1
|
Ndungo E, Bhaumik U, Liang Y, Chen WH, Travassos MA, Tapia MD, Kotloff KL, Levine MM, Pasetti MF. Shigella humoral immunity during the first 2 years of life in children from endemic areas. mBio 2025; 16:e0055525. [PMID: 40237475 PMCID: PMC12077217 DOI: 10.1128/mbio.00555-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Shigella is a leading cause of moderate-to-severe diarrhea, primarily affecting children in impoverished regions. Disease incidence is highest in toddlers 1-2 years of age. Acquisition of immunity over time reduces the risk of infection. However, the evolution of childhood immunity resulting from Shigella exposure and the host responses that mediate protection remain poorly understood. Gaining insight into the immunological features that develop over time and prevent subsequent Shigella infection is critical for guiding vaccine design. We examined the antigenic repertoire and magnitude of humoral immunity to Shigella in children < 2 years of age from three endemic areas-Bamako, Mali, and two sites (Afar and Tigray) in Ethiopia-using a qualified multiplex assay. Serum IgG and IgA specific to Shigella proteins (IpaB, IpaC, IpaD, IpaH, and VirG) and lipopolysaccharide (LPS) from S. flexneri 2a, S. flexneri 3a, S. flexneri 6, and S. sonnei were measured in three stratified age groups: 6-8, 12-17, and 18-24 months. The youngest group (6- to 8-month-olds) exhibited similar antibody profiles across all sites. By contrast, antibody levels in the 12- to 17- and 18- to 24-month-old age groups varied by both age and site, reflecting geographical differences in Shigella endemicity and exposure. Notably, most children in the 12- to 17-month-old age group had IgG levels below the adult protective thresholds identified in controlled human infection models, identifying a critical window of vulnerability that would require intervention. Our findings underscore the importance and value of Shigella serosurveillance in children from endemic regions to inform future vaccine rollout decisions.IMPORTANCEShigella is a major cause of moderate-to-severe diarrhea, the most affected being young children from poor resource countries. Shigella species easily acquire antibiotic resistance, presenting a challenge to infection control. The development of vaccines for young children has been hindered by a lack of understanding of what constitutes protective immunity. Here, for the first time, we investigated the magnitude and specificity of Shigella humoral immunity evoked by natural exposure in children 6 months to 2 years old living in Mali and Ethiopia (Shigella-endemic areas) using a qualified multiplex assay. Antibody profiles varied with age and region, revealing epidemiological trends. Children 12-17 months old were identified as the most vulnerable to infection. Antibodies specific for conserved Shigella proteins were higher in older children, affirming their potential as vaccine candidates. Shigella serosurveillance is useful in guiding public health interventions.
Collapse
Affiliation(s)
- Esther Ndungo
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ushasi Bhaumik
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yuanyuan Liang
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Wilbur H. Chen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mark A. Travassos
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Milagritos D. Tapia
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Karen L. Kotloff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Myron M. Levine
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Marcela F. Pasetti
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Scott TA, Baker KS, Trotter C, Jenkins C, Mostowy S, Hawkey J, Schmidt H, Holt KE, Thomson NR, Baker S. Shigella sonnei: epidemiology, evolution, pathogenesis, resistance and host interactions. Nat Rev Microbiol 2025; 23:303-317. [PMID: 39604656 DOI: 10.1038/s41579-024-01126-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
Shigella sonnei is a major cause of diarrhoea globally and is increasing in prevalence relative to other Shigella because of multiple demographic and environmental influences. This single-serotype species has traditionally received less attention in comparison to Shigella flexneri and Shigella dysenteriae, which were more common in low-income countries and more tractable in the laboratory. In recent years, we have learned that Shigella are highly complex and highly susceptible to environmental change, as exemplified by epidemiological trends and increasing relevance of S. sonnei. Ultimately, methods, tools and data generated from decades of detailed research into S. flexneri have been used to gain new insights into the epidemiology, microbiology and pathogenesis of S. sonnei. In parallel, widespread adoption of genomic surveillance has yielded insights into antimicrobial resistance, evolution and organism transmission. In this Review, we provide an overview of current knowledge of S. sonnei, highlighting recent insights into this globally disseminated antimicrobial-resistant pathogen and assessing how novel data may impact future vaccine development and implementation.
Collapse
Affiliation(s)
- Timothy A Scott
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Department of Medicine, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
| | - Kate S Baker
- Department of Clinical Microbiology, Immunology and Infection, University of Liverpool, Liverpool, UK
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Caroline Trotter
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | - Serge Mostowy
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Jane Hawkey
- Department of Infectious Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Hayden Schmidt
- Neutralizing Antibody Center, International AIDS Vaccine Initiative, San Diego, CA, USA
| | - Kathryn E Holt
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
- Department of Infectious Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Nicholas R Thomson
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Stephen Baker
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Department of Medicine, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
- International AIDS Vaccine Initiative, London, UK.
| |
Collapse
|
3
|
Randall AZ, Conti V, Nakakana U, Liang X, Teng AA, Di Pasquale AL, Kapulu M, Frenck R, Launay O, Ferruzzi P, Sciré AS, Marchetti E, Obiero C, Pablo JV, Edgar J, Bejon P, Shandling AD, Campo JJ, Yee A, Martin LB, Podda A, Micoli F. Protein-specific immune response elicited by the Shigella sonnei 1790GAHB GMMA-based candidate vaccine in adults with varying exposure to Shigella. mSphere 2025:e0105724. [PMID: 40237462 DOI: 10.1128/msphere.01057-24] [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: 12/16/2024] [Accepted: 02/25/2025] [Indexed: 04/18/2025] Open
Abstract
Shigella is a leading cause of diarrheal morbidity and mortality in young children from low- and middle-income countries. Here, we aimed to verify the ability of the generalized modules for membrane antigens (GMMA)-based Shigella sonnei candidate vaccine 1790GAHB to elicit an anti-protein antibody response. Serum samples from previous clinical trials in adults (a dose-escalation study and its extension in France, a vaccine efficacy study after human challenge in the United States, and a study in Kenya) were investigated using pan-proteome microarrays consisting of 3,150 full-length or fragmented Shigella proteins. Pre-/post-vaccination comparisons identified subsets of proteins that were highly immunoreactive and largely overlapped across all trials; the T3SS lipochaperone family protein (expressed on GMMA) was the most reactive in all studies. Responses to several microarray antigens correlated well with S. sonnei LPS serum IgG antibody levels. Overall, we confirmed the ability of GMMA to elicit an anti-protein IgG/IgA response; however, no association with protection against shigellosis was identified. In the challenge study, IgG response to seven antigens (IpaC, IpaB, IpaA, IpaD, IpaH, IpgC, and MxiD; not expressed on GMMA) was associated with a decreased risk of shigellosis. These antigens were observed to also have high IgG responses at baseline in individuals naturally exposed to Shigella and could constitute targets for future vaccine development.IMPORTANCEShigella remains a major cause of diarrheal disease, especially in children aged under 5 years from low-to-middle-income countries. No vaccine against shigellosis is yet widely available despite the high public health need. An ideal vaccine would provide protection against the most prevalent species, Shigella flexneri and Shigella sonnei; therefore, it could be relevant to identify common antigens. We developed a microarray containing 3,150 full-length or fragmented proteins selected across Shigella species. Sera collected in four clinical trials conducted in three countries of varying endemicity to evaluate a S. sonnei GMMA-based candidate vaccine were tested against these proteins. We identified several Shigella proteins (IpaC, IpaB, IpaA, IpaD, IpaH, IpgC, MxiD) that induced robust antibody response following experimental challenge or natural infection. These proteins correlated with a reduced risk of shigellosis after the S. sonnei challenge. We found no apparent role for anti-GMMA proteins' IgG or IgA response in protection against shigellosis.
Collapse
Affiliation(s)
| | | | | | - Xiaowu Liang
- Antigen Discovery, Inc. (ADI), Irvine, California, USA
| | - Andy A Teng
- Antigen Discovery, Inc. (ADI), Irvine, California, USA
| | | | - Melissa Kapulu
- Biosciences Department, Kenya Medical Research Institute (KEMRI)-Wellcome Trust Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Robert Frenck
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Odile Launay
- Université Paris Cité; Assistance Publique Hôpitaux de Paris, CIC Cochin Pasteur; Inserm, Paris, France
| | | | | | | | - Christina Obiero
- Clinical Research Department, Kenya Medical Research Institute (KEMRI)-Wellcome Trust Programme, Kilifi, Kenya
| | | | - Joshua Edgar
- Antigen Discovery, Inc. (ADI), Irvine, California, USA
| | - Philip Bejon
- Biosciences Department, Kenya Medical Research Institute (KEMRI)-Wellcome Trust Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | | | - Angela Yee
- Antigen Discovery, Inc. (ADI), Irvine, California, USA
| | | | - Audino Podda
- GSK Vaccines Institute for Global Health, Siena, Italy
| | | |
Collapse
|
4
|
Hwang W, Wantuch PL, Bernshtein B, Zhiteneva JA, Slater DM, Vater KH, Sridhar S, Oliver E, Roach DJ, Rao SR, Turbett SE, Knoot CJ, Harding CM, Amin MN, Cross AS, LaRocque RC, Rosen DA, Harris JB. Antibody responses in Klebsiella pneumoniae bloodstream infection: a prospective cohort study. THE LANCET. MICROBE 2025; 6:100988. [PMID: 39952262 DOI: 10.1016/j.lanmic.2024.100988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/08/2024] [Accepted: 08/28/2024] [Indexed: 02/17/2025]
Abstract
BACKGROUND Klebsiella pneumoniae is a leading cause of infection-related deaths globally, yet little is known about human antibody responses to invasive K pneumoniae. We sought to determine whether the O-specific polysaccharide antigen is immunogenic in humans with K pneumoniae bloodstream infection. We also sought to define the cross-reactivity of human antibody responses among structurally related K pneumoniae O-specific polysaccharide subtypes and to assess the effect of capsule production on O-specific polysaccharide-targeted antibody binding and function. METHODS In this prospective cohort study, we compared plasma antibody responses to O-specific polysaccharide in a cohort of consecutively enrolled patients with K pneumoniae bloodstream infection with controls, specifically a cohort of healthy individuals and a cohort of individuals with Enterococcus spp bloodstream infection. Patients were enrolled at the Massachusetts General Hospital, a tertiary hospital with affiliated clinics in the USA. We excluded patients whose isolates were not confirmed to be K pneumoniae by whole-genome sequencing. The primary outcome was the measurement of plasma IgG, IgM, and IgA antibody responses. We performed flow cytometry to measure the effects of K pneumoniae capsule production on O-specific polysaccharide antibody binding and O-specific polysaccharide antibody-mediated complement deposition, using patient isolates with variable levels of capsule production and isogenic capsule-deficient strains derived from these isolates. FINDINGS We enrolled 129 consecutive patients with suspected K pneumoniae bloodstream infection between July 24, 2021, and August 4, 2022, of whom 69 patients (44 [64%] male and 25 [36%] female) with confirmed K pneumoniae bloodstream infection were eligible for immunological evaluation. Common O-specific polysaccharide serotypes (O1, O2, O3, and O5) accounted for 57 (83%) of 69 infections. O-specific polysaccharide was immunogenic in patients with K pneumoniae bloodstream infection, and peak O-specific polysaccharide-IgG antibody responses in patients were ten-fold to 30-fold higher than antibody responses detected in healthy controls, depending on the serotype. There was cross-reactivity among similar O-specific polysaccharide subtypes, including the O1v1 and O1v2, O2v1 and O2v2, and O3 and O3b subtypes, as well as between the O1 and O2 types. Capsule produced by both hyperencapsulated and non-hyperencapsulated K pneumoniae inhibited O-specific polysaccharide-targeted antibody binding and function. INTERPRETATION O-specific polysaccharide was immunogenic in patients with K pneumoniae bloodstream infection, supporting its potential as a candidate vaccine antigen. The cross-reactivity observed between similar O-specific polysaccharide subtypes in patients with K pneumoniae bloodstream infection suggests that it might not be necessary to include all subtypes in an O-specific polysaccharide-based vaccine. However, these observations are tempered by the fact that capsule production, even in non-highly encapsulated strains, has the potential to interfere with O-specific polysaccharide antibody binding. This finding could limit the effectiveness of vaccines that exclusively target O-specific polysaccharide. FUNDING National Institute of Allergy and Infectious Diseases at the National Institutes of Health.
Collapse
Affiliation(s)
- Wontae Hwang
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Paeton L Wantuch
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Biana Bernshtein
- Ragon Institute of Massachusetts General Hospital, Harvard and MIT, Cambridge, MA, USA
| | - Julia A Zhiteneva
- Ragon Institute of Massachusetts General Hospital, Harvard and MIT, Cambridge, MA, USA
| | - Damien M Slater
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Kian Hutt Vater
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Sushmita Sridhar
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Elizabeth Oliver
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - David J Roach
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Sowmya R Rao
- Department of Global Health, Boston University of Public Health, Boston, MA, USA
| | - Sarah E Turbett
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | | | - Mohammed Nurul Amin
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan S Cross
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Regina C LaRocque
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David A Rosen
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jason B Harris
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Gomez Toledo A, Chowdhury S, Hjortswang E, Sorrentino JT, Lewis NE, Bläckberg A, Ekström S, Kjellström S, Izadi A, Olofsson B, Nordenfelt P, Malmström L, Rasmussen M, Malmström J. Dissecting the properties of circulating IgG against streptococcal pathogens through a combined systems antigenomics-serology workflow. Nat Commun 2025; 16:1942. [PMID: 39994218 PMCID: PMC11850916 DOI: 10.1038/s41467-025-57170-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
This study showcases an integrative mass spectrometry-based strategy combining systems antigenomics and systems serology to characterize human antibodies in clinical samples. This strategy involves using antibodies circulating in plasma to affinity-enrich antigenic proteins in biochemically fractionated pools of bacterial proteins, followed by their identification and quantification using mass spectrometry. A selected subset of the identified antigens is then expressed recombinantly to isolate antigen-specific IgG, followed by characterization of the structural and functional properties of these antibodies. We focused on Group A streptococcus (GAS), a major human pathogen lacking an approved vaccine. The data shows that both healthy and GAS-infected individuals have circulating IgG against conserved streptococcal proteins, including toxins and virulence factors. The antigenic breadth of these antibodies remains relatively constant across healthy individuals but changes considerably in GAS bacteremia. Moreover, antigen-specific IgG analysis reveals individual variation in titers, subclass distributions, and Fc-signaling capacity, despite similar epitope and Fc-glycosylation patterns. Finally, we show that GAS antibodies may cross-react with Streptococcus dysgalactiae (SD), a bacterial pathogen that occupies similar niches and causes comparable infections. Collectively, our results highlight the complexity of GAS-specific antibody responses and the versatility of our methodology to characterize immune responses to bacterial pathogens.
Collapse
Affiliation(s)
- Alejandro Gomez Toledo
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sounak Chowdhury
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Elisabeth Hjortswang
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - James T Sorrentino
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, CA, USA
| | - Nathan E Lewis
- Departments of Pediatrics and Bioengineering, University of California, San Diego, CA, USA
| | - Anna Bläckberg
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | | | | | - Arman Izadi
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Berit Olofsson
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Pontus Nordenfelt
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Lars Malmström
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Magnus Rasmussen
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Johan Malmström
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden.
- BioMS, Lund, Sweden.
| |
Collapse
|
6
|
Haldar R, Halder P, Koley H, Miyoshi SI, Das S. A newly developed oral infection mouse model of shigellosis for immunogenicity and protective efficacy studies of a candidate vaccine. Infect Immun 2025; 93:e0034624. [PMID: 39692481 PMCID: PMC11784180 DOI: 10.1128/iai.00346-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/06/2024] [Indexed: 12/19/2024] Open
Abstract
Shigella infection poses a significant public health challenge in the developing world. However, lack of a widely available mouse model that replicates human shigellosis creates a major bottleneck to better understanding of disease pathogenesis and development of newer drugs and vaccines. BALB/c mice pre-treated with streptomycin and iron (FeCl3) plus desferrioxamine intraperitoneally followed by oral infection with virulent Shigella flexneri 2a resulted in diarrhea, loss of body weight, bacterial colonization and progressive colitis characterized by disruption of epithelial lining, loss of crypt architecture with goblet cell depletion, increased polymorphonuclear infiltration into the mucosa, submucosal swelling (edema), and raised proinflammatory cytokines and chemokines in the large intestine. To evaluate the usefulness of the model for vaccine efficacy studies, mice were immunized intranasally with a recombinant protein vaccine containing Shigella invasion protein invasion plasmid antigen B (IpaB). Vaccinated mice conferred protection against Shigella, indicating that the model is suitable for testing of vaccine candidates. To protect both Shigella and Salmonella, a chimeric recombinant vaccine (rIpaB-T2544) was developed by fusing IpaB with Salmonella outer membrane protein T2544. Vaccinated mice developed antigen-specific serum IgG and IgA antibodies and a balanced Th1/Th2 response and were protected against oral challenge with Shigella (S. flexneri 2a, Shigella dysenteriae, and Shigella sonnei) using our present mouse model and Salmonella (Salmonella Typhi and Paratyphi) using an iron overload mouse model. We describe here the development of an oral Shigella infection model in wild-type mouse. This model was successfully used to demonstrate the immunogenicity and protective efficacy of a candidate protein subunit vaccine against Shigella.
Collapse
MESH Headings
- Animals
- Dysentery, Bacillary/prevention & control
- Dysentery, Bacillary/immunology
- Dysentery, Bacillary/microbiology
- Dysentery, Bacillary/pathology
- Disease Models, Animal
- Mice
- Shigella Vaccines/immunology
- Shigella Vaccines/administration & dosage
- Mice, Inbred BALB C
- Antibodies, Bacterial/blood
- Shigella flexneri/immunology
- Female
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Bacterial Proteins/immunology
- Bacterial Proteins/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Immunoglobulin G/blood
Collapse
Affiliation(s)
- Risha Haldar
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Prolay Halder
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Shin-ichi Miyoshi
- Division of Medicine, Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Okayama, Japan
| | - Santasabuj Das
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
- Division of Biological Science, ICMR-National Institute of Occupational Health, Ahmedabad, Gujarat, India
| |
Collapse
|
7
|
Desalegn G, Abrahamson C, Ross Turbyfill K, Pill-Pepe L, Bautista L, Tamilselvi CS, Dunn D, Kapoor N, Sullinger B, Herrera M, Oaks EV, Fairman J, Pasetti MF. A broad spectrum Shigella vaccine based on VirG 53-353 multiepitope region produced in a cell-free system. NPJ Vaccines 2025; 10:6. [PMID: 39805874 PMCID: PMC11731012 DOI: 10.1038/s41541-025-01064-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
Dysentery caused by Shigella species remains a major health threat to children in low- and middle-income countries. There is no vaccine available. The most advanced candidates, i.e., O-polysaccharide (OPS)-based conjugates, have limited coverage-only against the immunizing serotype. Vaccines based on Shigella conserved proteins are sought for their simplicity and capacity to prevent disease caused by multiple serotypes. We previously reported the broad protective capacity of VirGα, a conserved surface-exposed domain of Shigella virulence factor. Seeking to refine the vaccine antigenic target and achieve scalable manufacturing compatible with Good Manufacturing Practices, we mapped linear B-cell epitopes spanning the entire VirG protein sequence by probing the immune reactivity of 10-mer peptides (overlapping 4-8 aa) with sera from Shigella-infected rhesus monkeys. The surface-exposed VirG53-353 subregion of the passenger α-domain demonstrated the highest and strongest immunoreactivity. VirG53-353 was produced efficiently at a large scale (>150 mg/L) using cell-free protein synthesis. When administered to mice intramuscularly, VirG53-353 elicited robust antibody responses and conferred high levels of protection against the three most prevalent Shigella serotypes (S. flexneri 2a, 3a, and S. sonnei). VirG53-353 evoked the production of Th2-type cytokines by spleen cells from vaccinated mice. A new universal Shigella vaccine based on VirG53-353 meets the World Health Organization's preferred product specifications. The target antigen refinement and production improvement described here will facilitate the first-in-human studies.
Collapse
Affiliation(s)
- Girmay Desalegn
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - K Ross Turbyfill
- Department of Diarrheal Disease Research, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | | | | | - Chitradevi S Tamilselvi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dylan Dunn
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | - Edwin V Oaks
- Department of Diarrheal Disease Research, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Patuxent Research and Consulting Group, Gambrills, MD, USA
| | | | - Marcela F Pasetti
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
8
|
Bernshtein B, Kelly M, Cizmeci D, Zhiteneva JA, Macvicar R, Kamruzzaman M, Bhuiyan TR, Chowdhury F, Khan AI, Qadri F, Charles RC, Xu P, Kováč P, Clarkson KA, Kaminski RW, Alter G, Ryan ET. Determinants of immune responses predictive of protection against shigellosis in an endemic zone: a systems analysis of antibody profiles and function. THE LANCET. MICROBE 2024; 5:100889. [PMID: 39116906 PMCID: PMC11488819 DOI: 10.1016/s2666-5247(24)00112-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Shigella is the third leading global cause of moderate or severe diarrhoea among children younger than 5 years globally, and is the leading cause in children aged 24-59 months. The mechanism of protection against Shigella infection and disease in endemic areas is uncertain. We aimed to compare the Shigella-specific antibody responses in individuals living in Shigella-endemic and non-endemic areas, and to identify correlates of protection in a Shigella-endemic location. METHODS We applied a systems approach to retrospectively analyse serological responses to Shigella across endemic and non-endemic populations. We profiled serum samples collected from 44 individuals from the USA without previous exposure to Shigella and who were experimentally challenged with Shigella sonnei (non-endemic setting), and serum samples collected from 55 Peruvian army recruits (endemic setting). In the endemic setting, a subset of 37 samples collected from individuals infected with culture-confirmed Shigella flexneri 2a were divided into two groups: susceptible, which included individuals infected within 90 days of entering the camp (n=29); or resistant, which included individuals infected later than 90 days after entering the camp (n=8). We analysed Shigella-specific antibody isotype, subclass, and Fc receptor binding profiles across IpaB, IpaC, IpaD, and lipopolysaccharide from S flexneri 2a, 3a, and 6, and S sonnei, and O-specific polysaccharide (OSP) from S flexneri 2a and 3a and S sonnei. We also evaluated antibody-mediated complement deposition and innate immune cell activation. The main outcome of interest was the detection of antibody markers and functionality associated with protection against shigellosis in a high-burden endemic setting. FINDINGS Adults with endemic exposure to Shigella possessed broad and functional antibody responses across polysaccharide, glycolipid, and protein antigens compared with individuals from non-endemic regions. In a setting with high Shigella burden, elevated levels of OSP-specific Fcα receptor (FcαR) binding antibodies were associated with resistance to shigellosis, whereas total OSP-specific IgA was not, suggesting a potentially unique functionality. OSP-specific FcαR binding IgA found in resistant individuals activated bactericidal neutrophil functions including phagocytosis, degranulation, and production of reactive oxygen species. Moreover, IgA depletion from resistant serum significantly reduced binding of OSP-specific antibodies to FcαR and antibody-mediated activation of neutrophils and monocytes. INTERPRETATION Our findings suggest that OSP-specific functional IgA responses contribute to protective immunity against Shigella infection in a high-burden setting. These findings will assist in the development and evaluation of Shigella vaccines. FUNDING US National Institutes of Health.
Collapse
Affiliation(s)
- Biana Bernshtein
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA.
| | - Meagan Kelly
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Deniz Cizmeci
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Julia A Zhiteneva
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Ryan Macvicar
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Mohammad Kamruzzaman
- International Centre for Diarrheal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh; Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Taufiqur R Bhuiyan
- International Centre for Diarrheal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Fahima Chowdhury
- International Centre for Diarrheal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Ashraful Islam Khan
- International Centre for Diarrheal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Firdausi Qadri
- International Centre for Diarrheal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Richelle C Charles
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Peng Xu
- National Institute of Diabetes and Digestive and Kidney Diseases, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, MD, USA
| | - Pavol Kováč
- National Institute of Diabetes and Digestive and Kidney Diseases, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, MD, USA
| | - Kristen A Clarkson
- Department of Diarrheal Disease Research, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Robert W Kaminski
- Department of Diarrheal Disease Research, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Latham BioPharm Group, Cambridge, MA, USA
| | - Galit Alter
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Edward T Ryan
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
9
|
Boerth EM, Gong J, Roffler B, Hancock Z, Berger L, Song B, Malley SF, MacLennan CA, Zhang F, Malley R, Lu YJ. Evaluation of a Quadrivalent Shigella flexneri Serotype 2a, 3a, 6, and Shigella sonnei O-Specific Polysaccharide and IpaB MAPS Vaccine. Vaccines (Basel) 2024; 12:1091. [PMID: 39460258 PMCID: PMC11510904 DOI: 10.3390/vaccines12101091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/05/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Shigellosis is the leading cause of diarrheal deaths worldwide and is particularly dangerous in children under 5 years of age in low- and middle-income countries. Additionally, the rise in antibiotic resistance has highlighted the need for an effective Shigella vaccine. Previously, we have used the Multiple Antigen-Presenting System (MAPS) technology to generate monovalent and quadrivalent Salmonella MAPS vaccines that induce functional antibodies against Salmonella components. METHODS In this work, we detail the development of several monovalent vaccines using O-specific polysaccharides (OSPs) from four dominant serotypes, S. flexneri 2a, 3a, and 6, and S. sonnei. We tested several rhizavidin (rhavi) fusion proteins and selected a Shigella-specific protein IpaB. Quadrivalent MAPS were made with Rhavi-IpaB protein and tested in rabbits for immunogenicity. RESULTS Individual MAPS vaccines generated robust, functional antibody responses against both IpaB and the individual OSP component. Antibodies to IpaB were effective across Shigella serotypes. We also demonstrate that the OSP antibodies generated are specific to each homologous Shigella O type by performing ELISA and bactericidal assays. We combined the components of each MAPS vaccine to formulate a quadrivalent MAPS vaccine which elicited similar antibody and bactericidal responses compared to their monovalent counterparts. Finally, we show that the quadrivalent MAPS immune sera are functional against several clinical isolates of the serotypes used in the vaccine. CONCLUSIONS This quadrivalent MAPS Shigella vaccine is immunogenicity and warrants further study.
Collapse
Affiliation(s)
- Emily M. Boerth
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joyce Gong
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Becky Roffler
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Zoe Hancock
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lydia Berger
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Boni Song
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sasha F. Malley
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Calman A. MacLennan
- Enteric & Diarrheal Diseases, Global Health, Bill & Melinda Gates Foundation, 500 5th Ave. N, Seattle, WA 98109, USA
| | - Fan Zhang
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Richard Malley
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ying-Jie Lu
- Division of Infectious Diseases, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
10
|
Carreto-Binaghi LE, Sztein MB, Booth JS. Role of cellular effectors in the induction and maintenance of IgA responses leading to protective immunity against enteric bacterial pathogens. Front Immunol 2024; 15:1446072. [PMID: 39324143 PMCID: PMC11422102 DOI: 10.3389/fimmu.2024.1446072] [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: 06/08/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
The mucosal immune system is a critical first line of defense to infectious diseases, as many pathogens enter the body through mucosal surfaces, disrupting the balanced interactions between mucosal cells, secretory molecules, and microbiota in this challenging microenvironment. The mucosal immune system comprises of a complex and integrated network that includes the gut-associated lymphoid tissues (GALT). One of its primary responses to microbes is the secretion of IgA, whose role in the mucosa is vital for preventing pathogen colonization, invasion and spread. The mechanisms involved in these key responses include neutralization of pathogens, immune exclusion, immune modulation, and cross-protection. The generation and maintenance of high affinity IgA responses require a delicate balance of multiple components, including B and T cell interactions, innate cells, the cytokine milieu (e.g., IL-21, IL-10, TGF-β), and other factors essential for intestinal homeostasis, including the gut microbiota. In this review, we will discuss the main cellular components (e.g., T cells, innate lymphoid cells, dendritic cells) in the gut microenvironment as mediators of important effector responses and as critical players in supporting B cells in eliciting and maintaining IgA production, particularly in the context of enteric infections and vaccination in humans. Understanding the mechanisms of humoral and cellular components in protection could guide and accelerate the development of more effective mucosal vaccines and therapeutic interventions to efficiently combat mucosal infections.
Collapse
Affiliation(s)
- Laura E. Carreto-Binaghi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Laboratorio de Inmunobiologia de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Tumor Immunology and Immunotherapy Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Jayaum S. Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
11
|
Bernshtein B, Zhiteneva JA, Janardhanan J, Wagh C, Kelly M, Verma S, Jung W, Basher SR, Amin MA, Mahamud S, Rajib NH, Chowdhury F, Khan AI, Charles RC, Xu P, Kováč P, Chakraborty S, Kaminski RW, Alter G, Bhuiyan TR, Qadri F, Ryan ET. Limited O-specific polysaccharide (OSP)-specific functional antibody responses in young children with Shigella infection in Bangladesh. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611236. [PMID: 39345393 PMCID: PMC11429955 DOI: 10.1101/2024.09.04.611236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Shigellosis is the second leading cause of diarrheal death in children younger than five years of age globally. At present, there is no broadly licensed vaccine against shigella infection. Previous vaccine candidates have failed at providing protection for young children in endemic settings. Improved understanding of correlates of protection against Shigella infection and severe shigellosis in young children living in endemic settings is needed. Here, we applied a functional antibody profiling approach to define Shigella-specific antibody responses in young children versus older individuals with culture-confirmed shigellosis in Bangladesh, a Shigella endemic area. We analyzed Shigella-specific antibody isotypes, FcR binding and antibody-mediated innate immune cell activation in longitudinal serum samples collected at clinical presentation and up to 1 year later. We found that higher initial Shigella O-specific polysaccharide (OSP)-specific and protein-specific IgG and FcγR binding levels correlated with less severe disease regardless of patient age, but that individuals under 5 years of age developed a less prominent class switched, FcR-binding, functional and durable antibody response against both OSP and protein Shigella antigens than older individuals. Focusing on the largest cohort, we found that functional S. flexneri 2a OSP-specific responses were significantly induced only in individuals over age 5 years, and that these responses promoted monocyte phagocytosis and activation. Our findings suggest that in a Shigella endemic region, young children with shigellosis harbor a functional antibody response that fails to maximally activate monocytes; such a response may be important in facilitating subsequent innate cell clearance of Shigella, especially via recruitment and activation of polymorphonuclear cells capable of directly killing Shigella.
Collapse
|
12
|
Sugrue JA, Duffy D. Systems vaccinology studies - achievements and future potential. Microbes Infect 2024; 26:105318. [PMID: 38460935 DOI: 10.1016/j.micinf.2024.105318] [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: 07/02/2023] [Revised: 02/22/2024] [Accepted: 03/01/2024] [Indexed: 03/11/2024]
Abstract
Human immune responses to vaccination are variable both within and between populations. Systems vaccinology, which is the application of multi-omics technologies to vaccine studies, seeks to understand such variation and predict responses to optimise vaccine strategies. Here, we outline new approaches to systems vaccinology, focusing on the incorporation of additional cohorts, endpoints and technologies.
Collapse
Affiliation(s)
- Jamie A Sugrue
- Translational Immunology Unit, Institut Pasteur, Université de Paris Cité, F75015, Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université de Paris Cité, F75015, Paris, France.
| |
Collapse
|
13
|
Hwang W, Wantuch PL, Bernshtein B, Zhiteneva J, Slater D, Vater KH, Sridhar S, Oliver E, Roach DJ, Rao S, Turbett SE, Knoot CJ, Harding CM, Amin MN, Cross AS, LaRocque RC, Rosen DA, Harris JB. Antibody responses in Klebsiella pneumoniae bloodstream infection: a cohort study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.591958. [PMID: 38746292 PMCID: PMC11092611 DOI: 10.1101/2024.05.01.591958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background Klebsiella pneumonia (Kpn) is the fourth leading cause of infection-related deaths globally, yet little is known about human antibody responses to invasive Kpn. In this study, we sought to determine whether the O-specific polysaccharide (OPS) antigen, a vaccine candidate, is immunogenic in humans with Kpn bloodstream infection (BSI). We also sought to define the cross-reactivity of human antibody responses among structurally related Kpn OPS subtypes and to assess the impact of capsule production on OPS-targeted antibody binding and function. Methods We measured plasma antibody responses to OPS (and MrkA, a fimbrial protein) in a cohort of patients with Kpn BSI and compared these with controls, including a cohort of healthy individuals and a cohort of individuals with Enterococcus BSI. We performed flow cytometry to measure the impact of Kpn capsule production on whole cell antibody binding and complement deposition, utilizing patient isolates with variable levels of capsule production and isogenic capsule-deficient strains derived from these isolates. Findings We enrolled 69 patients with Kpn BSI. Common OPS serotypes accounted for 57/69 (83%) of infections. OPS was highly immunogenic in patients with Kpn BSI, and peak OPS-IgG antibody responses in patients were 10 to 30-fold higher than antibody levels detected in healthy controls, depending on the serotype. There was significant cross-reactivity among structurally similar OPS subtypes, including the O1v1/O1v2, O2v1/O2v2 and O3/O3b subtypes. Physiological amounts of capsule produced by both hyperencapsulated and non-hyperencapsulated Kpn significantly inhibited OPS-targeted antibody binding and function. Interpretation OPS was highly immunogenic in patients with Kpn BSI, supporting its potential as a candidate vaccine antigen. The strong cross-reactivity observed between similar OPS subtypes in humans with Kpn BSI suggests that it may not be necessary to include all subtypes in an OPS-based vaccine. However, these observations are tempered by the fact that capsule production, even in non-highly encapsulated strains, has the potential to interfere with OPS antibody binding. This may limit the effectiveness of vaccines that exclusively target OPS. Funding National Institute of Allergy and Infectious Diseases at the National Institutes of Health. Research in Context Evidence before this study: Despite the potential of O-specific polysaccharide (OPS) as a vaccine antigen against Klebsiella pneumoniae (Kpn), the immunogenicity of OPS in humans remains largely unstudied, creating a significant knowledge gap with regard to vaccine development. A search of PubMed for publications up to March 18, 2024, using the terms " Klebsiella pneumoniae " and "O-specific polysaccharide" or "O-antigen" or "lipopolysaccharide" revealed no prior studies addressing OPS antibody responses in humans with Kpn bloodstream infections (BSI). One prior study 1 evaluated antibody response to a single lipopolysaccharide (which contains one subtype of OPS) in humans with invasive Kpn infection; however, in this study OPS typing of the infecting strains and target antigen were not described. Added value of this study: Our investigation into OPS immunogenicity in a human cohort marks a significant advance. Analyzing plasma antibody responses in 69 patients with Kpn BSI, we found OPS to be broadly immunogenic across all the types and subtypes examined, and there was significant cross-reactivity among structurally related OPS antigens. We also demonstrated that Kpn capsule production inhibit OPS antibody binding and the activation of complement on the bacterial surface, even in classical Kpn strains expressing lower levels of capsule.Implications of all the available evidence: While the immunogenicity and broad cross-reactivity of OPS in humans with Kpn BSI suggests it is a promising vaccine candidate, the obstruction of OPS antibody binding and engagement by physiologic levels of Kpn capsule underscores the potential limitations of an exclusively OPS-antigen based vaccine for Kpn. Our study provides insights for the strategic development of vaccines aimed at combating Kpn infections, an important antimicrobial resistant pathogen.
Collapse
|
14
|
Baqar S, Bonavia A, Louis Bourgeois A, Campo JJ, Clifford A, Hanevik K, Hasso-Agopsowicz M, Hausdorff W, Kaminski R, MacLennan CA, Mantis N, Martin LB, Omore R, Pasetti M, Pavlinac P, Phalipon A, Poly F, Porter C, Ramasamy MN, Rogawski McQuade ET, Sztein MB, Walker R. The 2022 Vaccines Against Shigella and Enterotoxigenic Escherichia coli (VASE) Conference: Summary of breakout workshops. Vaccine 2024; 42:1445-1453. [PMID: 38036392 PMCID: PMC10953702 DOI: 10.1016/j.vaccine.2023.11.045] [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: 04/25/2023] [Revised: 08/25/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023]
Abstract
The global public health nonprofit organization PATH hosted the third Vaccines Against Shigella and Enterotoxigenic Escherichia coli (VASE) Conference in Washington, DC, from November 29 to December 1, 2022. This international gathering focused on cutting-edge research related to the development of vaccines against neglected diarrheal pathogens including Shigella, enterotoxigenic Escherichia coli (ETEC), Campylobacter, and non-typhoidal Salmonella. In addition to the conference's plenary content, the agenda featured ten breakout workshops on topics of importance to the enteric vaccine field. This unique aspect of VASE Conferences allows focused groups of attendees to engage in in-depth discussions on subjects of interest to the enteric vaccine development community. In 2022, the workshops covered a range of topics. Two focused on the public health value of enteric vaccines, with one examining how to translate evidence into policy and the other on the value proposition of potential combination vaccines against bacterial enteric pathogens. Two more workshops explored new tools for the development and evaluation of vaccines, with the first on integrating antigen/antibody technologies for mucosal vaccine and immunoprophylactic development, and the second on adjuvants specifically for Shigella vaccines for children in low- and middle-income countries. Another pair of workshops covered the status of vaccines against two emerging enteric pathogens, Campylobacter and invasive non-typhoidal Salmonella. The remaining four workshops examined the assessment of vaccine impact on acute and long-term morbidity. These included discussions on the nature and severity of intestinal inflammation; cellular immunity and immunological memory in ETEC and Shigella infections; clinical and microbiologic endpoints for Shigella vaccine efficacy studies in children; and intricacies of protective immunity to enteric pathogens. This article provides a brief summary of the presentations and discussions at each workshop in order to share these sessions with the broader enteric vaccine field.
Collapse
Affiliation(s)
| | - Aurelio Bonavia
- Bill & Melinda Gates Medical Research Institute, United States
| | | | | | | | - Kurt Hanevik
- University of Bergen, Norway; Norwegian National Advisory Unit for Tropical Infectious Diseases, Medical Department, Haukeland University Hospital, Norway
| | | | - William Hausdorff
- PATH, United States; Faculty of Medicine, Université Libre de Bruxelles, Belgium
| | | | - Calman A MacLennan
- Enteric and Diarrheal Diseases, Bill & Melinda Gates Foundation, United Kingdom; The Jenner Institute, United Kingdom
| | - Nicholas Mantis
- Wadsworth Center, New York State Department of Health, United States
| | | | - Richard Omore
- Kenya Medical Research Institute Center for Global Health Research, Kenya
| | | | | | | | | | - Chad Porter
- Naval Medical Research Command, United States
| | | | | | | | | |
Collapse
|
15
|
Benedicto-Matambo P, Avolio LN, Badji H, Batool R, Khanam F, Munga S, Tapia MD, Peñataro Yori P, Awuor AO, Ceesay BE, Cornick J, Cunliffe NA, Garcia Bardales PF, Heaney CD, Hotwani A, Ireen M, Taufiqul Islam M, Jallow O, Kaminski RW, Shapiama Lopez WV, Maiden V, Ikumapayi UN, Nyirenda R, Ochieng JB, Omore R, Paredes Olortegui M, Pavlinac PB, Pisanic N, Qadri F, Qureshi S, Rahman N, Rogawski McQuade ET, Schiaffino F, Secka O, Sonye C, Sultana S, Timite D, Traore A, Yousafzai MT, Taufiqur Rahman Bhuiyan M, Jahangir Hossain M, Jere KC, Kosek MN, Kotloff KL, Qamar FN, Sow SO, Platts-Mills JA. Exploring Natural Immune Responses to Shigella Exposure Using Multiplex Bead Assays on Dried Blood Spots in High-Burden Countries: Protocol From a Multisite Diarrhea Surveillance Study. Open Forum Infect Dis 2024; 11:S58-S64. [PMID: 38532958 PMCID: PMC10962721 DOI: 10.1093/ofid/ofad650] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024] Open
Abstract
Background Molecular diagnostics on human fecal samples have identified a larger burden of shigellosis than previously appreciated by culture. Evidence of fold changes in immunoglobulin G (IgG) to conserved and type-specific Shigella antigens could be used to validate the molecular assignment of type-specific Shigella as the etiology of acute diarrhea and support polymerase chain reaction (PCR)-based microbiologic end points for vaccine trials. Methods We will test dried blood spots collected at enrollment and 4 weeks later using bead-based immunoassays for IgG to invasion plasmid antigen B and type-specific lipopolysaccharide O-antigen for Shigella flexneri 1b, 2a, 3a, and 6 and Shigella sonnei in Shigella-positive cases and age-, site-, and season-matched test-negative controls from all sites in the Enterics for Global Health (EFGH) Shigella surveillance study. Fold antibody responses will be compared between culture-positive, culture-negative but PCR-attributable, and PCR-positive but not attributable cases and test-negative controls. Age- and site-specific seroprevalence distributions will be identified, and the association between baseline antibodies and Shigella attribution will be estimated. Conclusions The integration of these assays into the EFGH study will help support PCR-based attribution of acute diarrhea to type-specific Shigella, describe the baseline seroprevalence of conserved and type-specific Shigella antibodies, and support correlates of protection for immunity to Shigella diarrhea. These insights can help support the development and evaluation of Shigella vaccine candidates.
Collapse
Affiliation(s)
- Prisca Benedicto-Matambo
- School of Biomedical Sciences and Health Professions, Department of Medical Laboratory Sciences, Kamuzu University of Health Sciences, Blantyre, Malawi
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Department of Clinical Infection, Microbiology and Immunology, Liverpool, UK
| | - Lindsay N Avolio
- Department of Environmental Health & Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Henry Badji
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Rabab Batool
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Farhana Khanam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Stephen Munga
- Kenya Medical Research Institute, Center for Global Health Research (KEMRI-CGHR), Kisumu, Kenya
| | - Milagritos D Tapia
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Pablo Peñataro Yori
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Alex O Awuor
- Kenya Medical Research Institute, Center for Global Health Research (KEMRI-CGHR), Kisumu, Kenya
| | - Bubacarr E Ceesay
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Jennifer Cornick
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Department of Clinical Infection, Microbiology and Immunology, Liverpool, UK
| | - Nigel A Cunliffe
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Department of Clinical Infection, Microbiology and Immunology, Liverpool, UK
| | | | - Christopher D Heaney
- Department of Environmental Health & Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Aneeta Hotwani
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Mahzabeen Ireen
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Md Taufiqul Islam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Ousman Jallow
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | | | | | - Victor Maiden
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
| | - Usman Nurudeen Ikumapayi
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Ruth Nyirenda
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
| | - John Benjamin Ochieng
- Kenya Medical Research Institute, Center for Global Health Research (KEMRI-CGHR), Kisumu, Kenya
| | - Richard Omore
- Kenya Medical Research Institute, Center for Global Health Research (KEMRI-CGHR), Kisumu, Kenya
| | | | - Patricia B Pavlinac
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Nora Pisanic
- Department of Environmental Health & Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Firdausi Qadri
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Sonia Qureshi
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Nazia Rahman
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | | | - Francesca Schiaffino
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
- Faculty of Veterinary Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Ousman Secka
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Catherine Sonye
- Kenya Medical Research Institute, Center for Global Health Research (KEMRI-CGHR), Kisumu, Kenya
| | - Shazia Sultana
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Drissa Timite
- Centre pour le Développement des Vaccins du Mali, Bamako, Mali
| | - Awa Traore
- Centre pour le Développement des Vaccins du Mali, Bamako, Mali
| | | | - Md Taufiqur Rahman Bhuiyan
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - M Jahangir Hossain
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Khuzwayo C Jere
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Department of Clinical Infection, Microbiology and Immunology, Liverpool, UK
- School of Life Sciences & Health Professions, Department of Medical Laboratory Sciences, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Margaret N Kosek
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Karen L Kotloff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Farah Naz Qamar
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Samba O Sow
- Centre pour le Développement des Vaccins du Mali, Bamako, Mali
| | - James A Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
16
|
Venkatesan MM, Barnoy S, Frenck R, McNeal M, Baqar S. B memory cell responses to LPS, IVP and IpaB antigen after oral vaccination with Shigella sonnei vaccine candidates WRSs2 and WRSs3. PLoS One 2024; 19:e0290987. [PMID: 38232106 PMCID: PMC10793881 DOI: 10.1371/journal.pone.0290987] [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: 04/17/2023] [Accepted: 08/20/2023] [Indexed: 01/19/2024] Open
Abstract
B memory (BM) cell responses were evaluated using peripheral blood mononuclear cells that were collected and cryopreserved during a Phase 1 trial of two live Shigella sonnei vaccine candidates WRSs2 and WRSs3. An ELISpot assay was used to measure IgG+ and IgA+ BM cell responses against S. sonnei LPS, IVP and IpaB antigens. Analysis of BM cell responses at baseline, and on days 28 and 56 post vaccination indicate that after a single oral dose of WRSs2 and WRSs3, both groups of vaccinees induced IgG+ and IgA+ BM cell responses that were variable in magnitude among subjects and reached significance to IVP and IpaB at several doses. The responses generally peaked at d28 after vaccination. The baseline as well as post-vaccination levels of IgA+ BM cells were relatively higher than IgG+ BM cells, but the maximum fold-increase at d28/d56 over baseline was greater for IgG+ than IgA+ BM cell responses. Furthermore, at the three highest vaccine doses, >60-90% of subjects were considered responders indicating a ≥2-fold higher IgG+ BM cell responses to IVP and IpaB post vaccination, while fewer subjects indicated the same level of response to LPS.
Collapse
Affiliation(s)
- Malabi M. Venkatesan
- US Army Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Shoshana Barnoy
- US Army Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Robert Frenck
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Monica McNeal
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Shahida Baqar
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
17
|
Desalegn G, Tamilselvi CS, Lemme-Dumit JM, Heine SJ, Dunn D, Ndungo E, Kapoor N, Oaks EV, Fairman J, Pasetti MF. Shigella virulence protein VirG is a broadly protective antigen and vaccine candidate. NPJ Vaccines 2024; 9:2. [PMID: 38167387 PMCID: PMC10761965 DOI: 10.1038/s41541-023-00797-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Diarrhea caused by Shigella has been associated with high morbidity and mortality in young children worldwide. There are no licensed vaccines, and those clinically advanced have restricted coverage as they elicit serotype-specific immunity while disease is caused by multiple circulating serotypes. Our group had previously reported a close association between serum antibodies to the Shigella virulence factor VirG (or IcsA) and clinical protection in infected individuals. VirG is highly conserved among Shigella strains and appealing as a broad-spectrum vaccine candidate. In this study, we investigated the immunogenicity and protective capacity of VirG as a subunit vaccine in mice. The surface-exposed alpha (α) domain of VirG (VirGα) was produced as a recombinant protein. This region has almost identical immune reactivity to full-length VirG. Administered intramuscularly with alum, VirGα elicited robust immune responses and high protective efficacy against S. flexneri 2a and S. sonnei. Almost complete protection was afforded by VirGα given intranasally with the E. coli double mutant heat-labile toxin (dmLT). VirGα-specific antibodies recognized VirG expressed on live Shigella, and blocked Shigella adhesion and invasion to human colonic cells. These results show for the first time that VirGα is a promising cross-protective vaccine candidate to prevent Shigella infection.
Collapse
Affiliation(s)
- Girmay Desalegn
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Chitradevi S Tamilselvi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Jose M Lemme-Dumit
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Shannon J Heine
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Dylan Dunn
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Esther Ndungo
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Neeraj Kapoor
- Vaxcyte, Inc., 825 Industrial Road, San Carlos, CA, 94070, USA
| | - Edwin V Oaks
- Patuxent Research and Consulting Group, 3106 Arrowhead Farm Rd, Gambrills, MD, 21054, USA
| | - Jeff Fairman
- Vaxcyte, Inc., 825 Industrial Road, San Carlos, CA, 94070, USA
| | - Marcela F Pasetti
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
18
|
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
|
19
|
Smith C, Smith E, Chiu C, Hinton J, Perez Sepulveda B, Gordon M, Choy RK, Hill PW, Meiring JE, Darton TC, Carey ME, Cooke G, Gibani MM. The Challenge Non-Typhoidal Salmonella (CHANTS) Consortium: Development of a non-typhoidal Salmonella controlled human infection model: Report from a consultation group workshop, 05 July 2022, London, UK. Wellcome Open Res 2023; 8:111. [PMID: 37808389 PMCID: PMC10558987 DOI: 10.12688/wellcomeopenres.19012.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
Invasive non-typhoidal Salmonella disease (iNTS) is a major cause of morbidity and mortality globally, particularly as a cause of bloodstream infection in children and immunocompromised adults in sub-Saharan Africa. Vaccines to prevent non-typhoidal Salmonella (NTS) would represent a valuable public health tool in this setting to avert cases and prevent expansion of antimicrobial resistance. Several NTS and combination typhoidal-NTS vaccine candidates are in early-stage development, although the pathway to licensure is unclear due to challenges in conducting large phase III field trials. Controlled human infection models (CHIM) present an opportunity to accelerate vaccine development for a range of enteric pathogens. Several recent typhoidal Salmonella CHIMs have been conducted safely and have played pivotal roles in progressing vaccine candidates to pre-qualification and licensure. The Challenge Non-Typhoidal Salmonella (CHANTS) consortium has been formed with funding from the Wellcome Trust, to deliver the first NTS CHIM, which can act as a platform for future vaccine evaluation. This paper reports the conclusions of a consultation group workshop convened with key stakeholders. The aims of this meeting were to: (1) define the rationale for an NTS CHIM (2) map the NTS vaccine pipeline (3) refine study design and (4) establish potential future use cases.
Collapse
Affiliation(s)
- Christopher Smith
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Emma Smith
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Jay Hinton
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
| | - Blanca Perez Sepulveda
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
| | - Melita Gordon
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | | | - Peter W.S. Hill
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - James E. Meiring
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Thomas C. Darton
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Megan E. Carey
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Graham Cooke
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Malick M. Gibani
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - CHANTS Consortium
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- PATH, Seattle, Washington, 98121, USA
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
20
|
Smith C, Smith E, Chiu C, Hinton J, Perez Sepulveda B, Gordon M, Choy RK, Hill PW, Meiring JE, Darton TC, Carey ME, Cooke G, Gibani MM. The Challenge Non-Typhoidal Salmonella (CHANTS) Consortium: Development of a non-typhoidal Salmonella controlled human infection model: Report from a consultation group workshop, 05 July 2022, London, UK. Wellcome Open Res 2023; 8:111. [PMID: 37808389 PMCID: PMC10558987 DOI: 10.12688/wellcomeopenres.19012.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2023] [Indexed: 04/21/2024] Open
Abstract
Invasive non-typhoidal Salmonella disease (iNTS) is a major cause of morbidity and mortality globally, particularly as a cause of bloodstream infection in children and immunocompromised adults in sub-Saharan Africa. Vaccines to prevent non-typhoidal Salmonella (NTS) would represent a valuable public health tool in this setting to avert cases and prevent expansion of antimicrobial resistance. Several NTS and combination typhoidal-NTS vaccine candidates are in early-stage development, although the pathway to licensure is unclear due to challenges in conducting large phase III field trials. Controlled human infection models (CHIM) present an opportunity to accelerate vaccine development for a range of enteric pathogens. Several recent typhoidal Salmonella CHIMs have been conducted safely and have played pivotal roles in progressing vaccine candidates to pre-qualification and licensure. The Challenge Non-Typhoidal Salmonella (CHANTS) consortium has been formed with funding from the Wellcome Trust, to deliver the first NTS CHIM, which can act as a platform for future vaccine evaluation. This paper reports the conclusions of a consultation group workshop convened with key stakeholders. The aims of this meeting were to: (1) define the rationale for an NTS CHIM (2) map the NTS vaccine pipeline (3) refine study design and (4) establish potential future use cases.
Collapse
Affiliation(s)
- Christopher Smith
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Emma Smith
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Jay Hinton
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
| | - Blanca Perez Sepulveda
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
| | - Melita Gordon
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | | | - Peter W.S. Hill
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - James E. Meiring
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Thomas C. Darton
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Megan E. Carey
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Graham Cooke
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Malick M. Gibani
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - CHANTS Consortium
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- PATH, Seattle, Washington, 98121, USA
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
21
|
Boero E, Vezzani G, Micoli F, Pizza M, Rossi O. Functional assays to evaluate antibody-mediated responses against Shigella: a review. Front Cell Infect Microbiol 2023; 13:1171213. [PMID: 37260708 PMCID: PMC10227456 DOI: 10.3389/fcimb.2023.1171213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/27/2023] [Indexed: 06/02/2023] Open
Abstract
Shigella is a major global pathogen and the etiological agent of shigellosis, a diarrheal disease that primarily affects low- and middle-income countries. Shigellosis is characterized by a complex, multistep pathogenesis during which bacteria use multiple invasion proteins to manipulate and invade the intestinal epithelium. Antibodies, especially against the O-antigen and some invasion proteins, play a protective role as titres against specific antigens inversely correlate with disease severity; however, the context of antibody action during pathogenesis remains to be elucidated, especially with Shigella being mostly an intracellular pathogen. In the absence of a correlate of protection, functional assays rebuilding salient moments of Shigella pathogenesis can improve our understanding of the role of protective antibodies in blocking infection and disease. In vitro assays are important tools to build correlates of protection. Only recently animal models to recapitulate human pathogenesis, often not in full, have been established. This review aims to discuss in vitro assays to evaluate the functionality of anti-Shigella antibodies in polyclonal sera in light of the multistep and multifaced Shigella infection process. Indeed, measurement of antibody level alone may limit the evaluation of full vaccine potential. Serum bactericidal assay (SBA), and other functional assays such as opsonophagocytic killing assays (OPKA), and adhesion/invasion inhibition assays (AIA), are instead physiologically relevant and may provide important information regarding the role played by these effector mechanisms in protective immunity. Ultimately, the review aims at providing scientists in the field with new points of view regarding the significance of functional assays of choice which may be more representative of immune-mediated protection mechanisms.
Collapse
Affiliation(s)
- Elena Boero
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Giacomo Vezzani
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Mariagrazia Pizza
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| |
Collapse
|
22
|
Bernshtein B, Kelly M, Cizmeci D, Zhiteneva JA, Macvicar R, Kamruzzaman M, Bhuiyan TR, Chowdhury F, Khan AI, Qadri F, Charles RC, Xu P, Kováč P, Kaminski RW, Alter G, Ryan ET. Shigella O-specific polysaccharide functional IgA responses mediate protection against shigella infection in an endemic high-burden setting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.04.539451. [PMID: 37205407 PMCID: PMC10187263 DOI: 10.1101/2023.05.04.539451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Shigella is the second leading cause of diarrheal disease-related death in young children in low and middle income countries. The mechanism of protection against shigella infection and disease in endemic areas is uncertain. While historically LPS-specific IgG titers have been associated with protection in endemic settings, emerging deeper immune approaches have recently elucidated a protective role for IpaB-specific antibody responses in a controlled human challenge model in North American volunteers. To deeply interrogate potential correlates of immunity in areas endemic for shigellosis, here we applied a systems approach to analyze the serological response to shigella across endemic and non-endemic populations. Additionally, we analyzed shigella-specific antibody responses over time in the context of endemic resistance or breakthrough infections in a high shigella burden location. Individuals with endemic exposure to shigella possessed broad and functional antibody responses across both glycolipid and protein antigens compared to individuals from non-endemic regions. In high shigella burden settings, elevated levels of OSP-specific FcαR binding antibodies were associated with resistance to shigellosis. OSP-specific FcαR binding IgA found in resistant individuals activated bactericidal neutrophil functions including phagocytosis, degranulation and reactive oxygen species production. Moreover, IgA depletion from resistant serum significantly reduced binding of OSP-specific antibodies to FcαR and antibody mediated activation of neutrophils and monocytes. Overall, our findings suggest that OSP-specific functional IgA responses contribute to protective immunity against shigella infection in high-burden settings. These findings will assist in the development and evaluation of shigella vaccines.
Collapse
|
23
|
Portillo S. mSphere of Influence: Pertussis Vaccination and Antibodies in Mothers and Infants. mSphere 2023; 8:e0000923. [PMID: 36728427 PMCID: PMC9942552 DOI: 10.1128/msphere.00009-23] [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: 02/03/2023] Open
Abstract
Susana Portillo works in the field of mother-infant immunity with an emphasis on vaccination and prevention of respiratory diseases. In this mSphere of Influence, she reflects here on how two pertussis vaccine articles made an impact on her research. She discusses how much more remains to be understood about the role of maternal antibodies in preventing or reducing infant illnesses, their capacity to engage other immune components to deliver an efficient antimicrobial response, and their influence on the infant's own response to vaccination. She emphasizes the need for safe and effective interventions that strengthen maternal and infant immunity before and after birth.
Collapse
Affiliation(s)
- Susana Portillo
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|