaccine strain growth, shedding and stimulation of antibodies and cytokines. Following vaccination, mice were challenged with a virulent strain of S. Typhimurium and the protection conferred by the different vaccination routes was measured in terms of challenge suppression and animal survival. The main difference in immune stimulation found in this study was the development of a secretory IgA response in orally-vaccinated mice, which was absent in IV vaccinated mice. While both strains showed similar protection in terms of challenge suppression in systemic organs (spleen and liver) as well as survival, they differed in terms of challenge suppression of virulent pathogens in gut-associated organs. This difference in gut colonisation presents important questions around the ability of vaccines to prevent shedding and transmission. These findings demonstrate that while protection conferred by two vaccines can appear to be the same, the mechanisms controlling the protection can differ and have important implications for infection dynamics within a population.
Collapse
Affiliation(s)
- Clare L. Kinnear
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Biosciences, The University of Melbourne, Melbourne, Victoria, Australia
- * E-mail:
| | - Richard A. Strugnell
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
49
|
Garg R, Akhade AS, Yadav J, Qadri A. MyD88-dependent pro-inflammatory activity in Vi polysaccharide vaccine against typhoid promotes Ab switching to IgG. Innate Immun 2015; 21:778-83. [PMID: 26303218 PMCID: PMC4572389 DOI: 10.1177/1753425915599242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/14/2015] [Indexed: 01/02/2023] Open
Abstract
Vi capsular polysaccharide is currently in use as a vaccine against human typhoid caused by Salmonella Typhi. The vaccine efficacy correlates with IgG anti-Vi Abs. We have recently reported that Vi can generate inflammatory responses through activation of the TLR2/TLR1 complex. In the present study, we show that immunization with Vi produces IgM as well as IgG Abs in wild type mice. This ability is not compromised in mice deficient in T cells. However, immunization of mice lacking the TLR adaptor protein, MyD88, with Vi elicits only IgM Abs. These results suggest that MyD88-dependent pro-inflammatory ability of the Vi vaccine might be vital in generating IgG Abs with this T-independent Ag.
Collapse
Affiliation(s)
- Rohini Garg
- Hybridoma Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Ajay Suresh Akhade
- Hybridoma Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Jitender Yadav
- Hybridoma Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Ayub Qadri
- Hybridoma Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| |
Collapse
|
50
|
Xiong K, Chen Z, Zhu C, Li J, Hu X, Rao X, Cong Y. Safety and immunogenicity of an attenuated Salmonella enterica serovar Paratyphi A vaccine candidate. Int J Med Microbiol 2015; 305:563-71. [PMID: 26239100 DOI: 10.1016/j.ijmm.2015.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 07/02/2015] [Accepted: 07/22/2015] [Indexed: 02/08/2023] Open
Abstract
Enteric fever caused by Salmonella enterica serovar Paratyphi A has progressively increased in recent years and became a global health issue. Currently licensed typhoid vaccines do not confer adequate cross-immunoprotection against S. Paratyphi A infection. Therefore, vaccines specifically against enteric fever caused by S. Paratyphi A are urgently needed. In the present study, an attenuated vaccine strain was constructed from S. Paratyphi A CMCC50093 by the deletions of aroC and yncD. The obtained strain SPADD01 showed reduced survival within THP-1 cells and less bacterial burden in spleens and livers of infected mice compared with the wild-type strain. The 50% lethal doses of SPADD01 and the wild-type strain were assessed using a murine infection model. The virulence of SPADD01 is approximately 40,000-fold less than that of the wild-type strain. In addition, SPADD01 showed an excellent immunogenicity in mouse model. Single intranasal inoculation elicited striking humoral and mucosal immune responses in mice and yielded effective protection against lethal challenge of the wild-type strain. A high level of cross-reactive humoral immune response against LPS of Salmonella enterica serovar Typhi was also detected in immunized mice. However, SPADD01 vaccination only conferred a low level of cross-protection against S. Typhi. Our data suggest that SPADD01 is a promising vaccine candidate against S. Paratyphi A infection and deserves further evaluation in clinical trial. To date, no study has demonstrated a good cross-protection between serovars of S. Typhi and S. Paratyphi A, suggesting that the dominant protective antigens of both serovars are likely different and need to be defined in future study.
Collapse
Affiliation(s)
- Kun Xiong
- Department of Microbiology, Third Military Medical University, Chongqing, 400038, PR China.
| | - Zhijin Chen
- Department of Microbiology, Third Military Medical University, Chongqing, 400038, PR China.
| | - Chunyue Zhu
- Department of Microbiology, Third Military Medical University, Chongqing, 400038, PR China.
| | - Jianhua Li
- Department of Microbiology, Third Military Medical University, Chongqing, 400038, PR China.
| | - Xiaomei Hu
- Department of Microbiology, Third Military Medical University, Chongqing, 400038, PR China.
| | - Xiancai Rao
- Department of Microbiology, Third Military Medical University, Chongqing, 400038, PR China.
| | - Yanguang Cong
- Department of Microbiology, Third Military Medical University, Chongqing, 400038, PR China.
| |
Collapse
|