1
|
Rappuoli R, Alter G, Pulendran B. Transforming vaccinology. Cell 2024; 187:5171-5194. [PMID: 39303685 DOI: 10.1016/j.cell.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 09/22/2024]
Abstract
The COVID-19 pandemic placed the field of vaccinology squarely at the center of global consciousness, emphasizing the vital role of vaccines as transformative public health tools. The impact of vaccines was recently acknowledged by the award of the 2023 Nobel Prize in Physiology or Medicine to Katalin Kariko and Drew Weissman for their seminal contributions to the development of mRNA vaccines. Here, we provide a historic perspective on the key innovations that led to the development of some 27 licensed vaccines over the past two centuries and recent advances that promise to transform vaccines in the future. Technological revolutions such as reverse vaccinology, synthetic biology, and structure-based design transformed decades of vaccine failures into successful vaccines against meningococcus B and respiratory syncytial virus (RSV). Likewise, the speed and flexibility of mRNA vaccines profoundly altered vaccine development, and the advancement of novel adjuvants promises to revolutionize our ability to tune immunity. Here, we highlight exciting new advances in the field of systems immunology that are transforming our mechanistic understanding of the human immune response to vaccines and how to predict and manipulate them. Additionally, we discuss major immunological challenges such as learning how to stimulate durable protective immune response in humans.
Collapse
Affiliation(s)
| | - Galit Alter
- Moderna Therapeutics, Cambridge, MA 02139, USA.
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
2
|
Miles SL, Holt KE, Mostowy S. Recent advances in modelling Shigella infection. Trends Microbiol 2024; 32:917-924. [PMID: 38423917 DOI: 10.1016/j.tim.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 03/02/2024]
Abstract
Shigella is an important human-adapted pathogen which contributes to a large global burden of diarrhoeal disease. Together with the increasing threat of antimicrobial resistance and lack of an effective vaccine, there is great urgency to identify novel therapeutics and preventatives to combat Shigella infection. In this review, we discuss the development of innovative technologies and animal models to study mechanisms underlying Shigella infection of humans. We examine recent literature introducing (i) the organ-on-chip model, and its substantial contribution towards understanding the biomechanics of Shigella infection, (ii) the zebrafish infection model, which has delivered transformative insights into the epidemiological success of clinical isolates and the innate immune response to Shigella, (iii) a pioneering oral mouse model of shigellosis, which has helped to discover new inflammasome biology and protective mechanisms against shigellosis, and (iv) the controlled human infection model, which has been effective in translating basic research into human health impact and assessing suitability of novel vaccine candidates. We consider the recent contributions of each model and discuss where the future of modelling Shigella infection lies.
Collapse
Affiliation(s)
- Sydney L Miles
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Kathryn E Holt
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK; Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| |
Collapse
|
3
|
Richardson D, Savary-Trathen A, Fitzpatrick C, Williams D. Estimated prevalence and associations of sexually transmissible bacterial enteric pathogens in asymptomatic men who have sex with men: a systematic review and meta-analysis. Sex Transm Infect 2024:sextrans-2024-056183. [PMID: 38902026 DOI: 10.1136/sextrans-2024-056183] [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: 03/13/2024] [Accepted: 06/09/2024] [Indexed: 06/22/2024] Open
Abstract
OBJECTIVE The reservoir of sexually transmissible bacterial enteric pathogens in asymptomatic men who have sex with men (MSM) may impact future outbreaks, and the evolution of antimicrobial resistance. We aimed to estimate the pooled prevalence and explore any factors associated with Shigella spp, Campylobacter spp, diarrhoeagenic Escherichia coli and Salmonella spp in asymptomatic MSM using the random effects model. METHODS We searched Embase, MEDLINE, CINAHL and Web of Science Core Collections for manuscripts published up to February 2024. One author screened citations and abstracts; two authors independently conducted a full-text review. We included manuscripts which measured the prevalence of Shigella spp, Campylobacter spp, diarrhoeagenic E. coli and Salmonella spp in asymptomatic MSM. Quality and risk of bias was assessed independently by two authors using the Joanna Briggs Institute critical appraisal tools. We calculated pooled prevalence and CIs using the random effects model. RESULTS Six manuscripts were included in the final review. The manuscripts were from Australia (n=2), the UK (n=2), the Netherlands (n=1) and the USA (n=1) and included data from 3766 asymptomatic MSM tested for bacterial enteric pathogens. The prevalence of Shigella spp was 1.1% (95% CI 0.7% to 1.7%), Campylobacter spp 1.9% (95% CI 1.5% to 2.5%), diarrhoeagenic E. coli 3.8% (95% CI 2.1% to 6.7%) and Salmonella spp 0.3% (95% CI 0.1% to 0.6%). Two manuscripts demonstrated that the detection of bacterial enteric pathogen was more frequent in asymptomatic MSM using HIV-pre-exposure prophylaxis (PrEP), living with HIV, reporting <5 new sexual partners in the past 3 months, reporting insertive oral-anal sex and group sex compared with MSM testing negative. CONCLUSION Despite a small number of manuscripts, this review has estimated the pooled prevalence, and highlighted some possible associations with sexually transmissible bacterial enteric pathogens in asymptomatic MSM, which can inform future clinical guidelines, public health control strategies and research to increase our understanding of transmission and the evolution of antimicrobial resistance. PROSPERO REGISTRATION NUMBER CRD42024518700.
Collapse
Affiliation(s)
- Daniel Richardson
- Sexual Health & HIV medicine, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
- Global Health & Infection, Brighton and Sussex Medical School, Brighton, UK
| | - Amber Savary-Trathen
- Sexual Health & HIV medicine, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| | - Colin Fitzpatrick
- Sexual Health & HIV medicine, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| | - Deborah Williams
- Sexual Health & HIV medicine, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| |
Collapse
|
4
|
Tian X, Janes HE, Kublin JG. Statistical design and analysis of controlled human malaria infection trials. Malar J 2024; 23:133. [PMID: 38702775 PMCID: PMC11068571 DOI: 10.1186/s12936-024-04959-2] [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: 05/01/2023] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Malaria is a potentially life-threatening disease caused by Plasmodium protozoa transmitted by infected Anopheles mosquitoes. Controlled human malaria infection (CHMI) trials are used to assess the efficacy of interventions for malaria elimination. The operating characteristics of statistical methods for assessing the ability of interventions to protect individuals from malaria is uncertain in small CHMI studies. This paper presents simulation studies comparing the performance of a variety of statistical methods for assessing efficacy of intervention in CHMI trials. METHODS Two types of CHMI designs were investigated: the commonly used single high-dose design (SHD) and the repeated low-dose design (RLD), motivated by simian immunodeficiency virus (SIV) challenge studies. In the context of SHD, the primary efficacy endpoint is typically time to infection. Using a continuous time survival model, five statistical tests for assessing the extent to which an intervention confers partial or full protection under single dose CHMI designs were evaluated. For RLD, the primary efficacy endpoint is typically the binary infection status after a specific number of challenges. A discrete time survival model was used to study the characteristics of RLD versus SHD challenge studies. RESULTS In a SHD study with the continuous time survival model, log-rank test and t-test are the most powerful and provide more interpretable results than Wilcoxon rank-sum tests and Lachenbruch tests, while the likelihood ratio test is uniformly most powerful but requires knowledge of the underlying probability model. In the discrete time survival model setting, SHDs are more powerful for assessing the efficacy of an intervention to prevent infection than RLDs. However, additional information can be inferred from RLD challenge designs, particularly using a likelihood ratio test. CONCLUSIONS Different statistical methods can be used to analyze controlled human malaria infection (CHMI) experiments, and the choice of method depends on the specific characteristics of the experiment, such as the sample size allocation between the control and intervention groups, and the nature of the intervention. The simulation results provide guidance for the trade off in statistical power when choosing between different statistical methods and study designs.
Collapse
Affiliation(s)
- Xiaowen Tian
- Department of Biostatistics, University of Washington, 3980 15th Ave NE, Seattle, WA, 98195, USA.
| | - Holly E Janes
- Department of Biostatistics, University of Washington, 3980 15th Ave NE, Seattle, WA, 98195, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
- Department of Global Health, University of Washington, 3980 15th Ave NE, Seattle, WA, 98195, USA
| |
Collapse
|
5
|
Bose P, Chowdhury G, Halder G, Ghosh D, Deb AK, Kitahara K, Miyoshi SI, Morita M, Ramamurthy T, Dutta S, Mukhopadhyay AK. Prevalence and changing antimicrobial resistance profiles of Shigella spp. isolated from diarrheal patients in Kolkata during 2011-2019. PLoS Negl Trop Dis 2024; 18:e0011964. [PMID: 38377151 PMCID: PMC10906866 DOI: 10.1371/journal.pntd.0011964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 03/01/2024] [Accepted: 02/02/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND The primary aim of this study was to investigate the occurrence, characteristics, and antimicrobial resistance patterns of various Shigella serogroups isolated from patients with acute diarrhea of the Infectious Diseases Hospital in Kolkata from 2011-2019. PRINCIPAL FINDINGS During the study period, Shigella isolates were tested for their serogroups, antibiotic resistance pattern and virulence gene profiles. A total of 5.8% of Shigella spp. were isolated, among which S. flexneri (76.1%) was the highest, followed by S. sonnei (18.7%), S. boydii (3.4%), and S. dysenteriae (1.8%). Antimicrobial resistance against nalidixic acid was higher in almost all the Shigella isolates, while the resistance to β-lactamases, fluoroquinolones, tetracycline, and chloramphenicol diverged. The occurrence of multidrug resistance was found to be linked with various genes encoding drug-resistance, multiple mutations in the topoisomerase genes, and mobile genetic elements. All the isolates were positive for the invasion plasmid antigen H gene (ipaH). Dendrogram analysis of the plasmid and pulsed-field electrophoresis (PFGE) profiles revealed 70-80% clonal similarity among each Shigella serotype. CONCLUSION This comprehensive long-term surveillance report highlights the clonal diversity of clinical Shigella strains circulating in Kolkata, India, and shows alarming resistance trends towards recommended antibiotics. The elucidation of this study's outcome is helpful not only in identifying emerging antimicrobial resistance patterns of Shigella spp. but also in developing treatment guidelines appropriate for this region.
Collapse
Affiliation(s)
- Puja Bose
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Goutam Chowdhury
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
- Collaborative Research Centre of Okayama University for Infectious Diseases at ICMR-NICED, Kolkata, India
| | - Gourab Halder
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Debjani Ghosh
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Alok K. Deb
- Division of Epidemiology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Kei Kitahara
- Collaborative Research Centre of Okayama University for Infectious Diseases at ICMR-NICED, Kolkata, India
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shin-ichi Miyoshi
- Collaborative Research Centre of Okayama University for Infectious Diseases at ICMR-NICED, Kolkata, India
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masatomo Morita
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Thandavarayan Ramamurthy
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Shanta Dutta
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Asish Kumar Mukhopadhyay
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| |
Collapse
|
6
|
León Y, Honigsberg R, Rasko DA, Faherty CS. Gastrointestinal signals in supplemented media reveal a role in adherence for the Shigella flexneri sap autotransporter gene. Gut Microbes 2024; 16:2331985. [PMID: 38549437 PMCID: PMC10984119 DOI: 10.1080/19490976.2024.2331985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 03/14/2024] [Indexed: 04/02/2024] Open
Abstract
Shigella flexneri causes severe diarrheal disease worldwide. While many aspects of pathogenesis have been elucidated, significant knowledge gaps remain regarding the role of putative chromosomally-encoded virulence genes. The uncharacterized sap gene encoded on the chromosome has significant nucleotide sequence identity to the fluffy (flu) antigen 43 autotransporter gene in pathogenic Escherichia coli. Here, we constructed a Δsap mutant in S. flexneri strain 2457T and examined the effects of this mutation on bacterial cell aggregation, biofilm formation, and adherence to colonic epithelial cells. Analyses included the use of growth media supplemented with glucose and bile salts to replicate small intestinal signals encountered by S. flexneri. Deletion of the sap gene in 2457T affected epithelial cell adherence, resulted in quicker bacterial cell aggregation, but did not affect biofilm formation. This work highlights a functional role for the sap gene in S. flexneri pathogenesis and further demonstrates the importance of using relevant and appropriate gastrointestinal signals to characterize virulence genes of enteropathogenic bacteria.
Collapse
Affiliation(s)
- Yrvin León
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Raphael Honigsberg
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, USA
- École Normale Supérieure Paris-Saclay, Département d’Enseignement et de, Recherche de Biologie, Université Paris-Saclay, Gif-sur-Yvette, France
| | - David A. Rasko
- Institute for Genome Sciences, Center for Pathogen Research, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Christina S. Faherty
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Clarkson KA, Porter CK, Talaat KR, Kapulu MC, Chen WH, Frenck RW, Bourgeois AL, Kaminski RW, Martin LB. Shigella-Controlled Human Infection Models: Current and Future Perspectives. Curr Top Microbiol Immunol 2024; 445:257-313. [PMID: 35616717 PMCID: PMC7616482 DOI: 10.1007/82_2021_248] [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] [Indexed: 10/18/2022]
Abstract
Shigella-controlled human infection models (CHIMs) are an invaluable tool utilized by the vaccine community to combat one of the leading global causes of infectious diarrhea, which affects infants, children and adults regardless of socioeconomic status. The impact of shigellosis disproportionately affects children in low- and middle-income countries (LMICs) resulting in cognitive and physical stunting, perpetuating a cycle that must be halted. Shigella-CHIMs not only facilitate the early evaluation of enteric countermeasures and up-selection of the most promising products but also provide insight into mechanisms of infection and immunity that are not possible utilizing animal models or in vitro systems. The greater understanding of shigellosis obtained in CHIMs builds and empowers the development of new generation solutions to global health issues which are unattainable in the conventional laboratory and clinical settings. Therefore, refining, mining and expansion of safe and reproducible infection models hold the potential to create effective means to end diarrheal disease and associated co-morbidities associated with Shigella infection.
Collapse
Affiliation(s)
- Kristen A Clarkson
- Department of Diarrheal Disease Research, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Chad K Porter
- Enteric Disease Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Kawsar R Talaat
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, 624 North Broadway Street Hampton House, Baltimore, MD, 21205, USA
| | - Melissa C Kapulu
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi County Hospital, Off Bofa Road, Kilifi, 80108, Kenya
| | - Wilbur H Chen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Robert W Frenck
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - A Louis Bourgeois
- PATH Center for Vaccine Innovation and Access, 455 Massachusetts Avenue NW, Washington, DC, 20001, USA
| | - Robert W Kaminski
- Department of Diarrheal Disease Research, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Laura B Martin
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100, Siena, Italy.
| |
Collapse
|
8
|
De Silva PM, Bennett RJ, Kuhn L, Ngondo P, Debande L, Njamkepo E, Ho B, Weill FX, Marteyn BS, Jenkins C, Baker KS. Escherichia coli killing by epidemiologically successful sublineages of Shigella sonnei is mediated by colicins. EBioMedicine 2023; 97:104822. [PMID: 37806286 PMCID: PMC10579285 DOI: 10.1016/j.ebiom.2023.104822] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023] Open
Abstract
BACKGROUND Shigella sp. are enteric pathogens which causes >125 million cases of shigellosis annually. S. sonnei accounts for about a quarter of those cases and is increasingly prevalent in industrialising nations. Being an enteric pathogen, S. sonnei benefits from outcompeting gut commensals such as Escherichia coli to establish itself and cause disease. There are numerous mechanisms that bacterial pathogens use to outcompete its rivals including molecules called colicins. A Type 6 Secretion System (T6SS) was recently described as contributing to E. coli killing in S. sonnei. METHODS We used Bulk Phenotyping of Epidemiological Replicates (BPER) which combined bacterial Genome Wide Association Studies (bGWAS) and high throughput phenotyping on a collection of S. sonnei surveillance isolates to identify the genetic features associated with E. coli killing and explore their relationship with epidemiological behaviour. We further explored the presence of colicins and T6SS components in the isolates using genomics, laboratory experimentation, and proteomics. FINDINGS Our bGWAS analysis returned known and novel colicin and colicin related genes as significantly associated with E. coli killing. In silico analyses identified key colicin clusters responsible for the killing phenotype associated with epidemiologically successful sub-lineages. The killing phenotype was not associated with the presence of a T6SS. Laboratory analyses confirmed the presence of the key colicin clusters and that killing was contact-independent. INTERPRETATION Colicins are responsible for E. coli killing by S. sonnei, not a T6SS. This phenotype contributes to shaping the observed epidemiology of S. sonnei and may contribute to its increasing prevalence globally. BPER is an epidemiologically relevant approach to phenotypic testing that enables the rapid identification of genetic drivers of phenotypic changes, and assessment of their relevance to epidemiology in natural settings. FUNDING Biotechnology and Biological Sciences Research Council, Biotechnology and Biological Sciences Research Council Doctoral Training Partnership studentship, Wellcome Trust, Medical Research Council (UK), French National Research Agency.
Collapse
Affiliation(s)
- P Malaka De Silva
- Department of Clinical Infection, Microbiology, and Immunology, Institute for Infection, Veterinary, and Ecological Sciences (IVES), University of Liverpool, Liverpool, United Kingdom
| | - Rebecca J Bennett
- Department of Clinical Infection, Microbiology, and Immunology, Institute for Infection, Veterinary, and Ecological Sciences (IVES), University of Liverpool, Liverpool, United Kingdom
| | - Lauriane Kuhn
- Plateforme protéomique Strasbourg Esplanade FR1589 du CNRS, Université de Strasbourg, Strasbourg, France
| | - Patryk Ngondo
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR9002, F-67000, Strasbourg, France
| | - Lorine Debande
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR9002, F-67000, Strasbourg, France
| | - Elisabeth Njamkepo
- Institut Pasteur, Université Paris Cité, Unité des Bactéries pathogènes entériques, Centre National de Référence des Escherichia coli, Shigella et Salmonella, Paris, F-75015, France
| | - Brian Ho
- Institute of Structural and Molecular Biology, University College London and Birkbeck, London, UK
| | - François-Xavier Weill
- Institut Pasteur, Université Paris Cité, Unité des Bactéries pathogènes entériques, Centre National de Référence des Escherichia coli, Shigella et Salmonella, Paris, F-75015, France
| | - Benoît S Marteyn
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR9002, F-67000, Strasbourg, France
| | - Claire Jenkins
- Gastro and Food Safety (One Health) Division, UK Health Security Agency, Colindale, London, UK
| | - Kate S Baker
- Department of Clinical Infection, Microbiology, and Immunology, Institute for Infection, Veterinary, and Ecological Sciences (IVES), University of Liverpool, Liverpool, United Kingdom; Department of Genetics, University of Cambridge, Downing Place, Cambridge, UK.
| |
Collapse
|
9
|
Toapanta FR, Hu J, Meron-Sudai S, Mulard LA, Phalipon A, Cohen D, Sztein MB. Further characterization of Shigella-specific (memory) B cells induced in healthy volunteer recipients of SF2a-TT15, a Shigella flexneri 2a synthetic glycan-based vaccine candidate. Front Immunol 2023; 14:1291664. [PMID: 38022674 PMCID: PMC10653583 DOI: 10.3389/fimmu.2023.1291664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Shigellosis is common worldwide, and it causes significant morbidity and mortality mainly in young children in low- and middle- income countries. To date, there are not broadly available licensed Shigella vaccines. A novel type of conjugate vaccine candidate, SF2a-TT15, was developed against S. flexneri serotype 2a (SF2a). SF2a-TT15 is composed of a synthetic 15mer oligosaccharide, designed to act as a functional mimic of the SF2a O-antigen and covalently linked to tetanus toxoid (TT). SF2a-TT15 was recently shown to be safe and immunogenic in a Phase 1 clinical trial, inducing specific memory B cells and sustained antibody response up to three years after the last injection. In this manuscript, we advance the study of B cell responses to parenteral administration of SF2a-TT15 to identify SF2a LPS-specific B cells (SF2a+ B cells) using fluorescently labeled bacteria. SF2a+ B cells were identified mainly within class-switched B cells (SwB cells) in volunteers vaccinated with SF2a-TT15 adjuvanted or not with aluminium hydroxide (alum), but not in placebo recipients. These cells expressed high levels of CXCR3 and low levels of CD21 suggesting an activated phenotype likely to represent the recently described effector memory B cells. IgG SF2a+ SwB cells were more abundant than IgA SF2a + SwB cells. SF2a+ B cells were also identified in polyclonally stimulated B cells (antibody secreting cells (ASC)-transformed). SF2a+ ASC-SwB cells largely maintained the activated phenotype (CXCR3 high, CD21 low). They expressed high levels of CD71 and integrin α4β7, suggesting a high proliferation rate and ability to migrate to gut associated lymphoid tissues. Finally, ELISpot analysis showed that ASC produced anti-SF2a LPS IgG and IgA antibodies. In summary, this methodology confirms the ability of SF2a-TT15 to induce long-lived memory B cells, initially identified by ELISpots, which remain identifiable in blood up to 140 days following vaccination. Our findings expand and complement the memory B cell data previously reported in the Phase 1 trial and provide detailed information on the immunophenotypic characteristics of these cells. Moreover, this methodology opens the door to future studies at the single-cell level to better characterize the development of B cell immunity to Shigella.
Collapse
Affiliation(s)
- Franklin R. Toapanta
- Department of Medicine and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jingping Hu
- Department of Medicine and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Shiri Meron-Sudai
- School of Public Health, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Laurence A. Mulard
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Unité Chimie des Biomolécules, Paris, France
| | - Armelle Phalipon
- Institut Pasteur, Université Paris Cité, Laboratoire Innovation: Vaccins, Paris, France
| | - Dani Cohen
- School of Public Health, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Marcelo B. Sztein
- Department of Medicine and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
10
|
Baharvand A, Molaeipour L, Alesaeidi S, Shaddel R, Mashatan N, Amiriani T, Kiaei Sudkolaei M, Abbasian S, Talib Al-Naqeeb BZ, Kouhsari E. The increasing antimicrobial resistance of Shigella species among Iranian pediatrics: a systematic review and meta-analysis. Pathog Glob Health 2023; 117:611-622. [PMID: 36794800 PMCID: PMC10498791 DOI: 10.1080/20477724.2023.2179451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Shigellosis remains one of the global causes of morbidity and mortality. However, the global emergence of antibiotic resistance has become the leading cause of treatment failure in shigellosis. This review aimed to provide an updated picture of the antimicrobial resistance rates in Shigella species in Iranian pediatrics. METHODS A comprehensive systematic search was performed on PubMed, Scopus, Embase, and Web of Science until 28 July 2021. The meta-analysis was performed by computing the pooled using a random-effects model with Stata/SE software, v.17.1. The discrepancy within articles was surveyed by the forest plot in addition to the I2 statistic. All statistical interpretations were reported on a 95% confidence interval (CI) basis. RESULTS Totally, of 28 eligible studies published between 2008 and 2021. The pooled prevalence rate of multidrug-resistant (MDR) was 63% (95% CI 50-76). Regarding suggested antimicrobial agents for Shigella species, the prevalence of resistance for ciprofloxacin, azithromycin, and ceftriaxone as first- and second-line treatments for shigellosis were 3%, 30%, and 28%, respectively. In contrast, resistance to cefotaxime, cefixime, and ceftazidime was 39%, 35%, and 20%. Importantly, subgroup analyses indicated that an increase in resistance rates during the periods (2008-2014, 2015-2021) was recognized for ciprofloxacin (0 % to 6%) and ceftriaxone (6% to 42%). CONCLUSION Our findings revealed that ciprofloxacin is an effective drug for shigellosis in Iranian children. The substantially high prevalence estimation proposes that the first- and second-line treatments for shigellosis are the major threat to public health and active antibiotic treatment policies are essential.
Collapse
Affiliation(s)
| | - Leila Molaeipour
- Department of Epidemiology, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Sogol Alesaeidi
- Department of Pediatric Medicine, Imam Hossein Hospital, Resident of pediatric medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reyhane Shaddel
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Noushin Mashatan
- Graduated, School of Applied Sciences, University of Brighton, Brighton, UK
| | - Taghi Amiriani
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Melika Kiaei Sudkolaei
- Department of Nutrition, Faculty of Health, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sara Abbasian
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Ebrahim Kouhsari
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Laboratory Sciences, Faculty of Paramedicine, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
11
|
Quantitative dose-response analysis untangles host bottlenecks to enteric infection. Nat Commun 2023; 14:456. [PMID: 36709326 PMCID: PMC9884216 DOI: 10.1038/s41467-023-36162-3] [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] [Received: 10/18/2022] [Accepted: 01/17/2023] [Indexed: 01/30/2023] Open
Abstract
Host bottlenecks prevent many infections before the onset of disease by eliminating invading pathogens. By monitoring the diversity of a barcoded population of the diarrhea causing bacterium Citrobacter rodentium during colonization of its natural host, mice, we determine the number of cells that found the infection by establishing a replicative niche. In female mice the size of the pathogen's founding population scales with dose and is controlled by a severe yet slow-acting bottleneck. Reducing stomach acid or changing host genotype modestly relaxes the bottleneck without breaking the fractional relationship between dose and founders. In contrast, disrupting the microbiota causes the founding population to no longer scale with the size of the inoculum and allows the pathogen to infect at almost any dose, indicating that the microbiota creates the dominant bottleneck. Further, in the absence of competition with the microbiota, the diversity of the pathogen population slowly contracts as the population is overtaken by bacteria having lost the critical virulence island, the locus of enterocyte effacement (LEE). Collectively, our findings reveal that the mechanisms of protection by colonization bottlenecks are reflected in and can be generally defined by the impact of dose on the pathogen's founding population.
Collapse
|
12
|
Choy RKM, Bourgeois AL, Ockenhouse CF, Walker RI, Sheets RL, Flores J. Controlled Human Infection Models To Accelerate Vaccine Development. Clin Microbiol Rev 2022; 35:e0000821. [PMID: 35862754 PMCID: PMC9491212 DOI: 10.1128/cmr.00008-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The timelines for developing vaccines against infectious diseases are lengthy, and often vaccines that reach the stage of large phase 3 field trials fail to provide the desired level of protective efficacy. The application of controlled human challenge models of infection and disease at the appropriate stages of development could accelerate development of candidate vaccines and, in fact, has done so successfully in some limited cases. Human challenge models could potentially be used to gather critical information on pathogenesis, inform strain selection for vaccines, explore cross-protective immunity, identify immune correlates of protection and mechanisms of protection induced by infection or evoked by candidate vaccines, guide decisions on appropriate trial endpoints, and evaluate vaccine efficacy. We prepared this report to motivate fellow scientists to exploit the potential capacity of controlled human challenge experiments to advance vaccine development. In this review, we considered available challenge models for 17 infectious diseases in the context of the public health importance of each disease, the diversity and pathogenesis of the causative organisms, the vaccine candidates under development, and each model's capacity to evaluate them and identify correlates of protective immunity. Our broad assessment indicated that human challenge models have not yet reached their full potential to support the development of vaccines against infectious diseases. On the basis of our review, however, we believe that describing an ideal challenge model is possible, as is further developing existing and future challenge models.
Collapse
Affiliation(s)
- Robert K. M. Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | - A. Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Richard I. Walker
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Jorge Flores
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| |
Collapse
|
13
|
Porter CK, Detizio KJ, Maier N, Testa KJ, Talaat KR, Chen WH, Lyon CE, Gutierrez RL, Frenck R, Isidean SD, Kaminski RW, Alcala AN, Hanevik K, Sawe F, Kirkpatrick BD, Louis Bourgeois A. A site assessment tool for inpatient controlled human infection models for enteric disease pathogens. Clin Trials 2021; 19:116-118. [PMID: 34708664 DOI: 10.1177/17407745211052473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The use of the controlled human infection model to facilitate product development and to advance understanding of host-pathogen interactions is of increasing interest. While administering a virulent (or infective) organism to a susceptible host necessitates an ongoing evaluation of safety and ethical considerations, a central theme in conducting these studies in a safe and ethical manner that yields actionable data is their conduct in facilities well-suited to address their unique attributes. To that end, we have developed a framework for evaluating potential sites in which to conduct inpatient enteric controlled human infection model to ensure consistency and increase the likelihood of success.
Collapse
Affiliation(s)
- Chad K Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, USA
| | - Katherine J Detizio
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | | | - Kayla J Testa
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Kawsar R Talaat
- Center for Immunization Research, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Wilbur H Chen
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Caroline E Lyon
- Vaccine Testing Center, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| | - Ramiro L Gutierrez
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, USA
| | - Robert Frenck
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sandra D Isidean
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Robert W Kaminski
- Bacterial Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Ashley N Alcala
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Kurt Hanevik
- Norwegian National Advisory Unit on Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Frederick Sawe
- Kenya Medical Research Institute and United States Army Medical Research Directorate-Africa, Kericho, Kenya.,Henry M. Jackson Foundation Medical Research International, Bethesda, MD, USA
| | - Beth D Kirkpatrick
- Vaccine Testing Center, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| | | |
Collapse
|
14
|
Talaat KR, Alaimo C, Martin P, Bourgeois AL, Dreyer AM, Kaminski RW, Porter CK, Chakraborty S, Clarkson KA, Brubaker J, Elwood D, Frölich R, DeNearing B, Weerts H, Feijoo BL, Halpern J, Sack D, Riddle MS, Fonck VG. Human challenge study with a Shigella bioconjugate vaccine: Analyses of clinical efficacy and correlate of protection. EBioMedicine 2021; 66:103310. [PMID: 33862589 PMCID: PMC8054157 DOI: 10.1016/j.ebiom.2021.103310] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/15/2021] [Accepted: 03/12/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Shigellosis is a major cause of moderate to severe diarrhoea and dysentery in children under 5 years of age in low and middle-income countries. The Flexyn2a vaccine conjugates the O-polysaccharide of Shigella flexneri 2a to Pseudomonas aeruginosa exotoxin A. We describe a Phase 2b proof-of-concept challenge study that evaluated safety, immunogenicity, and efficacy of the Flexyn2a vaccine to protect against shigellosis. METHODS In this randomized, double blind, placebo-controlled trial, healthy adults were randomized 1:1 to receive Flexyn2a (10 µg) or placebo intramuscularly, twice, 4 weeks apart, followed by challenge 4 weeks later with 1500 colony forming units (CFUs) of S. flexneri 2a strain 2457T. The primary outcome was vaccine-induced protection. S. flexneri 2a lipopolysaccharide (LPS)-specific immune responses were assessed. FINDINGS Sixty-seven subjects were enrolled, 34 received vaccine and 33 placebo. The vaccine was well tolerated; the majority of adverse events were mild in nature. Thirty vaccinees and 29 placebo recipients received the S. flexneri 2a challenge. Vaccination resulted in a 30.2% reduction in shigellosis compared with placebo (13/30 vs. 18/29; p = 0.11; 95% CI -15 to 62.6). Vaccine efficacy was more robust against severe disease, reaching 51.7% (p = 0.015, 95% CI 5.3 to 77.9) against moderate/severe diarrhoea or dysentery concurrent with fever or severe enteric symptoms and 72.4% (p = 0.07) against more severe diarrhoea (≥10 lose stools or ≥1000 g loose stools/24 h). Vaccinated subjects were less likely to need early antibiotic intervention following challenge (protective efficacy 51.7%, p = 0.01; 95% CI 9 to 76.8). In those who developed shigellosis, vaccinated subjects had a lower disease severity score (p = 0.002) than placebo-recipients. Additionally, LPS-specific serum IgG responses in Flexyn2a recipients were associated with protection against disease (p = 0.0016) and with a decreased shigellosis disease score (p = 0.002). INTERPRETATION The Flexyn2a bioconjugate vaccine was immunogenic, well tolerated and protected against severe illness after Shigella challenge and is a promising Shigella vaccine construct. We identified a strong association between anti-S. flexneri 2a serum IgG and a reduction in disease outcomes. (Clinicaltrials.gov, NCT02646371.) FUNDING: Funding for this study was through a grant from the Wellcome Trust.
Collapse
Affiliation(s)
- Kawsar R Talaat
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States.
| | | | | | - A Louis Bourgeois
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States; Now at PATH Center for Vaccine Innovation and Access, Washington, DC, United States
| | | | - Robert W Kaminski
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Chad K Porter
- Naval Medical Research Center, Silver Spring, MD, United States
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kristen A Clarkson
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Jessica Brubaker
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Daniel Elwood
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | | | - Barbara DeNearing
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Hailey Weerts
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Brittany L Feijoo
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jane Halpern
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - David Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Mark S Riddle
- Naval Medical Research Center, Silver Spring, MD, United States
| | | |
Collapse
|
15
|
Clarkson KA, Talaat KR, Alaimo C, Martin P, Bourgeois AL, Dreyer A, Porter CK, Chakraborty S, Brubaker J, Elwood D, Frölich R, DeNearing B, Weerts HP, Feijoo B, Halpern J, Sack D, Riddle MS, Fonck VG, Kaminski RW. Immune response characterization in a human challenge study with a Shigella flexneri 2a bioconjugate vaccine. EBioMedicine 2021; 66:103308. [PMID: 33813141 PMCID: PMC8047506 DOI: 10.1016/j.ebiom.2021.103308] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/13/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Diarrheal diseases are a leading cause of global morbidity and mortality affecting all ages, but especially children under the age of five in resource-limited settings. Shigella is a leading contributor to diarrheal diseases caused by bacterial pathogens and is considered a significant antimicrobial resistance threat. While improvements in hygiene, and access to clean water help as control measures, vaccination remains one of the most viable options for significantly reducing morbidity and mortality. METHODS Flexyn2a is a bioconjugate vaccine manufactured using novel conjugation methodologies enzymatically linking the O-polysaccharide of S. flexneri 2a to exotoxin A of Pseudomonas aeruginosa. The protective capacity of Flexyn2a was assessed in a controlled human infection model after two intramuscular immunizations. Immune responses pre- and post-immunization and/or infection were investigated and are described here. FINDINGS Flexyn2a induced lipopolysaccharide (LPS)-specific serum IgG responses post-immunization which were associated with protection against shigellosis. Additionally, several other immune parameters, including memory B cell responses, bactericidal antibodies and serum IgA, were also elevated in vaccinees protected against shigellosis. Immunization with Flexyn2a also induced gut-homing, LPS-specific IgG and IgA secreting B cells, indicating the vaccine induced immune effectors functioning at the site of intestinal infection. INTERPRETATION Collectively, the results of these immunological investigations provide insights into protective immune mechanisms post-immunization with Flexyn2a which can be used to further guide vaccine development and may have applicability to the larger Shigella vaccine field. FUNDING Funding for this study was provided through a Wellcome Trust grant.
Collapse
Affiliation(s)
- Kristen A Clarkson
- Department of Enteric Infections, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Kawsar R Talaat
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | | | | | | | | | - Chad K Porter
- Naval Medical Research Center, Silver Spring, MD, United States
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jessica Brubaker
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Daniel Elwood
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | | | - Barbara DeNearing
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Hailey P Weerts
- Department of Enteric Infections, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Brittany Feijoo
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jane Halpern
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - David Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Mark S Riddle
- Naval Medical Research Center, Silver Spring, MD, United States
| | | | - Robert W Kaminski
- Department of Enteric Infections, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States.
| |
Collapse
|
16
|
Abstract
Shigella is a major cause of moderate to severe diarrhea largely affecting children (<5 years old) living in low- and middle-income countries. Several vaccine candidates are in development, and controlled human infection models (CHIMs) can be useful tools to provide an early assessment of vaccine efficacy and potentially support licensure. A lyophilized strain of S. sonnei 53G was manufactured and evaluated to establish a dose that safely and reproducibly induced a ≥60% attack rate. Samples were collected pre- and postchallenge to assess intestinal inflammatory responses, antigen-specific serum and mucosal antibody responses, functional antibody responses, and memory B cell responses. Infection with S. sonnei 53G induced a robust intestinal inflammatory response as well as antigen-specific antibodies in serum and mucosal secretions and antigen-specific IgA- and IgG-secreting B cells positive for the α4β7 gut-homing marker. There was no association between clinical disease outcomes and systemic or functional antibody responses postchallenge; however, higher lipopolysaccharide (LPS)-specific serum IgA- and IgA-secreting memory B cell responses were associated with a reduced risk of disease postchallenge. This study provides unique insights into the immune responses pre- and postinfection with S. sonnei 53G in a CHIM, which could help guide the rational design of future vaccines to induce protective immune responses more analogous to those triggered by infection.IMPORTANCE Correlate(s) of immunity have yet to be defined for shigellosis. As previous disease protects against subsequent infection in a serotype-specific manner, investigating immune response profiles pre- and postinfection provides an opportunity to identify immune markers potentially associated with the development of protective immunity and/or with a reduced risk of developing shigellosis postchallenge. This study is the first to report such an extensive characterization of the immune response after challenge with S. sonnei 53G. Results demonstrate an association of progression to shigellosis with robust intestinal inflammatory and mucosal gut-homing responses. An important finding in this study was the association of elevated Shigella LPS-specific serum IgA and memory B cell IgA responses at baseline with reduced risk of disease. The increased baseline IgA responses may contribute to the lack of dose response observed in the study and suggests that IgA responses should be further investigated as potential correlates of immunity.
Collapse
|
17
|
Abstract
Controlled human infection models (CHIMs) are invaluable tools utilized to understand the human response to infection, potentially leading to protective immune mechanisms and allowing efficacy testing of enteric countermeasures, including vaccines, antibiotics, and other products. The development of an improved Shigella CHIM for both Shigella sonnei and Shigella flexneri is consistent with international efforts, supported by international donors and the World Health Organization, focused on standardizing Shigella CHIMs and using them to accelerate Shigella vaccine development. The use of lyophilized Shigella challenge strains rather than plate-grown inoculum preparations is considered an important step forward in the standardization process. Furthermore, the results of studies such as this justify the development of lyophilized preparations for additional epidemiologically important S. flexneri serotypes, including S. flexneri 3a and S. flexneri 6. Controlled human infection models (CHIMs) are useful for vaccine development. To improve on existing models, we developed a CHIM using a lyophilized preparation of Shigella sonnei strain 53G produced using current good manufacturing practice (cGMP). Healthy adults were enrolled in an open-label dose-ranging study. Following administration of a dose of rehydrated S. sonnei strain 53G, subjects were monitored for development of disease. The first cohort received 500 CFU of 53G, and dosing of subsequent cohorts was based on results from the previous cohort. Subjects were administered ciprofloxacin on day 5 and discharged home on day 8. Subjects returned as outpatients for clinical checks and sample collection. Attack rates increased as the dose of S. sonnei was increased. Among those receiving the highest dose (1,760 CFU), 70% developed moderate to severe diarrhea, 50% had dysentery, and 40% had fever. Antilipopolysaccharide responses were observed across all cohorts. An S. sonnei CHIM using a lyophilized lot of strain 53G was established. A dose in the range of 1,500 to 2,000 CFU of 53G was selected as the dose for future challenge studies using this product. This model will enable direct comparison of study results between institutions and ensure better consistency over time in the challenge inoculum. IMPORTANCE Controlled human infection models (CHIMs) are invaluable tools utilized to understand the human response to infection, potentially leading to protective immune mechanisms and allowing efficacy testing of enteric countermeasures, including vaccines, antibiotics, and other products. The development of an improved Shigella CHIM for both Shigella sonnei and Shigella flexneri is consistent with international efforts, supported by international donors and the World Health Organization, focused on standardizing Shigella CHIMs and using them to accelerate Shigella vaccine development. The use of lyophilized Shigella challenge strains rather than plate-grown inoculum preparations is considered an important step forward in the standardization process. Furthermore, the results of studies such as this justify the development of lyophilized preparations for additional epidemiologically important S. flexneri serotypes, including S. flexneri 3a and S. flexneri 6.
Collapse
|
18
|
Bekeredjian-Ding I, Van Molle W, Baay M, Neels P. Human challenge trial workshop: Focus on quality requirements for challenge agents, Langen, Germany, October 22, 2019. Biologicals 2020; 66:53-61. [PMID: 32389512 DOI: 10.1016/j.biologicals.2020.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 01/01/2023] Open
Abstract
Controlled human infection models can be helpful to study pathogenesis and immune responses as a basis for the development of vaccines. In controlled human infection models, human challenge agents are used to infect healthy volunteers, therefore, ethical considerations include that the exposure studies need to be safe and results should be meaningful, e.g. contribute to a better cure. Both in the US and in Europe, the level of Good Manufacturing Practice required is related to the phase of the study ('sliding scale Good Manufacturing Practice'), and, hence, is much more open to speedy drug development than anticipated. Recommendations included: the development of guidelines for human challenge agents; a focus on strain selection, in particular with regard to strain infectivity, stability and purity; the use of whole genome sequencing; a reference repository of challenge agents, the need for early exchange with regulators to ensure acceptability of strain selection and manufacturing for later drug development; sharing of models and challenge agents.
Collapse
Affiliation(s)
- Isabelle Bekeredjian-Ding
- Paul-Ehrlich-Institut (PEI), Langen, Germany; Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.
| | | | - Marc Baay
- P95 Epidemiology & Pharmacovigilance, Leuven, Belgium.
| | - Pieter Neels
- International Alliance for Biological Standardization (IABS), Belgium.
| | | |
Collapse
|
19
|
Kuehl CJ, D'Gama JD, Warr AR, Waldor MK. An Oral Inoculation Infant Rabbit Model for Shigella Infection. mBio 2020; 11:e03105-19. [PMID: 31964739 PMCID: PMC6974573 DOI: 10.1128/mbio.03105-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023] Open
Abstract
Shigella species cause diarrheal disease globally. Shigellosis is typically characterized by bloody stools and colitis with mucosal damage and is the leading bacterial cause of diarrheal death worldwide. After the pathogen is orally ingested, it invades and replicates within the colonic epithelium through mechanisms that rely on its type III secretion system (T3SS). Currently, oral infection-based small animal models to study the pathogenesis of shigellosis are lacking. Here, we found that orogastric inoculation of infant rabbits with Shigella flexneri resulted in diarrhea and colonic pathology resembling that found in human shigellosis. Fasting animals prior to S. flexneri inoculation increased the frequency of disease. The pathogen colonized the colon, where both luminal and intraepithelial foci were observed. The intraepithelial foci likely arise through S. flexneri spreading from cell to cell. Robust S. flexneri intestinal colonization, invasion of the colonic epithelium, and epithelial sloughing all required the T3SS as well as IcsA, a factor required for bacterial spreading and adhesion in vitro Expression of the proinflammatory chemokine interleukin 8 (IL-8), detected with in situ mRNA labeling, was higher in animals infected with wild-type S. flexneri versus mutant strains deficient in icsA or T3SS, suggesting that epithelial invasion promotes expression of this chemokine. Collectively, our findings suggest that oral infection of infant rabbits offers a useful experimental model for studies of the pathogenesis of shigellosis and for testing of new therapeutics.IMPORTANCEShigella species are the leading bacterial cause of diarrheal death globally. The pathogen causes bacillary dysentery, a bloody diarrheal disease characterized by damage to the colonic mucosa and is usually spread through the fecal-oral route. Small animal models of shigellosis that rely on the oral route of infection are lacking. Here, we found that orogastric inoculation of infant rabbits with S. flexneri led to a diarrheal disease and colonic pathology reminiscent of human shigellosis. Diarrhea, intestinal colonization, and pathology in this model were dependent on the S. flexneri type III secretion system and IcsA, canonical Shigella virulence factors. Thus, oral infection of infant rabbits offers a feasible model to study the pathogenesis of shigellosis and to develop and test new therapeutics.
Collapse
Affiliation(s)
- Carole J Kuehl
- Division of Infectious Diseases, Brigham & Women's Hospital, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan D D'Gama
- Division of Infectious Diseases, Brigham & Women's Hospital, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Alyson R Warr
- Division of Infectious Diseases, Brigham & Women's Hospital, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew K Waldor
- Division of Infectious Diseases, Brigham & Women's Hospital, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Boston, Massachusetts, USA
| |
Collapse
|
20
|
MacLennan CA, Aguilar AO, Steele AD. Consensus Report on Shigella Controlled Human Infection Model: Introduction and Overview. Clin Infect Dis 2019; 69:S577-S579. [PMID: 31816066 PMCID: PMC6901124 DOI: 10.1093/cid/ciz886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In recent years, controlled human infection models (CHIMs) have become available for a range of infectious agents and have proved invaluable for understanding the disease process, pathogenesis, and mechanisms of immunity. CHIM studies have also contributed significantly to advancing development of a number of vaccines by providing an indication of vaccine efficacy. The Shigella CHIM has been established in 3 sites in the United States, and it is likely that the CHIM will play an important regulatory role for advancing the range of Shigella vaccine candidates that are currently in development. This supplement describes the harmonization of best practices across sites, with a view to maximizing the contribution that CHIM studies can make to Shigella vaccine development.
Collapse
|
21
|
Talaat KR, Bourgeois AL, Frenck RW, Chen WH, MacLennan CA, Riddle MS, Suvarnapunya AE, Brubaker JL, Kotloff KL, Porter CK. Consensus Report on Shigella Controlled Human Infection Model: Conduct of Studies. Clin Infect Dis 2019; 69:S580-S590. [PMID: 31816068 PMCID: PMC6901126 DOI: 10.1093/cid/ciz892] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Shigella causes morbidity and mortality worldwide, primarily affecting young children living in low-resource settings. It is also of great concern due to increasing antibiotic resistance, and is a priority organism for the World Health Organization. A Shigella vaccine would decrease the morbidity and mortality associated with shigellosis, improve child health, and decrease the need for antibiotics. Controlled human infection models (CHIMs) are useful tools in vaccine evaluation for early up- or down-selection of vaccine candidates and potentially useful in support of licensure. Over time, the methods employed in these models have become more uniform across sites performing CHIM trials, although some differences in conduct persist. In November 2017, a Shigella CHIM workshop was convened in Washington, District of Columbia. Investigators met to discuss multiple aspects of these studies, including study procedures, clinical and immunological endpoints, and shared experiences. This article serves as a uniform procedure by which to conduct Shigella CHIM studies.
Collapse
Affiliation(s)
- Kawsar R Talaat
- Center for Immunization Research, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Robert W Frenck
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Wilbur H Chen
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | | | - Mark S Riddle
- F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda
| | - Akamol E Suvarnapunya
- Department of Enteric Infections, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring
| | - Jessica L Brubaker
- Global Disease Epidemiology and Control Program, Department of International Health, Johns Hopkins Bloomberg School of Public Health
| | - Karen L Kotloff
- Division of Infectious Disease and Tropical Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - Chad K Porter
- Enteric Disease Department, Naval Medical Research Center, Silver Spring, Maryland
| |
Collapse
|
22
|
Hussen S, Mulatu G, Yohannes Kassa Z. Prevalence of Shigella species and its drug resistance pattern in Ethiopia: a systematic review and meta-analysis. Ann Clin Microbiol Antimicrob 2019; 18:22. [PMID: 31288806 PMCID: PMC6617577 DOI: 10.1186/s12941-019-0321-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/29/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Shigella species are a major cause of dysentery and may attribute for death worldwide. Currently antibiotic resistance became the critical challenges for management of infectious disease. The aim was to conduct a systematic review and meta-analysis of Shigella species and its drug resistance pattern in Ethiopia. METHODS A comprehensive literature search was conducted through internet searches using database of MEDLINE, PubMed, Google scholar, EMBASE, HINARI, Cochrane Library and reference lists of previous prevalence studies from January 1999 to November 2018. Results were presented in forest plot, tables and figures with 95% CI. The Cochrane Q test and I2 test statistic were used to test heterogeneity across studies. The Pooled estimate of Shigella species and its drug resistance pattern was computed by a random effects model. RESULTS The pooled prevalence of Shigella species in Ethiopia was 6.6% (95% CI 4.7-8.8). In the subgroup analysis, the highest prevalence was observed among patients in Health facility (8.5%, 95% CI 6.2-11.5) whereas the lowest prevalence was observed in Community based studies (1.6%, 95% CI 0.8-3.4). In addition, Shigella species were highly resistant to ampicillin, amoxicillin, erythromycin and multi-drug resistant (MDR) with the pooled resistance proportions of 83.1% (95% CI 75.7-88.6), 84.1% (95% CI 75.6-90.1), 86.5% (95% CI 70.9-94.4) and 83.2% (95% CI 77.1-87.9), respectively. On the other hand, comparably low resistance pattern was reported for ciprofloxacin 8.9% (95% CI 6.0-12.8), ceftriaxone 9.3% (95% CI 3.9-20.5), and norfloxacin 8.2% (95% CI 3.8-16.6) and gentamycin 17.3% (95% CI 11.2-25.9). Subgroup analyses indicated that study years were associated with a decreasing Shigella prevalence over time (p = 0.002). CONCLUSION The pooled estimate showed high burden of Shigella infection and its high proportion of drug resistance pattern to ampicillin, amoxicillin and erythromycin in Ethiopia. Therefore, initiating and scale up of performing drug susceptibility test for each shigellosis case, educate the community and health care providers on appropriate use of antibiotics need to be considered and strengthened.
Collapse
Affiliation(s)
- Siraj Hussen
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Hawassa University, Hawassa, Ethiopia
| | - Getamesay Mulatu
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Hawassa University, Hawassa, Ethiopia
| | - Zemenu Yohannes Kassa
- School of Nursing and Midwifery, College of Medicine and Health Sciences, Hawassa University, Hawassa, Ethiopia
| |
Collapse
|
23
|
Clinical endpoints for efficacy studies. Vaccine 2019; 37:4814-4822. [PMID: 30981626 DOI: 10.1016/j.vaccine.2019.03.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/19/2019] [Accepted: 03/21/2019] [Indexed: 12/21/2022]
Abstract
Well-established, validated and clinically meaningful primary and secondary endpoints are critical in advancing vaccines through proof of principal studies, licensure and pre-qualification. To that end, the field of vaccine development for Shigella, enterotoxigenic Escherichia coli (ETEC) as well as other enteric pathogens would benefit greatly from a focused review of clinical endpoints and the use of common endpoints across the field to enable study-to-study comparisons as well as comparative assessments between vaccine candidates. A workshop was conducted to review clinical endpoints from controlled human challenge studies, field studies in naïve adult travelers and pediatric studies in low-middle income countries and to develop a consensus on clinical endpoints for future vaccine trials. Following sequential presentations on different study designs (CHIM, travelers' efficacy and pediatric efficacy), workshop participants broke into three simultaneous workgroups focused on those study designs to discuss a number of topics key to clinical endpoints specific to each study design. Previously utilized endpoints were reviewed with an eye towards potentially novel endpoints for future studies and consideration of the disease parameters and spectrum of disease targeted for prevention. The strength of support among workshop participants for the use of various endpoints is summarized as are recommendations for additional endpoints to be considered in future studies. It is anticipated that this report will facilitate endpoint determination in future efficacy trials of vaccine candidates.
Collapse
|
24
|
Abstract
Zika virus (ZIKV), a previously little known arbovirus, caused an unprecedented outbreak in Latin America and the Caribbean throughout 2015 and 2016. The virus has been associated with the congenital Zika syndrome (CZS), which can occur with maternal ZIKV infection during any trimester and can result from asymptomatic infection. There is concern that even low levels of viremia can result in CZS, meaning an effective vaccine will need to induce very high levels of protection. Controlled human infection models (CHIMs), in which subjects are infected with a pathogen of interest, have been used to down-select vaccine candidates and have provided efficacy data in support of vaccine licensure.A ZIKV CHIM could be instrumental in determining which of the many ZIKV vaccine candidates provides the highest degree of protection and should be advanced in clinical development. The development of a ZIKV CHIM is not without challenges. The ZIKV, unlike other flaviviruses, is sexually and mosquito-transmitted, and an increase in the incidence of Guillain-Barré syndrome was reported in some countries during the ZIKV outbreak. These obstacles can be overcome with thoughtful study design to ensure maximal risk mitigation. If successful, a ZIKV CHIM could de-risk and accelerate ZIKV vaccine development.
Collapse
Affiliation(s)
- Anna P Durbin
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Stephen S Whitehead
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
25
|
Diemert DJ, Bottazzi ME, Plieskatt J, Hotez PJ, Bethony JM. Lessons along the Critical Path: Developing Vaccines against Human Helminths. Trends Parasitol 2018; 34:747-758. [PMID: 30064902 DOI: 10.1016/j.pt.2018.07.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/16/2022]
Abstract
Helminthic parasites are important targets for vaccine research as they infect an estimated 1 billion people worldwide. Despite significant progress in the discovery of defined antigens as candidates for vaccines, the potential of a helminth vaccine advancing to an investigational product to be tested in humans remains as challenging as it did 50 years ago. Candidate helminth vaccines must still advance along a 'critical path' of preclinical research, vaccine process development (which includes 'chemistry, manufacturing, and controls' or CMC), current good manufacturing practice (cGMP) production of the vaccine, and clinical trials. This path is highly targeted towards meeting the safety, immunogenicity, and efficacy criteria of regulatory bodies such as the US Food and Drug Administration (FDA). For nearly 20 years our product development partnership (PDP), the Texas Children's Hospital Center for Vaccine Development (TCH-CVD), has followed the critical paths of several novel subunit vaccines for the human hookworm Necator americanus and the intestinal trematode Schistosoma mansoni. Herein, we describe the critical lessons learned along this critical path.
Collapse
Affiliation(s)
- David J Diemert
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington DC, USA; Department of Medicine, The George Washington University, Washington DC, USA; Texas Children's Hospital Center for Vaccine Development - a Product Development Partnership, Houston, TX, USA
| | - Maria Elena Bottazzi
- Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA; Department of Biology, Baylor University, Waco, TX, USA; Texas Children's Hospital Center for Vaccine Development - a Product Development Partnership, Houston, TX, USA
| | - Jordan Plieskatt
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington DC, USA; Texas Children's Hospital Center for Vaccine Development - a Product Development Partnership, Houston, TX, USA
| | - Peter J Hotez
- Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA; Department of Biology, Baylor University, Waco, TX, USA; Texas Children's Hospital Center for Vaccine Development - a Product Development Partnership, Houston, TX, USA
| | - Jeffrey M Bethony
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington DC, USA; Texas Children's Hospital Center for Vaccine Development - a Product Development Partnership, Houston, TX, USA.
| |
Collapse
|
26
|
Roestenberg M, Hoogerwerf MA, Ferreira DM, Mordmüller B, Yazdanbakhsh M. Experimental infection of human volunteers. THE LANCET. INFECTIOUS DISEASES 2018; 18:e312-e322. [PMID: 29891332 DOI: 10.1016/s1473-3099(18)30177-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 02/08/2018] [Accepted: 03/01/2018] [Indexed: 12/13/2022]
Abstract
Controlled human infection (CHI) trials, in which healthy volunteers are experimentally infected, can accelerate the development of novel drugs and vaccines for infectious diseases of global importance. The use of CHI models is expanding from around 60 studies in the 1970s to more than 120 publications in this decade, primarily for influenza, rhinovirus, and malaria. CHI trials have provided landmark data for several registered drugs and vaccines, and have generated unprecedented scientific insights. Because of their invasive nature, CHI studies demand critical ethical review according to established frameworks. CHI-associated serious adverse events are rarely reported. Novel CHI models need standardised safety data from comparable CHI models to facilitate evidence-based risk assessments, as well as funds to produce challenge inoculum according to regulatory requirements. Advances such as the principle of controlled colonisation, the expansion of models to endemic areas, and the use of genetically attenuated strains will further broaden the scope of CHI trials.
Collapse
Affiliation(s)
| | | | | | - Benjamin Mordmüller
- Institute of Tropical Medicine and German Center for Infection Research, partner site Tübingen, University of Tübingen, Tübingen, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | | |
Collapse
|
27
|
Porter CK, Lynen A, Riddle MS, Talaat K, Sack D, Gutiérrez RL, McKenzie R, DeNearing B, Feijoo B, Kaminski RW, Taylor DN, Kirkpatrick BD, Bourgeois AL. Clinical endpoints in the controlled human challenge model for Shigella: A call for standardization and the development of a disease severity score. PLoS One 2018; 13:e0194325. [PMID: 29590182 PMCID: PMC5874036 DOI: 10.1371/journal.pone.0194325] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 02/28/2018] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Since 1946 the controlled human infection model (CHIM) for Shigella has been used to improve understanding of disease pathogenesis, describe clinical and immunologic responses to infection and as a tool for vaccine development. As the frequency and intent for use in vaccine comparisons increases, standardization of the primary endpoint definition is necessary. METHODS Subject-level data were obtained from previously conducted experimental Shigella CHIM studies. Signs and symptoms severity were categorized consistently across all studies. Sign and symptom correlations were estimated and univariate models were utilized to describe the association between stool output and other Shigella-attributable signs and symptoms. Multiple correspondence and hierarchical clustering analyses were performed to describe the co-occurrence of signs and symptoms. A disease score is proposed based on the co-occurrence of these events. RESULTS Data were obtained on 54 subjects receiving 800 to 2000 colony forming units (cfu) of S. flexneri. The median maximum 24 hour stool output was 514 ml (IQR: 300, 998 ml) with a median frequency of 6 (IQR: 4, 9). Subjects reported abdominal pain or cramps (81.5%), headache (66.7%) and anorexia (64.8%), 50.0% had a fever and 27.8% had gross blood in multiple loose stools. Multiple correspondence analyses highlighted co-occurrence of symptoms based on severity. A 3-parameter disease severity score predicted shigellosis endpoints and better differentiated disease spectrum. CONCLUSION Dichotomous endpoints for Shigella CHIM fail to fully account for disease variability. An ordinal disease score characterizing the breadth of disease severity may enable a better characterization of shigellosis and can decrease sample size requirements. Furthermore, the disease severity score may be a useful tool for portfolio management by enabling prioritization across vaccine candidates with comparable efficacy estimates using dichotomous endpoints.
Collapse
Affiliation(s)
- Chad K. Porter
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
- * E-mail:
| | - Amanda Lynen
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Mark S. Riddle
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Kawsar Talaat
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - David Sack
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Ramiro L. Gutiérrez
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Robin McKenzie
- School of Medicine, Johns Hopkins University, Baltimore, MD, United States of America
| | - Barbara DeNearing
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Brittany Feijoo
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Robert W. Kaminski
- Department of Enteric Infections, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - David N. Taylor
- Drug Development Global Program, PATH, Seattle, WA, United States of America
| | - Beth D. Kirkpatrick
- University of Vermont College of Medicine, Vaccine Testing Center, Department of Medicine, Burlington, VT, United States of America
| | - A. Louis Bourgeois
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
- Enteric Vaccine Initiative, PATH, Washington, DC, United States of America
| |
Collapse
|
28
|
Baay MFD, Richie TL, Neels P. Human challenge trials in vaccine development, Rockville, MD, USA, September 28-30, 2017. Biologicals 2018; 61:85-94. [PMID: 29573967 DOI: 10.1016/j.biologicals.2018.02.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 02/21/2018] [Indexed: 11/17/2022] Open
Abstract
The International Alliance for Biological Standardization organized the second workshop on human challenge trials (HCT) in Rockville, MD, in September 2017. The objective of this meeting was to examine the use of HCT, in response to the continuing human suffering caused by infectious diseases, preventable by the development of new and improved vaccines. For this, the approach of HCT could be valuable, as HCT can provide key safety, tolerability, immunogenicity, and efficacy data, and can be used to study host-pathogen biology. HCT can generate these data with speed, efficiency and minimal expense, albeit not with the same level of robustness as clinical trials. Incorporated wisely into a clinical development plan, HCT can support optimization or down-selection of new vaccine candidates, assuring that only the worthiest candidates progress to field testing. HCT may also provide pivotal efficacy data in support of licensure, particularly when field efficacy studies are not feasible. Many aspects of HCT were discussed by the participants, including new and existing models, standardization and ethics. A consensus was achieved that HCT, if ethically justified and performed with careful attention to safety and informed consent, should be pursued to promote and accelerate vaccine development.
Collapse
Affiliation(s)
- Marc F D Baay
- P95 Pharmacovigilance and Epidemiology Services, Leuven, Belgium.
| | - Thomas L Richie
- Sanaria Institute for Global Health and Tropical Medicine, Rockville, MD, USA.
| | - Pieter Neels
- International Alliance for Biological Standardization, Lyon, France.
| | | |
Collapse
|
29
|
Davis CL, Wahid R, Toapanta FR, Simon JK, Sztein MB. A clinically parameterized mathematical model of Shigella immunity to inform vaccine design. PLoS One 2018; 13:e0189571. [PMID: 29304144 PMCID: PMC5755796 DOI: 10.1371/journal.pone.0189571] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/16/2017] [Indexed: 11/28/2022] Open
Abstract
We refine and clinically parameterize a mathematical model of the humoral immune response against Shigella, a diarrheal bacteria that infects 80-165 million people and kills an estimated 600,000 people worldwide each year. Using Latin hypercube sampling and Monte Carlo simulations for parameter estimation, we fit our model to human immune data from two Shigella EcSf2a-2 vaccine trials and a rechallenge study in which antibody and B-cell responses against Shigella′s lipopolysaccharide (LPS) and O-membrane proteins (OMP) were recorded. The clinically grounded model is used to mathematically investigate which key immune mechanisms and bacterial targets confer immunity against Shigella and to predict which humoral immune components should be elicited to create a protective vaccine against Shigella. The model offers insight into why the EcSf2a-2 vaccine had low efficacy and demonstrates that at a group level a humoral immune response induced by EcSf2a-2 vaccine or wild-type challenge against Shigella′s LPS or OMP does not appear sufficient for protection. That is, the model predicts an uncontrolled infection of gut epithelial cells that is present across all best-fit model parameterizations when fit to EcSf2a-2 vaccine or wild-type challenge data. Using sensitivity analysis, we explore which model parameter values must be altered to prevent the destructive epithelial invasion by Shigella bacteria and identify four key parameter groups as potential vaccine targets or immune correlates: 1) the rate that Shigella migrates into the lamina propria or epithelium, 2) the rate that memory B cells (BM) differentiate into antibody-secreting cells (ASC), 3) the rate at which antibodies are produced by activated ASC, and 4) the Shigella-specific BM carrying capacity. This paper underscores the need for a multifaceted approach in ongoing efforts to design an effective Shigella vaccine.
Collapse
Affiliation(s)
- Courtney L. Davis
- Natural Science Division, Pepperdine University, Malibu, CA, United States of America
- * E-mail:
| | - Rezwanul Wahid
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Franklin R. Toapanta
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Jakub K. Simon
- Merck & Co. Inc. Kenilworth, NJ, United States of America
| | - Marcelo B. Sztein
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
30
|
Mani S, Wierzba T, Walker RI. Status of vaccine research and development for Shigella. Vaccine 2016; 34:2887-2894. [DOI: 10.1016/j.vaccine.2016.02.075] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 02/10/2016] [Indexed: 12/21/2022]
|
31
|
Porter CK, Riddle MS, Alcala AN, Sack DA, Harro C, Chakraborty S, Gutierrez RL, Savarino SJ, Darsley M, McKenzie R, DeNearing B, Steinsland H, Tribble DR, Bourgeois AL. An Evidenced-Based Scale of Disease Severity following Human Challenge with Enteroxigenic Escherichia coli. PLoS One 2016; 11:e0149358. [PMID: 26938983 PMCID: PMC4777366 DOI: 10.1371/journal.pone.0149358] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/29/2016] [Indexed: 11/30/2022] Open
Abstract
Background Experimental human challenge models have played a major role in enhancing our understanding of infectious diseases. Primary outcomes have typically utilized overly simplistic outcomes that fail to entirely account for complex illness syndromes. We sought to characterize clinical outcomes associated with experimental infection with enterotoxigenic Escherichia coli (ETEC) and to develop a disease score. Methods Data were obtained from prior controlled human ETEC infection studies. Correlation and univariate regression across sign and symptom severity was performed. A multiple correspondence analysis was conducted. A 3-parameter disease score with construct validity was developed in an iterative fashion, compared to standard outcome definitions and applied to prior vaccine challenge trials. Results Data on 264 subjects receiving seven ETEC strains at doses from 1x105 to 1x1010 cfu were used to construct a standardized dataset. The strongest observed correlation was between vomiting and nausea (r = 0.65); however, stool output was poorly correlated with subjective activity-impacting outcomes. Multiple correspondence analyses showed covariability in multiple signs and symptoms, with severity being the strongest factor corresponding across outcomes. The developed disease score performed well compared to standard outcome definitions and differentiated disease in vaccinated and unvaccinated subjects. Conclusion Frequency and volumetric definitions of diarrhea severity poorly characterize ETEC disease. These data support a disease severity score accounting for stool output and other clinical signs and symptoms. Such a score could serve as the basis for better field trial outcomes and gives an additional outcome measure to help select future vaccines that warrant expanded testing in pivotal pre-licensure trials.
Collapse
Affiliation(s)
- Chad K. Porter
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
- * E-mail:
| | - Mark S. Riddle
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Ashley N. Alcala
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - David A. Sack
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Clayton Harro
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Subhra Chakraborty
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Ramiro L. Gutierrez
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Stephen J. Savarino
- Enteric Disease Department, Infectious Disease Directorate, Naval Medical Research Center, Silver Spring, MD, United States of America
| | | | - Robin McKenzie
- School of Medicine, Johns Hopkins University, Baltimore, MD, United States of America
| | - Barbara DeNearing
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Hans Steinsland
- Centre for Intervention Science in Maternal and Child Health (CISMAC), Centre for International Health, and Department of Biomedicine, University of Bergen, Bergen, Norway
| | - David R. Tribble
- Infectious Disease Clinical Research Program, Bethesda, MD, United States of America
| | - A. Louis Bourgeois
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
- PATH, Washington, DC, United States of America
| |
Collapse
|
32
|
Zak DE, Aderem A. Systems integration of innate and adaptive immunity. Vaccine 2015; 33:5241-8. [PMID: 26102534 DOI: 10.1016/j.vaccine.2015.05.098] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/21/2015] [Accepted: 05/28/2015] [Indexed: 12/20/2022]
Abstract
The pathogens causing AIDS, malaria, and tuberculosis have proven too complex to be overcome by classical approaches to vaccination. The complexities of human immunology and pathogen-induced modulation of the immune system mandate new approaches to vaccine discovery and design. A new field, systems vaccinology, weds holistic analysis of innate and adaptive immunity within a quantitative framework to enable rational design of new vaccines that elicit tailored protective immune responses. A key step in the approach is to discover relationships between the earliest innate inflammatory responses to vaccination and the subsequent vaccine-induced adaptive immune responses and efficacy. Analysis of these responses in clinical studies is complicated by the inaccessibility of relevant tissue compartments (such as the lymph node), necessitating reliance upon peripheral blood responses as surrogates. Blood transcriptomes, although indirect to vaccine mechanisms, have proven very informative in systems vaccinology studies. The approach is most powerful when innate and adaptive immune responses are integrated with vaccine efficacy, which is possible for malaria with the advent of a robust human challenge model. This is more difficult for AIDS and tuberculosis, given that human challenge models are lacking and efficacy observed in clinical trials has been low or highly variable. This challenge can be met by appropriate clinical trial design for partially efficacious vaccines and by analysis of natural infection cohorts. Ultimately, systems vaccinology is an iterative approach in which mechanistic hypotheses-derived from analysis of clinical studies-are evaluated in model systems, and then used to guide the development of new vaccine strategies. In this review, we will illustrate the above facets of the systems vaccinology approach with case studies.
Collapse
Affiliation(s)
- Daniel E Zak
- The Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, 307 Westlake Ave N, Suite 500, Seattle, WA 98109, USA
| | - Alan Aderem
- The Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, 307 Westlake Ave N, Suite 500, Seattle, WA 98109, USA.
| |
Collapse
|
33
|
Darton TC, Blohmke CJ, Moorthy VS, Altmann DM, Hayden FG, Clutterbuck EA, Levine MM, Hill AVS, Pollard AJ. Design, recruitment, and microbiological considerations in human challenge studies. THE LANCET. INFECTIOUS DISEASES 2015; 15:840-51. [PMID: 26026195 DOI: 10.1016/s1473-3099(15)00068-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 01/13/2015] [Accepted: 01/15/2015] [Indexed: 12/26/2022]
Abstract
Since the 18th century a wealth of knowledge regarding infectious disease pathogenesis, prevention, and treatment has been accumulated from findings of infection challenges in human beings. Partly because of improvements to ethical and regulatory guidance, human challenge studies-involving the deliberate exposure of participants to infectious substances-have had a resurgence in popularity in the past few years, in particular for the assessment of vaccines. To provide an overview of the potential use of challenge models, we present historical reports and contemporary views from experts in this type of research. A range of challenge models and practical approaches to generate important data exist and are used to expedite vaccine and therapeutic development and to support public health modelling and interventions. Although human challenge studies provide a unique opportunity to address complex research questions, participant and investigator safety is paramount. To increase the collaborative effort and future success of this area of research, we recommend the development of consensus frameworks and sharing of best practices between investigators. Furthermore, standardisation of challenge procedures and regulatory guidance will help with the feasibility for using challenge models in clinical testing of new disease intervention strategies.
Collapse
Affiliation(s)
- Thomas C Darton
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK
| | - Christoph J Blohmke
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK.
| | - Vasee S Moorthy
- Department of Immunisation, Vaccines and Biologicals, WHO, Geneva, Switzerland
| | | | - Frederick G Hayden
- Department of Medicine, University of Virginia School of Medicine, Charlottesville VA, USA
| | - Elizabeth A Clutterbuck
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK
| | - Myron M Levine
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Adrian V S Hill
- The Jenner Institute Laboratories, University of Oxford, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
34
|
Abstract
Salmonella, Shigella, and Yersinia cause a well-characterized spectrum of disease in humans, ranging from asymptomatic carriage to hemorrhagic colitis and fatal typhoidal fever. These pathogens are responsible for millions of cases of food-borne illness in the United States each year, with substantial costs measured in hospitalizations and lost productivity. In the developing world, illness caused by these pathogens is not only more prevalent but also associated with a greater case-fatality rate. Classic methods for identification rely on selective media and serology, but newer methods based on mass spectrometry and polymerase chain reaction show great promise for routine clinical testing.
Collapse
|
35
|
Abstract
Renewed awareness of the substantial morbidity and mortality that Shigella infection causes among young children in developing countries, combined with technological innovations in vaccinology, has led to the development of novel vaccine strategies in the past 5 years. Along with advancement of classic vaccines in clinical trials and new sophisticated measurements of immunological responses, much new data has been produced, lending promise to the potential for production of safe and effective Shigella vaccines. Herein, we review the latest progress in Shigella vaccine development within the framework of persistent obstacles.
Collapse
|