1
|
Finn A, Guiso N, Wirsing von König CH, Martinón-Torres F, Palmu AA, Bonanni P, Bakhache P, Maltezou HC, Van Damme P. How to improve pertussis vaccination in pregnancy: a European expert review. Expert Rev Vaccines 2025; 24:175-182. [PMID: 40042539 DOI: 10.1080/14760584.2025.2473328] [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: 09/27/2024] [Accepted: 01/27/2025] [Indexed: 03/15/2025]
Abstract
INTRODUCTION Pertussis vaccination in pregnancy is a safe and highly effective strategy to protect young infants against severe pertussis, but cases continue to occur. In November 2023, the authors of this paper met to discuss difficulties faced by pertussis vaccination programs in pregnant women in Europe, and the need and potential for new vaccines. AREAS COVERED We summarize current pertussis epidemiology, the status of pertussis vaccination in pregnancy in Europe, followed by a summary of the meeting on benefits of pertussis-only vaccines and pertussis vaccines with improved immunogenicity, including a review of available vaccines. EXPERT OPINION Ongoing surveillance and registers documenting vaccine uptake in pregnant women are important to monitor changes in pertussis epidemiology and estimated effectiveness of maternal pertussis vaccination programs in individual countries. While current programs have been effective, Tdap or Tdap-IPV combined vaccines are not the ideal choice but are the only vaccines available for pertussis immunization in pregnancy in Europe. Pertussis-only vaccine would avoid exposing women to unnecessary tetanus and diphtheria boosters in every pregnancy. Recombinant pertussis vaccines with higher immunogenicity could prolong passive immune protection against pertussis in young infants.
Collapse
Affiliation(s)
- Adam Finn
- Bristol Vaccine Centre, Schools of Population Health Science and of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | | | | | - Federico Martinón-Torres
- Translational Pediatrics and Infectious Diseases, Hospital Clínico Universitario de Santiago (SERGAS) and University of Santiago de Compostela (USC), Santiago de Compostela, Spain
- Genetics, Vaccines and Pediatric Infectious Diseases Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Arto A Palmu
- FVR - Finnish Vaccine Research, Tampere, Finland
| | - Paolo Bonanni
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Pierre Bakhache
- INFOVAC, French Association of Ambulatory Pediatrics, Toulon, France
| | - Helena C Maltezou
- Directorate of Research, Studies and Documentation, National Public Health Organization, Athens, Greece
| | - Pierre Van Damme
- Centre for the Evaluation of Vaccination, Vaccinopolis, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
2
|
Goldsmith JA, Nguyen AW, Wilen RE, Wijagkanalan W, McLellan JS, Maynard JA. Structural basis for neutralizing antibody binding to pertussis toxin. Proc Natl Acad Sci U S A 2025; 122:e2419457122. [PMID: 40172968 PMCID: PMC12002313 DOI: 10.1073/pnas.2419457122] [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: 09/24/2024] [Accepted: 03/03/2025] [Indexed: 04/04/2025] Open
Abstract
Pertussis toxin (PT) is a key protective antigen in vaccine- and natural immunity-mediated protection from Bordetella pertussis infection. Despite its importance, no PT-neutralizing epitopes have been characterized structurally. To define neutralizing epitopes and identify key structural elements to preserve during PT antigen design, we determined a 3.6 Å cryoelectron microscopy structure of genetically detoxified PT (PTg) bound to hu11E6 and hu1B7, two potently neutralizing anti-PT antibodies with complementary mechanisms: disruption of toxin adhesion to cells and intracellular activities, respectively. Hu11E6 binds the paralogous S2 and S3 subunits of PTg via a conserved epitope but surprisingly did not span the previously identified sialic acid-binding site implicated in toxin adhesion. Hu11E6 specifically prevented PTg binding to sialylated N-glycans and a sialylated model receptor, as demonstrated by high-throughput glycan array analysis and ELISA, while a T cell activation assay showed that it blocks PTg mitogenic activities to define its neutralizing mechanism. Hu1B7 bound a quaternary epitope spanning the S1 and S5 subunits, although functional studies of hu1B7 variants suggested that S5 binding is not involved in its PT neutralization mechanism. These results structurally define neutralizing epitopes on PT, improving our molecular understanding of immune protection from B. pertussis and providing key information for the future development of PT immunogens.
Collapse
Affiliation(s)
- Jory A. Goldsmith
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Annalee W. Nguyen
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX78712
| | - Rebecca E. Wilen
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX78712
| | | | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Jennifer A. Maynard
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX78712
| |
Collapse
|
3
|
Kang HM, Lee TJ, Park SE, Choi SH. Pertussis in the Post-COVID-19 Era: Resurgence, Diagnosis, and Management. Infect Chemother 2025; 57:13-30. [PMID: 40183651 PMCID: PMC11972920 DOI: 10.3947/ic.2024.0117] [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: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 04/05/2025] Open
Abstract
Pertussis is endemic worldwide, with epidemics occurring every 2 to 5 years despite a high vaccination coverage. After limited circulation during the coronavirus disease 2019 (COVID-19) pandemic, pertussis cases have increased rapidly worldwide since mid-late 2023, returning to pre-pandemic patterns. In Korea, 90 cases of pertussis were reported from April 2020 to May 2023, with elderly individuals aged ≥65 years accounting for 48.9%. Pertussis cases have increased sharply since June 2024, showing a nationwide epidemic, with a large increase among adolescents aged 13-15 years. As of August 2024, the national incidence rate of pertussis was estimated to be 37.75 per 100,000 population, with the highest incidence of 526.2 per 100,000 population in 13-year-olds. In Europe, during 2023-2024, an increase in pertussis incidence among infants was observed, along with large increases in 10-19-year-olds. In China, the number of reported cases of pertussis has increased rapidly since late 2023, with an age shift to older children, increase of vaccine escape, and a marked increase in the prevalence of macrolide-resistant Bordetella pertussis. The recent global resurgence of pertussis is due to decreased opportunities for boosting immunity by natural infection during the COVID-19 pandemic in combination with waning of immunity-induced pertussis vaccines.
Collapse
Affiliation(s)
- Hyun Mi Kang
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Taek-Jin Lee
- Department of Pediatrics, CHA University School of Medicine, Pocheon, Korea
| | - Su Eun Park
- Department of Pediatrics, School of Medicine, Pusan National University, Busan, Korea
| | - Soo-Han Choi
- Department of Pediatrics, School of Medicine, Pusan National University, Busan, Korea.
| |
Collapse
|
4
|
Puthanakit T, Tangsathapornpong A, Anugulruengkitt S, Nantanee R, Bunjoungmanee P, Mansouri S, Fortuna L, Wijagkanalan W, Tantawichien T. A reduced-dose recombinant pertussis vaccine booster in Thai adolescents: a phase 2/3, observer-blinded, randomised controlled, non-inferiority trial. THE LANCET. CHILD & ADOLESCENT HEALTH 2024; 8:900-909. [PMID: 39572123 DOI: 10.1016/s2352-4642(24)00173-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND A resurgence of pertussis has increased the demand for low-cost vaccines. The aim of this study was to test the immunogenicity of a booster acellular monovalent pertussis vaccine containing reduced-dose (2 μg) recombinant pertussis toxin (PT) and 5 μg filamentous haemagglutinin (FHA; apgen) against a version of apgen containing tetanus and reduced-dose diphtheria toxoids (Tdapgen) and a licensed vaccine containing chemically detoxified PT and FHA combined with tetanus toxoid and reduced-dose diphtheria toxoid (Tdapchem). METHODS This phase 2/3, observer-blinded, randomised, controlled, non-inferiority trial was done in adolescents aged 9-17 years at two clinical research centres in Bangkok and Pathum Thani, Thailand. Eligible participants were screened and randomly assigned (1:1:1) to receive one booster dose of apgen, Tdapgen, or Tdapchem vaccine. Participants were followed up until day 336 post-immunisation. The primary endpoint was non-inferior seroconversion rates in Tdapgen and Tdapchem vaccine groups, with seroconversion rate defined as the proportion of participants with at least a four-fold increase on day 28 post-immunisation relative to baseline of anti-PT and anti-FHA IgG. The non-inferiority for seroconversion rates of anti-PT and anti-FHA IgG was defined as the lower bound of the two-sided 95% CI of the seroconversion rate for Tdapgen compared with Tdapchem exceeding -10%. Immunogenicity was analysed in the per-protocol population. All safety data were collected, and the prevalence of adverse events was analysed in the intention-to-treat population. This trial was registered on the Thai Clinical Trial Registry (TCTR20181031001). FINDINGS Between June 18, and Aug 3, 2019, 450 adolescents (mean age 12·1 years, SD 2·5) were enrolled and randomly assigned (150 participants in each group). Day 28 anti-PT IgG seroconversion rates were 141 (94%) of 150 participants who received Tdapgen (95% CI 88·8-97·0) and 105 (71%) of 149 participants who received Tdapchem (62·7-77·2; p<0·0001). Day 28 anti-FHA IgG seroconversion rates were 144 (96%) of 150 participants who received Tdapgen (91·4-98·3) and 124 (83%) of 149 participants who received Tdapchem (76·4-88·4; p<0·0001). The difference in seroconversion rates was 23·5% (95% CI 15·3-31·8) for anti-PT IgG and 12·8% (6·0-19·6) for anti-FHA IgG, when comparing the Tdapgen versus the Tdapchem vaccine group. No vaccine-related serious adverse events were reported. INTERPRETATION Recombinant Tdapgen vaccine showed non-inferior immunogenicity compared with Tdapchem at day 28 in terms of seroconversion rate of anti-PT IgG and anti-FHA IgG relative to baseline. The reduced-dose approach for Tdapgen vaccines thus presents as a potentially cost-saving booster strategy to protect adolescents against pertussis. FUNDING Office of National Higher Education Science Research and Innovation Policy Council (Programme Management Unit Competitiveness), Thailand, and BioNet-Asia.
Collapse
Affiliation(s)
- Thanyawee Puthanakit
- Centre of Excellence for Paediatric Infectious Diseases and Vaccines and Department of Paediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Suvaporn Anugulruengkitt
- Centre of Excellence for Paediatric Infectious Diseases and Vaccines and Department of Paediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Rapisa Nantanee
- Centre of Excellence for Paediatric Infectious Diseases and Vaccines and Department of Paediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pornumpa Bunjoungmanee
- Department of Paediatrics, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | | | | | | | - Terapong Tantawichien
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
5
|
Galeas-Pena M, Hirsch A, Kuang E, Hoffmann J, Gellings P, Brown JB, Limbert VM, Callahan CL, McLachlan JB, Morici LA. A novel outer membrane vesicle adjuvant improves vaccine protection against Bordetella pertussis. NPJ Vaccines 2024; 9:190. [PMID: 39406780 PMCID: PMC11480359 DOI: 10.1038/s41541-024-00990-1] [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: 05/09/2023] [Accepted: 10/07/2024] [Indexed: 10/18/2024] Open
Abstract
Pertussis is a vaccine-preventable respiratory disease caused by the Gram negative coccobacillus Bordetella pertussis. The licensed acellular pertussis (aP) vaccines protect against disease but do not prevent bacterial colonization and transmission. Here, we developed and tested an intranasal vaccine composed of aP antigens combined with T-vant, a novel adjuvant derived from bacterial outer membrane vesicles, that elicits both mucosal and systemic immune responses. We hypothesized that immunization of mice with aP-T-vant would enhance mucosal immunity and eliminate B. pertussis in the respiratory tract. In contrast to mice immunized intramuscularly with the licensed aP vaccine, intranasal immunization with aP-T-vant eliminated bacteria in both the lung and nasopharynx. Protection was associated with IFN-gamma and IL-17-producing, non-circulating CD4 + T cells in the lung and nasopharynx, and sterilizing immunity in the nasopharynx was dependent on IL-17. Novel mucosal adjuvants, such as T-vant, warrant further investigation to enhance the efficacy of next generation pertussis vaccines.
Collapse
Affiliation(s)
- Michelle Galeas-Pena
- Tulane University School of Medicine, Dept. of Microbiology and Immunology, New Orleans, LA, 70112, USA
| | - Allyson Hirsch
- Tulane University School of Medicine, Dept. of Microbiology and Immunology, New Orleans, LA, 70112, USA
| | - Erin Kuang
- Tulane University School of Medicine, Dept. of Microbiology and Immunology, New Orleans, LA, 70112, USA
| | - Joseph Hoffmann
- Tulane University School of Medicine, Dept. of Microbiology and Immunology, New Orleans, LA, 70112, USA
| | - Patrick Gellings
- Tulane University School of Medicine, Dept. of Microbiology and Immunology, New Orleans, LA, 70112, USA
| | - Jasmine B Brown
- Tulane University School of Medicine, Dept. of Microbiology and Immunology, New Orleans, LA, 70112, USA
| | - Vanessa M Limbert
- Tulane University School of Medicine, Dept. of Microbiology and Immunology, New Orleans, LA, 70112, USA
| | - Claire L Callahan
- Tulane University School of Medicine, Dept. of Microbiology and Immunology, New Orleans, LA, 70112, USA
| | - James B McLachlan
- Tulane University School of Medicine, Dept. of Microbiology and Immunology, New Orleans, LA, 70112, USA.
| | - Lisa A Morici
- Tulane University School of Medicine, Dept. of Microbiology and Immunology, New Orleans, LA, 70112, USA.
| |
Collapse
|
6
|
Goldsmith JA, Nguyen AW, Wilen RE, Wijagkanalan W, McLellan JS, Maynard JA. Structural Basis for Antibody Neutralization of Pertussis Toxin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614357. [PMID: 39386618 PMCID: PMC11463501 DOI: 10.1101/2024.09.23.614357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Pertussis toxin (PT) is a key protective antigen in vaccine- and natural immunity-mediated protection from Bordetella pertussis infection. Despite its importance, no PT-neutralizing epitopes have been characterized structurally. To define neutralizing epitopes and identify key structural elements to preserve during PT antigen design, we determined a 3.6 Å cryo-electron microscopy structure of genetically detoxified PT (PTg) bound to hu11E6 and hu1B7, two potently neutralizing anti-PT antibodies with complementary mechanisms: disruption of toxin adhesion to cells and intracellular activities, respectively. Hu11E6 bound the paralogous S2 and S3 subunits of PTg via a conserved epitope, but surprisingly did not span the sialic acid binding site implicated in toxin adhesion. High-throughput glycan array analysis showed that hu11E6 specifically prevents PTg binding to sialylated N-glycans, while a T cell activation assay showed that hu11E6 blocks PTg mitogenic activities to define the neutralizing mechanism. Hu1B7 bound a quaternary epitope spanning the S1 and S5 subunits, although functional studies of hu1B7 variants suggested that S5 binding is not involved in its PT neutralization mechanism. These results are the first to structurally define neutralizing epitopes on PT, improving our molecular understanding of immune protection from B. pertussis and providing key information for the future development of PT immunogens.
Collapse
Affiliation(s)
- Jory A Goldsmith
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA 78712
| | - Annalee W Nguyen
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas, USA 78712
| | - Rebecca E Wilen
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas, USA 78712
| | | | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA 78712
| | - Jennifer A Maynard
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas, USA 78712
| |
Collapse
|
7
|
Pyles GM, Huckaby AB, Gutierrez MDLP, Witt WT, Mateu-Borrás M, Dublin SR, Rocuskie-Marker C, Sesti BN, Peasak K, Bitzer GJ, Rader N, Weaver KL, Boehm DT, Fitzgerald N, Chapman J, Ulicny S, Damron FH, Barbier M. Virus-like particles displaying the mature C-terminal domain of filamentous hemagglutinin are immunogenic and protective against Bordetella pertussis respiratory infection in mice. Infect Immun 2024; 92:e0027024. [PMID: 39023271 PMCID: PMC11320929 DOI: 10.1128/iai.00270-24] [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: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/20/2024] Open
Abstract
Bordetella pertussis, the bacterium responsible for whooping cough, remains a significant public health challenge despite the existing licensed pertussis vaccines. Current acellular pertussis vaccines, though having favorable reactogenicity and efficacy profiles, involve complex and costly production processes. In addition, acellular vaccines have functional challenges such as short-lasting duration of immunity and limited antigen coverage. Filamentous hemagglutinin (FHA) is an adhesin of B. pertussis that is included in all multivalent pertussis vaccine formulations. Antibodies to FHA have been shown to prevent bacterial attachment to respiratory epithelial cells, and T cell responses to FHA facilitate cell-mediated immunity. In this study, FHA's mature C-terminal domain (MCD) was evaluated as a novel vaccine antigen. MCD was conjugated to virus-like particles via SpyTag-SpyCatcher technology. Prime-boost vaccine studies were performed in mice to characterize immunogenicity and protection against the intranasal B. pertussis challenge. MCD-SpyVLP was more immunogenic than SpyTag-MCD antigen alone, and in Tohama I strain challenge studies, improved protection against challenge was observed in the lungs at day 3 and in the trachea and nasal wash at day 7 post-challenge. Furthermore, a B. pertussis strain encoding genetically inactivated pertussis toxin was used to evaluate MCD-SpyVLP vaccine immunity. Mice vaccinated with MCD-SpyVLP had significantly lower respiratory bacterial burden at both days 3 and 7 post-challenge compared to mock-vaccinated animals. Overall, these data support the use of SpyTag-SpyCatcher VLPs as a platform for use in vaccine development against B. pertussis and other pathogens.
Collapse
Affiliation(s)
- Gage M. Pyles
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Annalisa B. Huckaby
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Maria de la Paz Gutierrez
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - William T. Witt
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Margalida Mateu-Borrás
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Spencer R. Dublin
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Carleena Rocuskie-Marker
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Bethany N. Sesti
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Kerrington Peasak
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Graham J. Bitzer
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Nathaniel Rader
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Kelly L. Weaver
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Dylan T. Boehm
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Nicholas Fitzgerald
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Joshua Chapman
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Samuel Ulicny
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - F. Heath Damron
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Mariette Barbier
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| |
Collapse
|
8
|
Bitzer GJ, Fitzgerald NA, DeJong MA, Cunningham C, Chapman JA, Boehm DT, Pyles GM, Huckaby AB, Miller SJ, Dublin SR, Warden MD, Barbier M, Damron FH. Immunization with an mRNA DTP vaccine protects against pertussis in rats. Infect Immun 2024; 92:e0052023. [PMID: 39016553 PMCID: PMC11320933 DOI: 10.1128/iai.00520-23] [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] [Received: 12/13/2023] [Accepted: 06/28/2024] [Indexed: 07/18/2024] Open
Abstract
Bordetella pertussis is a Gram-negative bacterium that is the causative agent of the respiratory disease known as pertussis. Since the switch to the acellular vaccines of DTaP and Tap, pertussis cases in the US have risen and cyclically fallen. We have observed that mRNA pertussis vaccines are immunogenic and protective in mice. Here, we further evaluated the pertussis toxoid mRNA antigen and refined the formulation based on optimal pertussis toxin neutralization in vivo. We next evaluated the mRNA pertussis vaccine in Sprague-Dawley rats using an aerosol B. pertussis challenge model paired with whole-body plethysmography to monitor coughing and respiratory function. Female Sprague-Dawley rats were primed and boosted with either commercially available vaccines (DTaP or wP-DTP), an mRNA-DTP vaccine, or mock-vaccinated. The mRNA-DTP vaccine was immunogenic in rats and induced antigen-specific IgG antibodies comparable to DTaP. Rats were then aerosol challenged with a streptomycin-resistant emerging clinical isolate D420Sm1. Bacterial burden was assessed at days 1 and 9 post-challenge, and the mRNA vaccine reduced burden equal to both DTaP and wP-DTP. Whole-body plethysmography revealed that mRNA-DTP vaccinated rats were well protected against coughing which was comparable to the non-challenged group. These data suggest that an mRNA-DTP vaccine is immunogenic in rats and provides protection against aerosolized B. pertussis challenge in Sprague-Dawley rats.
Collapse
Affiliation(s)
- Graham J. Bitzer
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Nicholas A. Fitzgerald
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Megan A. DeJong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Casey Cunningham
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Joshua A. Chapman
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Dylan T. Boehm
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Gage M. Pyles
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Annalisa B. Huckaby
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Sarah J. Miller
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Spencer R. Dublin
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Matthew D. Warden
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - F. Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- West Virginia University Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
9
|
Wolf MA, O'Hara JM, Bitzer GJ, Narayanan E, Boehm DT, Bevere JR, DeJong MA, Hall JM, Wong TY, Falcone S, Deal CE, Richards A, Green S, Nguyen B, King E, Ogega C, Russo L, Sen-Kilic E, Plante O, Himansu S, Barbier M, Carfi A, Damron FH. Multivalent mRNA-DTP vaccines are immunogenic and provide protection from Bordetella pertussis challenge in mice. NPJ Vaccines 2024; 9:103. [PMID: 38858423 PMCID: PMC11164898 DOI: 10.1038/s41541-024-00890-4] [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: 06/12/2023] [Accepted: 05/13/2024] [Indexed: 06/12/2024] Open
Abstract
Acellular multivalent vaccines for pertussis (DTaP and Tdap) prevent symptomatic disease and infant mortality, but immunity to Bordetella pertussis infection wanes significantly over time resulting in cyclic epidemics of pertussis. The messenger RNA (mRNA) vaccine platform provides an opportunity to address complex bacterial infections with an adaptable approach providing Th1-biased responses. In this study, immunogenicity and challenge models were used to evaluate the mRNA platform with multivalent vaccine formulations targeting both B. pertussis antigens and diphtheria and tetanus toxoids. Immunization with mRNA formulations were immunogenetic, induced antigen specific antibodies, as well as Th1 T cell responses. Upon challenge with either historical or contemporary B. pertussis strains, 6 and 10 valent mRNA DTP vaccine provided protection equal to that of 1/20th human doses of either DTaP or whole cell pertussis vaccines. mRNA DTP immunized mice were also protected from pertussis toxin challenge as measured by prevention of lymphocytosis and leukocytosis. Collectively these pre-clinical mouse studies illustrate the potential of the mRNA platform for multivalent bacterial pathogen vaccines.
Collapse
Affiliation(s)
- M Allison Wolf
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | | | - Graham J Bitzer
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | | | - Dylan T Boehm
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Justin R Bevere
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Megan A DeJong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Jesse M Hall
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Ting Y Wong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | | | | | | | | | | | | | | | | | - Emel Sen-Kilic
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | | | | | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | | | - F Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA.
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA.
| |
Collapse
|
10
|
Taton M, Willems F, Widomski C, Georges D, Martin C, Jiang Y, Renard K, Konopnicki D, Cogan A, Necsoi C, Matagne A, De Wit S, Ackerman ME, Marchant A, Dauby N. HIV-related immune activation attenuates polyfunctional IgG and memory B-cell responses to Tdap immunization during pregnancy. EBioMedicine 2024; 104:105179. [PMID: 38848615 PMCID: PMC11192781 DOI: 10.1016/j.ebiom.2024.105179] [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: 04/14/2023] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Maternal pertussis vaccination with Tdap vaccine is recommended to protect newborns from severe postnatal infection. HIV-exposed uninfected (HEU) infants have a higher incidence of pertussis infection and may particularly benefit from maternal immunization. The impact of HIV infection on the quality of IgG and memory B cell (MBC) responses to Tdap vaccination in pregnant women (PW) living with HIV (PWH) is unknown. METHODS In this observational study, humoral immune responses to Tdap vaccination, including IgG levels, Fc-dependent effector functions, and MBC frequencies, were measured before and after vaccination in 40 PWH and 42 HIV-uninfected PW. Placental transfer of IgG and avidity were assessed in cord blood (CB). Soluble and cellular immune activation markers were quantified at baseline. FINDINGS One month after vaccination, PWH had lower frequencies of MBC compared with HIV-uninfected PW. At delivery, PWH had attenuated pertussis-specific IgG levels and Fc-dependent effector functions. Reduced levels of maternal vaccine polyfunctional IgG and IgG avidity were transferred to HEU as compared to HIV-unexposed newborns. After adjustment with ethnicity, maternal antibody levels and gestational age at vaccination, HIV infection was independently associated with decreased levels of PT specific-IgG in CB. Both maternal and neonatal pertussis-specific IgG responses as well as PT-specific IgG avidity were inversely correlated with maternal sCD14 levels before vaccination among PWH. INTERPRETATION Maternal HIV infection is associated with attenuated humoral immune responses to Tdap vaccination that correlate with sCD14. Suboptimal transfer of maternal immunity may further increase the risk of severe pertussis infection in HEU infants. FUNDING This work was supported by IRIS Fund managed by the Foundation Roi Baudouin [2017J1820690206902], Association Vésale pour la Recherche Médicale and the Medical Council of CHU Saint-Pierre and has been funded in part with Federal funds from the National Institute of Allergy and Infectious Diseases, National Institutes of Health, US Department of Health and Human Services, under Award No. U19AI145825. N.D. is a clinical researcher and A.M. is Research Director at the Fonds de la Recherche Scientifique (F.R.S.-FNRS), Belgium. M.E.A. was partially supported by NIHNIAID1U19AI14825. This article is published with the support of the Fondation Universitaire of Belgium.
Collapse
Affiliation(s)
- Martin Taton
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Fabienne Willems
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Cyprien Widomski
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles (ULB), , Brussels, Belgium
| | - Daphnée Georges
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles (ULB), , Brussels, Belgium; Faculty of Sciences, Université de Liège, Liège, Belgium
| | - Charlotte Martin
- Department of Infectious Diseases, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Yiwei Jiang
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles (ULB), , Brussels, Belgium
| | - Katty Renard
- Clinical Research Unit, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Deborah Konopnicki
- Department of Infectious Diseases, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alexandra Cogan
- Department of Gynecology and Obstetrics, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Coca Necsoi
- Department of Infectious Diseases, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - André Matagne
- Faculty of Sciences, Université de Liège, Liège, Belgium
| | - Stéphane De Wit
- Department of Infectious Diseases, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA; Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Arnaud Marchant
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles (ULB), , Brussels, Belgium
| | - Nicolas Dauby
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium; Department of Infectious Diseases, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
11
|
DeJong MA, Wolf MA, Bitzer GJ, Hall JM, Fitzgerald NA, Pyles GM, Huckaby AB, Petty JE, Lee K, Barbier M, Bevere JR, Ernst RK, Damron FH. BECC438b TLR4 agonist supports unique immune response profiles from nasal and muscular DTaP pertussis vaccines in murine challenge models. Infect Immun 2024; 92:e0022323. [PMID: 38323817 PMCID: PMC10929442 DOI: 10.1128/iai.00223-23] [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] [Received: 06/05/2023] [Accepted: 12/08/2023] [Indexed: 02/08/2024] Open
Abstract
The protection afforded by acellular pertussis vaccines wanes over time, and there is a need to develop improved vaccine formulations. Options to improve the vaccines involve the utilization of different adjuvants and administration via different routes. While intramuscular (IM) vaccination provides a robust systemic immune response, intranasal (IN) vaccination theoretically induces a localized immune response within the nasal cavity. In the case of a Bordetella pertussis infection, IN vaccination results in an immune response that is similar to natural infection, which provides the longest duration of protection. Current acellular formulations utilize an alum adjuvant, and antibody levels wane over time. To overcome the current limitations with the acellular vaccine, we incorporated a novel TLR4 agonist, BECC438b, into both IM and IN acellular formulations to determine its ability to protect against infection in a murine airway challenge model. Following immunization and challenge, we observed that DTaP + BECC438b reduced bacterial burden within the lung and trachea for both administration routes when compared with mock-vaccinated and challenged (MVC) mice. Interestingly, IN administration of DTaP + BECC438b induced a Th1-polarized immune response, while IM vaccination polarized toward a Th2 immune response. RNA sequencing analysis of the lung demonstrated that DTaP + BECC438b activates biological pathways similar to natural infection. Additionally, IN administration of DTaP + BECC438b activated the expression of genes involved in a multitude of pathways associated with the immune system. Overall, these data suggest that BECC438b adjuvant and the IN vaccination route can impact efficacy and responses of pertussis vaccines in pre-clinical mouse models.
Collapse
Affiliation(s)
- Megan A. DeJong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - M. Allison Wolf
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Graham J. Bitzer
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Jesse M. Hall
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Nicholas A. Fitzgerald
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Gage M. Pyles
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Annalisa B. Huckaby
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Jonathan E. Petty
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Katherine Lee
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Justin R. Bevere
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, Maryland, USA
| | - F. Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| |
Collapse
|
12
|
Kapil P, Wang Y, Zimmerman L, Gaykema M, Merkel TJ. Repeated Bordetella pertussis Infections Are Required to Reprogram Acellular Pertussis Vaccine-Primed Host Responses in the Baboon Model. J Infect Dis 2024; 229:376-383. [PMID: 37565807 PMCID: PMC10873172 DOI: 10.1093/infdis/jiad332] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/31/2023] [Accepted: 08/09/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND The United States has experienced a resurgence of pertussis following the introduction of acellular pertussis (aP) vaccines. This is likely due to the failure of aP vaccines to induce durable immunity and prevent infection, carriage, and transmission. METHODS To evaluate the impact of aP vaccination on the immune response to infection and test the ability of infection to reprogram aP-imprinted immune responses, we challenged unvaccinated and aP-vaccinated baboons with Bordetella pertussis multiple times and accessed the immune responses and outcomes of infections after each exposure. RESULTS Multiple infections were required to elicit T-helper 17 responses and protection in aP-vaccinated animals comparable to responses seen in unvaccinated animals after a single challenge. Even after 3 challenges, T-helper 1 responses were not observed in aP-vaccinated animals. Immunoglobulin G responses to vaccine and nonvaccine antigens were not negatively affected in aP-vaccinated animals. CONCLUSIONS Our results indicate that it is possible to retrain aP-primed immune responses, but it will likely require an optimal booster and multiple doses. Our results in the baboon model suggest that circulation of B. pertussis in aP-vaccinated populations is concentrated in the younger age bands of the population, providing information that can guide improved modeling of B. pertussis epidemiology in aP-vaccinated populations.
Collapse
Affiliation(s)
- Parul Kapil
- Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yihui Wang
- Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lindsey Zimmerman
- Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mara Gaykema
- Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Tod J Merkel
- Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
13
|
Dubois V, Chatagnon J, Depessemier M, Locht C. Maternal acellular pertussis vaccination in mice impairs cellular immunity to Bordetella pertussis infection in offspring. JCI Insight 2023; 8:e167210. [PMID: 37581930 PMCID: PMC10561720 DOI: 10.1172/jci.insight.167210] [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: 11/15/2022] [Accepted: 08/08/2023] [Indexed: 08/17/2023] Open
Abstract
Given the resurgence of pertussis, several countries have introduced maternal tetanus, diphtheria, and acellular pertussis (aP) vaccination during pregnancy to protect young infants against severe pertussis. Although protective against the disease, the effect of maternal aP vaccination on bacterial colonization of the offspring is unknown. Here, we used a mouse model to demonstrate that maternal aP immunization, either before or during pregnancy, protects pups from lung colonization by Bordetella pertussis. However, maternal aP vaccination resulted in significantly prolonged nasal carriage of B. pertussis by inhibiting the natural recruitment of IL-17-producing resident memory T cells and ensuing neutrophil influx in the nasal tissue, especially of those with proinflammatory and cytotoxic properties. Prolonged nasal carriage after aP vaccination is due to IL-4 signaling, as prolonged nasal carriage is abolished in IL-4Rα-/- mice. The effect of maternal aP vaccination can be transferred transplacentally to the offspring or via breastfeeding and is long-lasting, as it persists into adulthood. Maternal aP vaccination may, thus, augment the B. pertussis reservoir.
Collapse
|
14
|
Taton M, Willems F, Widomski C, Martin C, Jiang Y, Renard K, Cogan A, Necsoi C, Ackerman ME, Marchant A, Dauby N. Impact of pregnancy on polyfunctional IgG and memory B cell responses to Tdap immunization. Vaccine 2023; 41:4009-4018. [PMID: 37244810 DOI: 10.1016/j.vaccine.2023.05.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 04/21/2023] [Accepted: 05/14/2023] [Indexed: 05/29/2023]
Abstract
BACKGROUND Maternal pertussis immunization using Tdap vaccine is recommended in many countries to protect newborns from severe post-natal infection. Immunological changes during pregnancy may influence the response to vaccines. The quality of IgG and memory B cell responses to Tdap immunization in pregnant women has not yet been described. METHODS The impact of pregnancy on the response to Tdap vaccination was assessed by comparing humoral immune responses in 42 pregnant and 39 non-pregnant women. The levels of serum pertussis antigens and tetanus toxoid-specific IgG, IgG subclasses, IgG Fc-mediated effector functions, as well as memory B cell frequencies were assessed before and at several time points after vaccination. RESULTS Tdap immunization induced similar levels of pertussis and tetanus-specific IgG and IgG subclasses in pregnant and non-pregnant women. Pregnant women produced IgG promoting complement deposition, and neutrophils and macrophages phagocytosis at levels comparable to non-pregnant women. They were also able to expand pertussis and tetanus-specific memory B cells at similar frequencies as non-pregnant women, suggesting equivalent "boostability". Higher levels of vaccine-specific IgG, IgG subclasses, and IgG Fc-mediated effector functions were detected in cord blood as compared to maternal blood, indicating efficient transport across the placenta. CONCLUSIONS This study demonstrates that pregnancy does not affect the quality of effector IgG and memory B cell responses to Tdap immunization and that polyfunctional IgG are efficiently transferred across the placenta. REGISTRY'S URL AND THE TRIAL'S REGISTRATION NUMBER ClinicalTrials.Gov (NCT03519373).
Collapse
Affiliation(s)
- Martin Taton
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Faculty of Medicine, Université libre de Bruxelles (ULB), Lennik Rd. 808, Anderlecht 1070, Brussels, Belgium.
| | - Fabienne Willems
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Faculty of Medicine, Université libre de Bruxelles (ULB), Lennik Rd. 808, Anderlecht 1070, Brussels, Belgium.
| | - Cyprien Widomski
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Faculty of Medicine, Université libre de Bruxelles (ULB), Lennik Rd. 808, Anderlecht 1070, Brussels, Belgium.
| | - Charlotte Martin
- Department of Infectious Diseases, CHU Saint-Pierre, Université libre de Bruxelles (ULB), Rue Haute 322, Brussels 1000, Brussels, Belgium.
| | - Yiwei Jiang
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Faculty of Medicine, Université libre de Bruxelles (ULB), Lennik Rd. 808, Anderlecht 1070, Brussels, Belgium.
| | - Katty Renard
- Clinical Research Unit, CHU Saint-Pierre, Université libre de Bruxelles (ULB), Rue Haute 322, Brussels 1000, Brussels, Belgium.
| | - Alexandra Cogan
- Department of Gynecology and Obstetrics, CHU Saint-Pierre, Université libre de Bruxelles (ULB), Rue Haute 322, Brussels 1000, Brussels, Belgium.
| | - Coca Necsoi
- Department of Infectious Diseases, CHU Saint-Pierre, Université libre de Bruxelles (ULB), Rue Haute 322, Brussels 1000, Brussels, Belgium.
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Rope Ferry Rd. 1, Hanover, NH 03755, USA; Thayer School of Engineering, Dartmouth College, Thayer Dr. 15, Hanover, NH 03755, USA.
| | - Arnaud Marchant
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Faculty of Medicine, Université libre de Bruxelles (ULB), Lennik Rd. 808, Anderlecht 1070, Brussels, Belgium.
| | - Nicolas Dauby
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Faculty of Medicine, Université libre de Bruxelles (ULB), Lennik Rd. 808, Anderlecht 1070, Brussels, Belgium; Department of Infectious Diseases, CHU Saint-Pierre, Université libre de Bruxelles (ULB), Rue Haute 322, Brussels 1000, Brussels, Belgium.
| |
Collapse
|
15
|
Keech C, Miller VE, Rizzardi B, Hoyle C, Pryor MJ, Ferrand J, Solovay K, Thalen M, Noviello S, Goldstein P, Gorringe A, Cavell B, He Q, Barkoff AM, Rubin K, Locht C. Immunogenicity and safety of BPZE1, an intranasal live attenuated pertussis vaccine, versus tetanus-diphtheria-acellular pertussis vaccine: a randomised, double-blind, phase 2b trial. Lancet 2023; 401:843-855. [PMID: 36906345 DOI: 10.1016/s0140-6736(22)02644-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/08/2022] [Accepted: 12/19/2022] [Indexed: 03/11/2023]
Abstract
BACKGROUND Bordetella pertussis epidemics persist as transmission remains unabated despite high acellular pertussis vaccination rates. BPZE1, a live attenuated intranasal pertussis vaccine, was designed to prevent B pertussis infection and disease. We aimed to assess the immunogenicity and safety of BPZE1 compared with the tetanus-diphtheria-acellular pertussis vaccine (Tdap). METHODS In this double-blind, phase 2b trial at three research centres in the USA, healthy adults aged 18-50 years were randomly assigned (2:2:1:1) via a permuted block randomisation schedule to receive BPZE1 vaccination followed by BPZE1 attenuated challenge, BPZE1 vaccination followed by placebo challenge, Tdap followed by BPZE1 attenuated challenge, or Tdap followed by placebo challenge. On day 1, lyophilised BPZE1 was reconstituted with sterile water and given intranasally (0·4 mL delivered to each nostril), whereas Tdap was given intramuscularly. To maintain masking, participants in the BPZE1 groups received an intramuscular saline injection, and those in the Tdap groups received intranasal lyophilised placebo buffer. The attenuated challenge took place on day 85. The primary immunogenicity endpoint was the proportion of participants achieving nasal secretory IgA seroconversion against at least one B pertussis antigen on day 29 or day 113. Reactogenicity was assessed up to 7 days after vaccination and challenge, and adverse events were recorded for 28 days after vaccination and challenge. Serious adverse events were monitored throughout the study. This trial is registered with ClinicalTrials.gov, NCT03942406. FINDINGS Between June 17 and Oct 3, 2019, 458 participants were screened and 280 were randomly assigned to the main cohort: 92 to the BPZE1-BPZE1 group, 92 to the BPZE1-placebo group, 46 to the Tdap-BPZE1 group, and 50 to the Tdap-placebo group. Seroconversion of at least one B pertussis-specific nasal secretory IgA was recorded in 79 (94% [95% CI 87-98]) of 84 participants in the BPZE1-BPZE1 group, 89 (95% [88-98]) of 94 in the BPZE1-placebo group, 38 (90% [77-97]) of 42 in the Tdap-BPZE1 group, and 42 (93% [82-99]) of 45 in the Tdap-placebo group. BPZE1 induced broad and consistent B pertussis-specific mucosal secretory IgA responses, whereas Tdap did not induce consistent mucosal secretory IgA responses. Both vaccines were well tolerated, with mild reactogenicity and no serious adverse events related to study vaccination. INTERPRETATION BPZE1 induced nasal mucosal immunity and produced functional serum responses. BPZE1 has the potential to avert B pertussis infections, which ultimately could lead to reduced transmission and diminished epidemic cycles. These results should be confirmed in large phase 3 trials. FUNDING ILiAD Biotechnologies.
Collapse
Affiliation(s)
| | - Vicki E Miller
- DM Clinical Research, Texas Center for Drug Development, Houston, TX, USA
| | | | | | | | | | | | - Marcel Thalen
- ILiAD Biotechnologies, Weston, FL, USA; BioLyo Technologies, Ghent, Belgium
| | | | | | - Andrew Gorringe
- United Kingdom Health Security Agency, Porton Down, Salisbury, UK
| | - Breeze Cavell
- United Kingdom Health Security Agency, Porton Down, Salisbury, UK
| | - Qiushui He
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | | | - Camille Locht
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
16
|
Gregg KA, Wang Y, Warfel J, Schoenfeld E, Jankowska E, Cipollo JF, Mayho M, Boinett C, Prasad D, Brickman TJ, Armstrong SK, Parkhill J, Da Silva Antunes R, Sette A, Papin JF, Wolf R, Merkel TJ. Antigen Discovery for Next-Generation Pertussis Vaccines Using Immunoproteomics and Transposon-Directed Insertion Sequencing. J Infect Dis 2023; 227:583-591. [PMID: 36575950 PMCID: PMC10169431 DOI: 10.1093/infdis/jiac502] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Despite high vaccination rates, the United States has experienced a resurgence in reported cases of pertussis after switching to the acellular pertussis vaccine, indicating a need for improved vaccines that enhance infection control. METHODS Bordetella pertussis antigens recognized by convalescent-baboon serum and nasopharyngeal wash were identified by immunoproteomics and their subcellular localization predicted. Genes essential or important for persistence in the baboon airway were identified by transposon-directed insertion-site sequencing (TraDIS) analysis. RESULTS In total, 314 B. pertussis antigens were identified by convalescent baboon serum and 748 by nasopharyngeal wash. Thirteen antigens were identified as immunogenic in baboons, essential for persistence in the airway by TraDIS, and membrane-localized: BP0840 (OmpP), Pal, OmpA2, BP1485, BamA, Pcp, MlaA, YfgL, BP2197, BP1569, MlaD, ComL, and BP0183. CONCLUSIONS The B. pertussis antigens identified as immunogenic, essential for persistence in the airway, and membrane-localized warrant further investigation for inclusion in vaccines designed to reduce or prevent carriage of bacteria in the airway of vaccinated individuals.
Collapse
Affiliation(s)
- Kelsey A Gregg
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yihui Wang
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jason Warfel
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Elizabeth Schoenfeld
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ewa Jankowska
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - John F Cipollo
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | | | - Deepika Prasad
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Timothy J Brickman
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sandra K Armstrong
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, California, USA.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - James F Papin
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Roman Wolf
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Tod J Merkel
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
17
|
Sedney CJ, Caulfield A, Dewan KK, Blas-Machado U, Callender M, Manley NR, Harvill ET. Novel murine model reveals an early role for pertussis toxin in disrupting neonatal immunity to Bordetella pertussis. Front Immunol 2023; 14:1125794. [PMID: 36855631 PMCID: PMC9968397 DOI: 10.3389/fimmu.2023.1125794] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
The increased susceptibility of neonates to specific pathogens has previously been attributed to an underdeveloped immune system. More recent data suggest neonates have effective protection against most pathogens but are particularly susceptible to those that target immune functions specific to neonates. Bordetella pertussis (Bp), the causative agent of "whooping cough", causes more serious disease in infants attributed to its production of pertussis toxin (PTx), although the neonate-specific immune functions it targets remain unknown. Problematically, the rapid development of adult immunity in mice has confounded our ability to study interactions of the neonatal immune system and its components, such as virtual memory T cells which are prominent prior to the maturation of the thymus. Here, we examine the rapid change in susceptibility of young mice and define a period from five- to eight-days-old during which mice are much more susceptible to Bp than mice even a couple days older. These more narrowly defined "neonatal" mice display significantly increased susceptibility to wild type Bp but very rapidly and effectively respond to and control Bp lacking PTx, more rapidly even than adult mice. Thus, PTx efficiently blocks some very effective form(s) of neonatal protective immunity, potentially providing a tool to better understand the neonatal immune system. The rapid clearance of the PTx mutant correlates with the early accumulation of neutrophils and T cells and suggests a role for PTx in disrupting their accumulation. These results demonstrate a striking age-dependent response to Bp, define an early age of extreme susceptibility to Bp, and demonstrate that the neonatal response can be more efficient than the adult response in eliminating bacteria from the lungs, but these neonatal functions are substantially blocked by PTx. This refined definition of "neonatal" mice may be useful in the study of other pathogens that primarily infect neonates, and PTx may prove a particularly valuable tool for probing the poorly understood neonatal immune system.
Collapse
Affiliation(s)
- Colleen J. Sedney
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Amanda Caulfield
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Kaylan K. Dewan
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Uriel Blas-Machado
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Maiya Callender
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Nancy R. Manley
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
| | - Eric T. Harvill
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
18
|
Blockade of the Adenylate Cyclase Toxin Synergizes with Opsonizing Antibodies to Protect Mice against Bordetella pertussis. mBio 2022; 13:e0152722. [PMID: 35920558 PMCID: PMC9426472 DOI: 10.1128/mbio.01527-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bordetella produces an array of virulence factors, including the adenylate cyclase toxin (ACT), which is essential, immunogenic in humans, and highly conserved. Despite mediating immune-evasive functions as a leukotoxin, ACT’s potential role as a protective antigen is unclear. To better understand the contributions of humoral anti-ACT immunity, we evaluated protection against Bordetella pertussis by antibodies binding structurally defined ACT epitopes in a mouse pneumonia model. An ACT-neutralizing antibody, but not a nonneutralizing antibody or an isotype control, significantly increased mouse survival after lethal challenge with B. pertussis. When modified to impair Fc effector functions, the neutralizing antibody retained protective capabilities, indicating that protection was mediated by the blockade of the interactions of ACT with its αMβ2 integrin receptor. After infection with a lower bacterial dose, ACT neutralization synergistically reduced lung bacterial colonization levels when combined with an opsonic antibody binding the surface antigen pertactin. Notably, protection was significantly enhanced when antibodies were administered intranasally as opposed to systemically, indicating that local immune responses are key to antibody-mediated protection against ACT and pertactin. These data reconcile previous conflicting reports to indicate that neutralizing anti-ACT antibodies support the phagocytosis of opsonized B. pertussis and thereby contribute to pertussis protection in vivo.
Collapse
|
19
|
DeJong MA, Wolf MA, Bitzer GJ, Hall JM, Sen-Kilic E, Blake JM, Petty JE, Wong TY, Barbier M, Campbell JD, Bevere JR, Damron FH. CpG 1018® adjuvant enhances Tdap immune responses against Bordetella pertussis in mice. Vaccine 2022; 40:5229-5240. [PMID: 35927132 DOI: 10.1016/j.vaccine.2022.07.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022]
Abstract
Bordetella pertussis is the causative agent of whooping cough (pertussis), a severe respiratory disease that can be fatal, particularly in infants. Despite high vaccine coverage, pertussis remains a problem because the currently used DTaP and Tdap vaccines do not completely prevent infection or transmission. It is well established that the alum adjuvant is a potential weakness of the acellular vaccines because the immunity provided by it is short-term. We aimed to evaluate the potential of CpG 1018® adjuvant to improve antibody responses and enhance protection against B. pertussis challenge in a murine model. A titrated range of Tdap vaccine doses were evaluated in order to best identify the adjuvant capability of CpG 1018. Antibody responses to pertussis toxin (PT), filamentous hemagglutinin (FHA), or the whole bacterium were increased due to the inclusion of CpG 1018. In B. pertussis intranasal challenge studies, we observed improved protection and bacterial clearance from the lower respiratory tract due to adding CpG 1018 to 1/20th the human dose of Tdap. Further, we determined that Tdap and Tdap + CpG 1018 were both capable of facilitating clearance of strains that do not express pertactin (PRN-), which are rising in prevalence. Functional phenotyping of antibodies revealed that the inclusion of CpG 1018 induced more bacterial opsonization and antibodies of the Th1 phenotype (IgG2a and IgG2b). This study demonstrates the potential of adding CpG 1018 to Tdap to improve immunogenicity and protection against B. pertussis compared to the conventional, alum-only adjuvanted Tdap vaccine.
Collapse
Affiliation(s)
- Megan A DeJong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - M Allison Wolf
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Graham J Bitzer
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Jesse M Hall
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Emel Sen-Kilic
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Jeanna M Blake
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Jonathan E Petty
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Ting Y Wong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | | | - Justin R Bevere
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - F Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA.
| |
Collapse
|
20
|
Abstract
![]()
The paradigm of antivirulence
therapy dictates that bacterial pathogens
are specifically disarmed but not killed by neutralizing their virulence
factors. Clearance of the invading pathogen by the immune system is
promoted. As compared to antibiotics, the pathogen-selective antivirulence
drugs hold promise to minimize collateral damage to the beneficial
microbiome. Also, selective pressure for resistance is expected to
be lower because bacterial viability is not directly affected. Antivirulence
drugs are being developed for stand-alone prophylactic and therapeutic
treatments but also for combinatorial use with antibiotics. This Review
focuses on drug modalities that target bacterial exotoxins after the
secretion or release-upon-lysis. Exotoxins have a significant and
sometimes the primary role as the disease-causing virulence factor,
and thereby they are attractive targets for drug development. We describe
the key pre-clinical and clinical trial data that have led to the
approval of currently used exotoxin-targeted drugs, namely the monoclonal
antibodies bezlotoxumab (toxin B/TcdB, Clostridioides difficile), raxibacumab (anthrax toxin, Bacillus anthracis), and obiltoxaximab (anthrax toxin, Bacillus anthracis), but also to challenges with some of the promising leads. We also
highlight the recent developments in pre-clinical research sector
to develop exotoxin-targeted drug modalities, i.e., monoclonal antibodies,
antibody fragments, antibody mimetics, receptor analogs, neutralizing
scaffolds, dominant-negative mutants, and small molecules. We describe
how these exotoxin-targeted drug modalities work with high-resolution
structural knowledge and highlight their advantages and disadvantages
as antibiotic alternatives.
Collapse
Affiliation(s)
- Moona Sakari
- Institute of Biomedicine, Research Unit for Infection and Immunity, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Arttu Laisi
- Institute of Biomedicine, Research Unit for Infection and Immunity, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Arto T. Pulliainen
- Institute of Biomedicine, Research Unit for Infection and Immunity, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| |
Collapse
|
21
|
Ernst K. Novel Strategies to Inhibit Pertussis Toxin. Toxins (Basel) 2022; 14:187. [PMID: 35324684 PMCID: PMC8951090 DOI: 10.3390/toxins14030187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 11/25/2022] Open
Abstract
Pertussis, also known as whooping cough, is a respiratory disease caused by infection with Bordetella pertussis, which releases several virulence factors, including the AB-type pertussis toxin (PT). The characteristic symptom is severe, long-lasting paroxysmal coughing. Especially in newborns and infants, pertussis symptoms, such as leukocytosis, can become life-threatening. Despite an available vaccination, increasing case numbers have been reported worldwide, including Western countries such as Germany and the USA. Antibiotic treatment is available and important to prevent further transmission. However, antibiotics only reduce symptoms if administered in early stages, which rarely occurs due to a late diagnosis. Thus, no causative treatments against symptoms of whooping cough are currently available. The AB-type protein toxin PT is a main virulence factor and consists of a binding subunit that facilitates transport of an enzyme subunit into the cytosol of target cells. There, the enzyme subunit ADP-ribosylates inhibitory α-subunits of G-protein coupled receptors resulting in disturbed cAMP signaling. As an important virulence factor associated with severe symptoms, such as leukocytosis, and poor outcomes, PT represents an attractive drug target to develop novel therapeutic strategies. In this review, chaperone inhibitors, human peptides, small molecule inhibitors, and humanized antibodies are discussed as novel strategies to inhibit PT.
Collapse
Affiliation(s)
- Katharina Ernst
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
22
|
Silva RP, DiVenere AM, Amengor D, Maynard JA. Antibodies binding diverse pertactin epitopes protect mice from B. pertussis infection. J Biol Chem 2022; 298:101715. [PMID: 35151691 PMCID: PMC8931430 DOI: 10.1016/j.jbc.2022.101715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 11/27/2022] Open
Abstract
Infection by the bacterium Bordetella pertussis continues to cause considerable morbidity and mortality worldwide. Many current acellular pertussis vaccines include the antigen pertactin, which has presumptive adhesive and immunomodulatory activities, but is rapidly lost from clinical isolates after the introduction of these vaccines. To better understand the contributions of pertactin antibodies to protection and pertactin's role in pathogenesis, we isolated and characterized recombinant antibodies binding four distinct epitopes on pertactin. We demonstrate that four of these antibodies bind epitopes that are conserved across all three classical Bordetella strains, and competition assays further showed that antibodies binding these epitopes are also elicited by B. pertussis infection of baboons. Surprisingly, we found that representative antibodies binding each epitope protected mice against experimental B. pertussis infection. A cocktail of antibodies from each epitope group protected mice against a subsequent lethal dose of B. pertussis and greatly reduced lung colonization levels after sublethal challenge. Each antibody reduced B. pertussis lung colonization levels up to 100-fold when administered individually, which was significantly reduced when antibody effector functions were impaired, with no antibody mediating antibody-dependent complement-induced lysis. These data suggest that antibodies binding multiple pertactin epitopes protect primarily by the same bactericidal mechanism, which overshadows contributions from blockade of other pertactin functions. These antibodies expand the available tools to further dissect pertactin's role in infection and understand the impact of antipertactin antibodies on bacterial fitness.
Collapse
|
23
|
Locht C, Antoine R. The History of Pertussis Toxin. Toxins (Basel) 2021; 13:623. [PMID: 34564627 PMCID: PMC8472871 DOI: 10.3390/toxins13090623] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 08/27/2021] [Accepted: 09/03/2021] [Indexed: 11/20/2022] Open
Abstract
Besides the typical whooping cough syndrome, infection with Bordetella pertussis or immunization with whole-cell vaccines can result in a wide variety of physiological manifestations, including leukocytosis, hyper-insulinemia, and histamine sensitization, as well as protection against disease. Initially believed to be associated with different molecular entities, decades of research have provided the demonstration that these activities are all due to a single molecule today referred to as pertussis toxin. The three-dimensional structure and molecular mechanisms of pertussis toxin action, as well as its role in protective immunity have been uncovered in the last 50 years. In this article, we review the history of pertussis toxin, including the paradigm shift that occurred in the 1980s which established the pertussis toxin as a single molecule. We describe the role molecular biology played in the understanding of pertussis toxin action, its role as a molecular tool in cell biology and as a protective antigen in acellular pertussis vaccines and possibly new-generation vaccines, as well as potential therapeutical applications.
Collapse
Affiliation(s)
- Camille Locht
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France;
| | | |
Collapse
|
24
|
Barkoff AM, Knuutila A, Mertsola J, He Q. Evaluation of Anti-PT Antibody Response after Pertussis Vaccination and Infection: The Importance of Both Quantity and Quality. Toxins (Basel) 2021; 13:toxins13080508. [PMID: 34437379 PMCID: PMC8402585 DOI: 10.3390/toxins13080508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/21/2022] Open
Abstract
Pertussis toxin (PT) is considered the main virulence factor causing whooping cough or pertussis. The protein is widely studied and its composition was revealed and sequenced already during the 1980s. The human immune system creates a good response against PT when measured in quantity. However, the serum anti-PT antibodies wane rapidly, and only a small amount of these antibodies are found a few years after vaccination/infection. Therefore, multiple approaches to study the functionality (quality) of these antibodies, e.g., avidity, neutralizing capacity, and epitope specificity, have been investigated. In addition, the long-term B cell memory (Bmem) to PT is crucial for good protection throughout life. In this review, we summarize the findings from functional PT antibody and Bmem studies. These results are discussed in line with the quantity of serum anti-PT antibodies. PT neutralizing antibodies and anti-PT antibodies with proper avidity are crucial for good protection against the disease, and certain epitopes have been identified to have multiple functions in the protection. Although PT-specific Bmem responses are detectable at least five years after vaccination, long-term surveillance is lacking. Variation of the natural boosting of circulating Bordetella pertussis in communities is an important confounding factor in these memory studies.
Collapse
Affiliation(s)
- Alex-Mikael Barkoff
- Research Center for Infection and Immunity, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.-M.B.); (A.K.); (J.M.)
| | - Aapo Knuutila
- Research Center for Infection and Immunity, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.-M.B.); (A.K.); (J.M.)
| | - Jussi Mertsola
- Research Center for Infection and Immunity, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.-M.B.); (A.K.); (J.M.)
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, 20520 Turku, Finland
| | - Qiushui He
- Research Center for Infection and Immunity, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.-M.B.); (A.K.); (J.M.)
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
- Correspondence: ; Tel.: +358-40-472-2255
| |
Collapse
|
25
|
Pitisuttithum P, Dhitavat J, Sirivichayakul C, Pitisuthitham A, Sabmee Y, Chinwangso P, Kerdsomboon C, Fortuna L, Spiegel J, Chauhan M, Poredi IK, van den Biggelaar AH, Wijagkanalan W, Viviani S, Mansouri S, Pham HT. Antibody persistence 2 and 3 years after booster vaccination of adolescents with recombinant acellular pertussis monovalent aP gen or combined TdaP gen vaccines. EClinicalMedicine 2021; 37:100976. [PMID: 34386749 PMCID: PMC8343263 DOI: 10.1016/j.eclinm.2021.100976] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/20/2021] [Accepted: 06/04/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Recombinant pertussis vaccines inducing long-lasting immune responses could help to control the rise in pertussis. We here report on persisting antibody responses 2 and 3 years after booster vaccination with a new generation recombinant acellular pertussis vaccine. METHODS Participants of a phase 2/3 randomised-controlled clinical trial with a monovalent pertussis vaccine containing genetically inactivated pertussis toxin (aPgen) or its tetanus and diphtheria toxoids combination (TdaPgen), or a chemically detoxified comparator vaccine (Tdapchem), (originally conducted between July and August 2015) were invited to participate in observational studies of persisting antibody responses 2 and 3 years after vaccination. Serum IgG against pertussis toxin (PT-IgG) and filamentous hemagglutinin (FHA-IgG) were assessed by ELISA, and PT-neutralising antibodies (PT-Nab) by Chinese Hamster Ovary cell assay. FINDINGS Waning of antibodies stabilised in aPgen and TdaPgen vaccinees 2 and 3 years after vaccination. Three years post-vaccination PT-neutralising antibodies remained 4·6-fold (95% Confidence Interval (CI) 2·6-8·1) and 3·7-fold (95% CI 2·2-6·1) higher, PT-IgG antibodies 3·0-fold (95% CI 2·2-4·1) and 2·5-fold (95% CI 1·9-3·3) higher, and FHA-IgG antibodies 1·8-fold (95% CI 1·3-2·5) and 1·6-fold (95% CI 1·2-2·1) higher than baseline in aPgen and TdaPgen recipients, respectively. In the Tdapchem group, PT-neutralising and PT-IgG and FHA-IgG antibodies were back at baseline levels 2 years post-vaccination. Three years post-vaccination seroconversion rates for PT-neutralising antibodies were 65·0% (95% CI 44·1-85·9) and 55·0% (95% CI 33·2-76·8) in aPgen and TdaPgen recipients, respectively. INTERPRETATION Considering the persistence of elevated antibody responses 3 years post-booster vaccination, genetically detoxified monovalent aPgen and TdaPgen vaccines can be expected to induce longer-lasting protection than chemically inactivated Tdap vaccines. FUNDING BioNet-Asia.
Collapse
Affiliation(s)
- Punnee Pitisuttithum
- Vaccine Trial Centre, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Jittima Dhitavat
- Vaccine Trial Centre, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Chukiat Sirivichayakul
- Vaccine Trial Centre, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Arom Pitisuthitham
- Vaccine Trial Centre, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Yupa Sabmee
- Vaccine Trial Centre, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| | - Pailinrut Chinwangso
- BioNet-Asia Co., Ltd., 19 Soi Udomsuk 37, Sukhumvit 103 Road, Bangjak, Prakanong, Bangkok 10260, Thailand
| | - Chawanee Kerdsomboon
- BioNet-Asia Co., Ltd., 19 Soi Udomsuk 37, Sukhumvit 103 Road, Bangjak, Prakanong, Bangkok 10260, Thailand
| | - Librada Fortuna
- BioNet-Asia Co., Ltd., 19 Soi Udomsuk 37, Sukhumvit 103 Road, Bangjak, Prakanong, Bangkok 10260, Thailand
| | - Jane Spiegel
- BioNet-Asia Co., Ltd., 19 Soi Udomsuk 37, Sukhumvit 103 Road, Bangjak, Prakanong, Bangkok 10260, Thailand
| | - Mukesh Chauhan
- BioNet-Asia Co., Ltd., 19 Soi Udomsuk 37, Sukhumvit 103 Road, Bangjak, Prakanong, Bangkok 10260, Thailand
| | - Indrajeet Kumar Poredi
- BioNet-Asia Co., Ltd., 19 Soi Udomsuk 37, Sukhumvit 103 Road, Bangjak, Prakanong, Bangkok 10260, Thailand
| | | | - Wassana Wijagkanalan
- BioNet-Asia Co., Ltd., 19 Soi Udomsuk 37, Sukhumvit 103 Road, Bangjak, Prakanong, Bangkok 10260, Thailand
| | - Simonetta Viviani
- BioNet-Asia Co., Ltd., 19 Soi Udomsuk 37, Sukhumvit 103 Road, Bangjak, Prakanong, Bangkok 10260, Thailand
| | - Souad Mansouri
- BioNet-Asia Co., Ltd., 19 Soi Udomsuk 37, Sukhumvit 103 Road, Bangjak, Prakanong, Bangkok 10260, Thailand
| | - Hong Thai Pham
- BioNet-Asia Co., Ltd., 19 Soi Udomsuk 37, Sukhumvit 103 Road, Bangjak, Prakanong, Bangkok 10260, Thailand
| |
Collapse
|
26
|
Locht C. Long-lived immunity to genetically detoxified pertussis vaccines. EClinicalMedicine 2021; 37:101014. [PMID: 34278279 PMCID: PMC8267544 DOI: 10.1016/j.eclinm.2021.101014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 11/18/2022] Open
Affiliation(s)
- Camille Locht
- U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille F-59000, France
| |
Collapse
|
27
|
Dubois V, Locht C. Mucosal Immunization Against Pertussis: Lessons From the Past and Perspectives. Front Immunol 2021; 12:701285. [PMID: 34211481 PMCID: PMC8239240 DOI: 10.3389/fimmu.2021.701285] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/26/2021] [Indexed: 01/11/2023] Open
Abstract
Background Current vaccination strategies against pertussis are sub-optimal. Optimal protection against Bordetella pertussis, the causative agent of pertussis, likely requires mucosal immunity. Current pertussis vaccines consist of inactivated whole B. pertussis cells or purified antigens thereof, combined with diphtheria and tetanus toxoids. Although they are highly protective against severe pertussis disease, they fail to elicit mucosal immunity. Compared to natural infection, immune responses following immunization are short-lived and fail to prevent bacterial colonization of the upper respiratory tract. To overcome these shortcomings, efforts have been made for decades, and continue to be made, toward the development of mucosal vaccines against pertussis. Objectives In this review we systematically analyzed published literature on protection conferred by mucosal immunization against pertussis. Immune responses mounted by these vaccines are summarized. Method The PubMed Library database was searched for published studies on mucosal pertussis vaccines. Eligibility criteria included mucosal administration and the evaluation of at least one outcome related to efficacy, immunogenicity and safety. Results While over 349 publications were identified by the search, only 63 studies met the eligibility criteria. All eligible studies are included here. Initial attempts of mucosal whole-cell vaccine administration in humans provided promising results, but were not followed up. More recently, diverse vaccination strategies have been tested, including non-replicating and replicating vaccine candidates given by three different mucosal routes: orally, nasally or rectally. Several adjuvants and particulate formulations were tested to enhance the efficacy of non-replicating vaccines administered mucosally. Most novel vaccine candidates were only tested in animal models, mainly mice. Only one novel mucosal vaccine candidate was tested in baboons and in human trials. Conclusion Three vaccination strategies drew our attention, as they provided protective and durable immunity in the respiratory tract, including the upper respiratory tract: acellular vaccines adjuvanted with lipopeptide LP1569 and c-di-GMP, outer membrane vesicles and the live attenuated BPZE1 vaccine. Among all experimental vaccines, BPZE1 is the only one that has advanced into clinical development.
Collapse
Affiliation(s)
- Violaine Dubois
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Camille Locht
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
28
|
Joma M, Fovet CM, Seddiki N, Gressens P, Laforge M. COVID-19 and Pregnancy: Vertical Transmission and Inflammation Impact on Newborns. Vaccines (Basel) 2021; 9:391. [PMID: 33921113 PMCID: PMC8071483 DOI: 10.3390/vaccines9040391] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 12/18/2022] Open
Abstract
The COVID-19 pandemic is ongoing and we are still compiling new findings to decipher and understand SARS-CoV-2 infection during pregnancy. No reports encompass any conclusive confirmation of vertical transmission. Nevertheless, cases of fetal distress and multiple organ failure have been reported, as well as rare cases of fetal demise. While clinicians and scientists continue to seek proof of vertical transmission, they miss the greater point, namely the cause of preterm delivery. In this review, we suggest that the cause might not be due to the viral infection but the fetal exposure to maternal inflammation or cytokine storm that translates into a complication of COVID-19. This statement is extrapolated from previous experience with infections and inflammation which were reported to be fatal by increasing the risk of preterm delivery and causing abnormal neonatal brain development and resulting in neurological disorders like atypical behavioral phenotype or autistic syndrome. Given the potentially fatal consequences on neonate health, we highlight the urgent need for an animal model to study vertical transmission. The preclinical model will allow us to make the link between SARS-COV-2 infection, inflammation and long-term follow-up of child brain development.
Collapse
Affiliation(s)
- Mohamed Joma
- Université de Paris, NeuroDiderot, Inserm, 75019 Paris, France; (M.J.); (P.G.)
| | - Claire-Maelle Fovet
- INSERM U1184, CEA, IDMIT Department, Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB), Université Paris-Saclay, 92265 Fontenay-aux-Roses, France; (C.-M.F.); (N.S.)
| | - Nabila Seddiki
- INSERM U1184, CEA, IDMIT Department, Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB), Université Paris-Saclay, 92265 Fontenay-aux-Roses, France; (C.-M.F.); (N.S.)
| | - Pierre Gressens
- Université de Paris, NeuroDiderot, Inserm, 75019 Paris, France; (M.J.); (P.G.)
| | - Mireille Laforge
- Université de Paris, NeuroDiderot, Inserm, 75019 Paris, France; (M.J.); (P.G.)
| |
Collapse
|
29
|
Locht C. The Path to New Pediatric Vaccines against Pertussis. Vaccines (Basel) 2021; 9:vaccines9030228. [PMID: 33807962 PMCID: PMC7998139 DOI: 10.3390/vaccines9030228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 12/21/2022] Open
Abstract
Whooping cough, or pertussis, mostly caused by Bordetella pertussis, is a respiratory disease that affects all age groups, but severe and fatal pertussis occurs almost exclusively in young children. The widespread use of whole-cell and, more recently, of acellular vaccines has substantially reduced the disease incidence. However, it has not been eliminated in any part of the world and has made a worrisome rebound in several areas. Cocoon and maternal immunization have been implemented in several countries but have their intrinsic limitations. To effectively control pertussis, novel vaccines are needed that protect against disease and prevent B. pertussis infection and transmission, which is not the case for current vaccines. Several approaches are contemplated, including alternative administration routes, such as nasal immunization, improvement of acellular vaccines by adding more antigens and T-cell-promoting adjuvants, and the development of novel vaccines, such as outer membrane vesicles and live attenuated vaccines. Among them, only a live attenuated vaccine has so far been assessed for safety and immunogenicity in preclinical models other than mice and is in clinical development. Before any of these vaccines can be used in neonates, extensive safety and immunogenicity assessment in pre-clinical neonatal models and in carefully designed clinical trials is necessary. The aim of this review is to discuss the current pertussis problem, implemented strategies to resolve it, the value of animal models and novel vaccine approaches.
Collapse
Affiliation(s)
- Camille Locht
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| |
Collapse
|
30
|
Wolf MA, Boehm DT, DeJong MA, Wong TY, Sen-Kilic E, Hall JM, Blackwood CB, Weaver KL, Kelly CO, Kisamore CA, Bitzer GJ, Bevere JR, Barbier M, Damron FH. Intranasal Immunization with Acellular Pertussis Vaccines Results in Long-Term Immunity to Bordetella pertussis in Mice. Infect Immun 2021; 89:e00607-20. [PMID: 33318136 PMCID: PMC8097269 DOI: 10.1128/iai.00607-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
Bordetella pertussis colonizes the respiratory mucosa of humans, inducing an immune response seeded in the respiratory tract. An individual, once convalescent, exhibits long-term immunity to the pathogen. Current acellular pertussis (aP) vaccines do not induce the long-term immune response observed after natural infection in humans. In this study, we evaluated the durability of protection from intranasal (i.n.) pertussis vaccines in mice. Mice that convalesced from B. pertussis infection served as a control group. Mice were immunized with a mock vaccine (phosphate-buffered saline [PBS]), aP only, or an aP base vaccine combined with one of the following adjuvants: alum, curdlan, or purified whole glucan particles (IRI-1501). We utilized two study designs: short term (challenged 35 days after priming vaccination) and long term (challenged 6 months after boost). The short-term study demonstrated that immunization with i.n. vaccine candidates decreased the bacterial burden in the respiratory tract, reduced markers of inflammation, and induced significant serum and lung antibody titers. In the long-term study, protection from bacterial challenge mirrored the results observed in the short-term challenge study. Immunization with pertussis antigens alone was surprisingly protective in both models; however, the alum and IRI-1501 adjuvants induced significant B. pertussis-specific IgG antibodies in both the serum and lung and increased numbers of anti-B. pertussis IgG-secreting plasma cells in the bone marrow. Our data indicate that humoral responses induced by the i.n. vaccines correlated with protection, suggesting that long-term antibody responses can be protective.
Collapse
Affiliation(s)
- M Allison Wolf
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Dylan T Boehm
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Megan A DeJong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Ting Y Wong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Emel Sen-Kilic
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Jesse M Hall
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Catherine B Blackwood
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Kelly L Weaver
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Claire O Kelly
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Caleb A Kisamore
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Graham J Bitzer
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Justin R Bevere
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - F Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| |
Collapse
|
31
|
Cole LE, Zhang J, Pacheco KM, Lhéritier P, Anosova NG, Piolat J, Zheng L, Reveneau N. Immunological Distinctions between Acellular and Whole-Cell Pertussis Immunizations of Baboons Persist for at Least One Year after Acellular Vaccine Boosting. Vaccines (Basel) 2020; 8:vaccines8040729. [PMID: 33276673 PMCID: PMC7761625 DOI: 10.3390/vaccines8040729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/19/2020] [Accepted: 11/26/2020] [Indexed: 01/17/2023] Open
Abstract
While both whole-cell (wP) and acellular pertussis (aP) vaccines have been highly effective at reducing the global pertussis disease burden, there are concerns that compared to wP vaccination, the immune responses to aP vaccination may wane more rapidly. To gain insights into the vaccine elicited immune responses, pre-adult baboons were immunized with either aP or wP vaccines, boosted with an aP vaccine, and observed over a nearly two-year period. Priming with a wP vaccine elicited a more Th17-biased response than priming with aP, whereas priming with an aP vaccine led to a more Th2-biased response than priming with wP. These differences were maintained after aP vaccine boost immunizations. Compared to aP, animals primed with a wP vaccine exhibited greater numbers of pertussis specific memory B cells. While aP and wP vaccine priming initially elicited similar levels of anti-pertussis toxin antibody, titers declined more rapidly in aP vaccine primed animals leading to a 4-fold difference. Both wP and aP vaccine immunization could induce serum bactericidal activity (SBA); however, only one wP vaccine immunization was required to elicit SBA while multiple aP vaccine immunizations were required to elicit lower, less durable SBA titers. In conclusion, when compared to aP vaccine, priming with wP vaccine elicits distinct cellular and humoral immune responses that persist after aP vaccine boosting.
Collapse
Affiliation(s)
- Leah E. Cole
- Sanofi Pasteur, Cambridge, MA 02139, USA; (J.Z.); (K.M.P.); (N.G.A.)
- Correspondence: (L.E.C.); (N.R.); Tel.: +1-617-866-4473 (L.E.C.); +33-4-37-66-8510 (N.R.)
| | - Jinrong Zhang
- Sanofi Pasteur, Cambridge, MA 02139, USA; (J.Z.); (K.M.P.); (N.G.A.)
| | - Kristl M. Pacheco
- Sanofi Pasteur, Cambridge, MA 02139, USA; (J.Z.); (K.M.P.); (N.G.A.)
| | | | | | - Julie Piolat
- Sanofi Pasteur, 69280 Marcy L’Etoile, France; (P.L.); (J.P.)
| | | | - Nathalie Reveneau
- Sanofi Pasteur, 69280 Marcy L’Etoile, France; (P.L.); (J.P.)
- Correspondence: (L.E.C.); (N.R.); Tel.: +1-617-866-4473 (L.E.C.); +33-4-37-66-8510 (N.R.)
| |
Collapse
|
32
|
Lesne E, Cavell BE, Freire-Martin I, Persaud R, Alexander F, Taylor S, Matheson M, van Els CACM, Gorringe A. Acellular Pertussis Vaccines Induce Anti-pertactin Bactericidal Antibodies Which Drives the Emergence of Pertactin-Negative Strains. Front Microbiol 2020; 11:2108. [PMID: 32983069 PMCID: PMC7481377 DOI: 10.3389/fmicb.2020.02108] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022] Open
Abstract
Despite high vaccination coverage, Bordetella pertussis the causative agent of whooping cough is still a health concern worldwide. A resurgence of pertussis cases has been reported, particularly in countries using acellular vaccines with waning immunity and pathogen adaptation thought to be responsible. A better understanding of protective immune responses is needed for the development of improved vaccines. In our study, B. pertussis strain B1917 variants presenting a single gene deletion were generated to analyze the role of vaccine components or candidate vaccine antigens as targets for bactericidal antibodies generated after acellular vaccination or natural infection. Our results show that acellular vaccination generates bactericidal antibodies that are only directed against pertactin. Serum bactericidal assay performed with convalescent samples show that disease induces bactericidal antibodies against Prn but against other antigen(s) as well. Four candidate vaccine antigens (CyaA, Vag8, BrkA, and TcfA) have been studied but were not targets for complement-mediated bactericidal antibodies after natural infection. We confirm that Vag8 and BrkA are involved in complement resistance and would be targeted by blocking antibodies. Our study suggests that the emergence and the widespread circulation of Prn-deficient strains is driven by acellular vaccination and the generation of bactericidal antibodies targeting Prn.
Collapse
Affiliation(s)
- Elodie Lesne
- Public Health England, Porton Down, United Kingdom
| | | | | | - Ruby Persaud
- Public Health England, Porton Down, United Kingdom
| | | | | | | | - Cécile A. C. M. van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | | |
Collapse
|