1
|
Matera MG, Capristo C, de Novellis V, Cazzola M. The ongoing challenge of prevention of pertussis in infants: what's new in 2024? Expert Rev Anti Infect Ther 2025; 23:247-263. [PMID: 40051224 DOI: 10.1080/14787210.2025.2476010] [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: 10/02/2024] [Accepted: 02/28/2025] [Indexed: 05/03/2025]
Abstract
INTRODUCTION Pertussis, a respiratory disease caused primarily by Bordetella pertussis, is undergoing a resurgence despite decades of high rates of vaccination. The prevention of pertussis in infants presents several challenges, including the waning immunity of the acellular pertussis (aP) vaccine, the limited protection afforded to newborns before they complete the vaccine series, and the existence of gaps in maternal vaccination. Furthermore, the unwillingness or refusal of a considerable number of individuals, including some healthcare workers, to receive vaccinations represents another significant challenge. AREAS COVERED This narrative review provides an updated overview of the ongoing challenge of preventing pertussis in infants and discusses some possible solutions. EXPERT OPINION The ongoing challenge of preventing pertussis in infants is multifaceted. To address these challenging issues, a multi-pronged approach is required. This approach should be designed to address various barriers and increase uptake. It should include measures to strengthen maternal vaccination programs, ensure timely infant vaccinations, improve public education, and continue research into more effective vaccines with longer-lasting immunity.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Carlo Capristo
- Unit of Pediatrics, Department of Woman, Child and General and Specialized Surgery, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Vito de Novellis
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|
2
|
Leontari K, Lianou A, Tsantes AG, Filippatos F, Iliodromiti Z, Boutsikou T, Paliatsou S, Chaldoupis AE, Ioannou P, Mpakosi A, Iacovidou N, Sokou R. Pertussis in Early Infancy: Diagnostic Challenges, Disease Burden, and Public Health Implications Amidst the 2024 Resurgence, with Emphasis on Maternal Vaccination Strategies. Vaccines (Basel) 2025; 13:276. [PMID: 40266155 PMCID: PMC11945951 DOI: 10.3390/vaccines13030276] [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: 01/01/2025] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 04/24/2025] Open
Abstract
Bordetella pertussis is the causative agent of pertussis or whooping cough, an acute and highly contagious respiratory infection that can have serious and fatal complications such as pneumonia, encephalopathy, and seizures, especially for newborns. The disease is endemic not only in the European Union (EU)/European Economic Area (EEA) but also globally. Larger outbreaks are anticipated every three to five years, even in countries where vaccination rates are high. Despite the high pertussis vaccination coverage in developed countries and a low rate of pertussis incidence for many years, especially during the COVID-19 pandemic, the incidence of pertussis has been on the rise again, with outbreaks in some places, which is referred to as "re-emergence of pertussis". The aim of this review is to underscore the critical importance of achieving high vaccination coverage, particularly among pregnant women, to safeguard vulnerable neonates from pertussis during their early months, before they are eligible for vaccination. This aligns with the need to address diagnostic challenges, mitigate disease severity, and strengthen public health strategies in light of the ongoing 2024 Bordetella pertussis resurgence.
Collapse
Affiliation(s)
- Konstantina Leontari
- Neonatal Department, School of Medicine, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (K.L.); (S.P.); (N.I.)
| | - Alexandra Lianou
- Neonatal Intensive Care Unit, General Hospital of Nikaia “Agios Panteleimon”, 18454 Piraeus, Greece
| | - Andreas G. Tsantes
- Microbiology Department, “Saint Savvas” Oncology Hospital, 11522 Athens, Greece
| | - Filippos Filippatos
- Neonatal Department, School of Medicine, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (K.L.); (S.P.); (N.I.)
- First Department of Pediatrics, National and Kapodistrian University of Athens, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
| | - Zoi Iliodromiti
- Neonatal Department, School of Medicine, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (K.L.); (S.P.); (N.I.)
| | - Theodora Boutsikou
- Neonatal Department, School of Medicine, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (K.L.); (S.P.); (N.I.)
| | - Styliani Paliatsou
- Neonatal Department, School of Medicine, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (K.L.); (S.P.); (N.I.)
| | - Anastasios E. Chaldoupis
- Laboratory of Haematology and Blood Bank Unit, School of Medicine, “Attiko” Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Petros Ioannou
- School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Alexandra Mpakosi
- Department of Microbiology, General Hospital of Nikaia “Agios Panteleimon”, 18454 Piraeus, Greece;
| | - Nicoletta Iacovidou
- Neonatal Department, School of Medicine, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (K.L.); (S.P.); (N.I.)
| | - Rozeta Sokou
- Neonatal Department, School of Medicine, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (K.L.); (S.P.); (N.I.)
- Neonatal Intensive Care Unit, General Hospital of Nikaia “Agios Panteleimon”, 18454 Piraeus, Greece
| |
Collapse
|
3
|
Fathy Mohamed Y, Fernandez RC. Programming Bordetella pertussis lipid A to promote adjuvanticity. Microb Cell Fact 2024; 23:250. [PMID: 39272136 PMCID: PMC11401268 DOI: 10.1186/s12934-024-02518-7] [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: 06/18/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Bordetella pertussis is the causative agent of whooping cough or pertussis. Although both acellular (aP) and whole-cell pertussis (wP) vaccines protect against disease, the wP vaccine, which is highly reactogenic, is better at preventing colonization and transmission. Reactogenicity is mainly attributed to the lipid A moiety of B. pertussis lipooligosaccharide (LOS). Within LOS, lipid A acts as a hydrophobic anchor, engaging with TLR4-MD2 on host immune cells to initiate both MyD88-dependent and TRIF-dependent pathways, thereby influencing adaptive immune responses. Lipid A variants, such as monophosphoryl lipid A (MPLA) can also act as adjuvants. Adjuvants may overcome the shortcomings of aP vaccines. RESULTS This work used lipid A modifying enzymes from other bacteria to produce an MPLA-like adjuvant strain in B. pertussis. We created B. pertussis strains with distinct lipid A modifications, which were validated using MALDI-TOF. We engineered a hexa-acylated monophosphorylated lipid A that markedly decreased human TLR4 activation and activated the TRIF pathway. The modified lipooligosaccharide (LOS) promoted IRF3 phosphorylation and type I interferon production, similar to MPLA responses. We generated three other variants with increased adjuvanticity properties and reduced endotoxicity. Pyrogenicity studies using the Monocyte Activation Test (MAT) revealed that these four lipid A variants significantly decreased the IL-6, a marker for fever, response in peripheral blood mononuclear cells (PBMCs). CONCLUSION These findings pave the way for developing wP vaccines that are possibly less reactogenic and designing adaptable adjuvants for current vaccine formulations, advancing more effective immunization strategies against pertussis.
Collapse
Affiliation(s)
- Yasmine Fathy Mohamed
- Department of Microbiology & Immunology, The University of British Columbia, Vancouver, British Columbia, V6T1Z3, Canada
- Department of Microbiology & Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Rachel C Fernandez
- Department of Microbiology & Immunology, The University of British Columbia, Vancouver, British Columbia, V6T1Z3, Canada.
| |
Collapse
|
4
|
Kim AR, Sette A, da Silva Antunes R. Adaptive immune response to bordetella pertussis during vaccination and infection: emerging perspectives and unanswered questions. Expert Rev Vaccines 2024; 23:705-714. [PMID: 39037200 PMCID: PMC11306532 DOI: 10.1080/14760584.2024.2383745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
INTRODUCTION Whooping cough, also known as pertussis, remains a significant challenge as a vaccine-preventable disease worldwide. Since the switch from the whole-cell Pertussis (wP) vaccine to the acellular Pertussis vaccine (aP), cases of whooping cough have increased in countries using the aP vaccine. Understanding the immune system's response to pertussis vaccines and infection is crucial for improving current vaccine efficacy. AREAS COVERED This review of the literature using PubMed records offers an overview of the qualitative differences in antibody and T cell responses to B. pertussis (BP) in vaccination and infection, and their potential association with decreased efficacy of the aP vaccine in preventing infection and subclinical colonization. We further discuss how asymptomatic infections and carriage are widespread among vaccinated human populations, and explore methodologies that can be employed for their detection, to better understand their impact on adaptive immune responses and identify key features necessary for protection against the disease. EXPERT OPINION An underappreciated human BP reservoir, stemming from the decreased capacity of the aP vaccine to prevent subclinical infection, offers an alternative explanation for the increased incidence of clinical disease and recurrent outbreaks.
Collapse
Affiliation(s)
- A-Reum Kim
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Disease and Global Public Health, University of California San Diego (UCSD), La Jolla, CA, 92037, USA
| | | |
Collapse
|
5
|
Abu-Raya B, Esser MJ, Nakabembe E, Reiné J, Amaral K, Diks AM, Imede E, Way SS, Harandi AM, Gorringe A, Le Doare K, Halperin SA, Berkowska MA, Sadarangani M. Antibody and B-cell Immune Responses Against Bordetella Pertussis Following Infection and Immunization. J Mol Biol 2023; 435:168344. [PMID: 37926426 DOI: 10.1016/j.jmb.2023.168344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Neither immunization nor recovery from natural infection provides life-long protection against Bordetella pertussis. Replacement of a whole-cell pertussis (wP) vaccine with an acellular pertussis (aP) vaccine, mutations in B. pertussis strains, and better diagnostic techniques, contribute to resurgence of number of cases especially in young infants. Development of new immunization strategies relies on a comprehensive understanding of immune system responses to infection and immunization and how triggering these immune components would ensure protective immunity. In this review, we assess how B cells, and their secretory products, antibodies, respond to B. pertussis infection, current and novel vaccines and highlight similarities and differences in these responses. We first focus on antibody-mediated immunity. We discuss antibody (sub)classes, elaborate on antibody avidity, ability to neutralize pertussis toxin, and summarize different effector functions, i.e. ability to activate complement, promote phagocytosis and activate NK cells. We then discuss challenges and opportunities in studying B-cell immunity. We highlight shared and unique aspects of B-cell and plasma cell responses to infection and immunization, and discuss how responses to novel immunization strategies better resemble those triggered by a natural infection (i.e., by triggering responses in mucosa and production of IgA). With this comprehensive review, we aim to shed some new light on the role of B cells and antibodies in the pertussis immunity to guide new vaccine development.
Collapse
Affiliation(s)
- Bahaa Abu-Raya
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.
| | - Mirjam J Esser
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Eve Nakabembe
- Centre for Neonatal and Paediatric Infectious Diseases Research, St George's, University of London, Cranmer Terrace, London SW17 0RE, UK; Department of Obstetrics and Gynaecology, Makerere University College of Health Sciences, Upper Mulago Hill Road, Kampala, P.O. Box 7072, Uganda
| | - Jesús Reiné
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom; Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Kyle Amaral
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Annieck M Diks
- Department of Immunology, Leiden University Medical Center, Albinusdreef 2, Leiden ZA 2333, the Netherlands
| | - Esther Imede
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Sing Sing Way
- Department of Pediatrics, Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Ali M Harandi
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Andrew Gorringe
- UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK
| | - Kirsty Le Doare
- Centre for Neonatal and Paediatric Infectious Diseases Research, St George's, University of London, Cranmer Terrace, London SW17 0RE, UK; Makerere University-Johns Hopkins University Research Collaboration, MU-JHU, Upper Mulago Hill, Kampala, P.O. Box 23491, Uganda
| | - Scott A Halperin
- Canadian Center for Vaccinology, Departments of Pediatrics and Microbiology and Immunology, Dalhousie University, Izaak Walton Killam Health Centre, and Nova Scotia Health Authority, Halifax, NS, Canada
| | - Magdalena A Berkowska
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
6
|
Miguelena Chamorro B, De Luca K, Swaminathan G, Longet S, Mundt E, Paul S. Bordetella bronchiseptica and Bordetella pertussis: Similarities and Differences in Infection, Immuno-Modulation, and Vaccine Considerations. Clin Microbiol Rev 2023; 36:e0016422. [PMID: 37306571 PMCID: PMC10512794 DOI: 10.1128/cmr.00164-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023] Open
Abstract
Bordetella pertussis and Bordetella bronchiseptica belong to the genus Bordetella, which comprises 14 other species. B. pertussis is responsible for whooping cough in humans, a severe infection in children and less severe or chronic in adults. These infections are restricted to humans and currently increasing worldwide. B. bronchiseptica is involved in diverse respiratory infections in a wide range of mammals. For instance, the canine infectious respiratory disease complex (CIRDC), characterized by a chronic cough in dogs. At the same time, it is increasingly implicated in human infections, while remaining an important pathogen in the veterinary field. Both Bordetella can evade and modulate host immune responses to support their persistence, although it is more pronounced in B. bronchiseptica infection. The protective immune responses elicited by both pathogens are comparable, while there are important characteristics in the mechanisms that differ. However, B. pertussis pathogenesis is more difficult to decipher in animal models than those of B. bronchiseptica because of its restriction to humans. Nevertheless, the licensed vaccines for each Bordetella are different in terms of formulation, route of administration and immune responses induced, with no known cross-reaction between them. Moreover, the target of the mucosal tissues and the induction of long-lasting cellular and humoral responses are required to control and eliminate Bordetella. In addition, the interaction between both veterinary and human fields are essential for the control of this genus, by preventing the infections in animals and the subsequent zoonotic transmission to humans.
Collapse
Affiliation(s)
- Beatriz Miguelena Chamorro
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Lyon, France
- Boehringer Ingelheim, Global Innovation, Saint-Priest, France
| | - Karelle De Luca
- Boehringer Ingelheim, Global Innovation, Saint-Priest, France
| | | | - Stéphanie Longet
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Lyon, France
- CIC Inserm 1408 Vaccinology, Saint-Etienne, France
| | - Egbert Mundt
- Boehringer Ingelheim, Global Innovation, Saint-Priest, France
| | - Stéphane Paul
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Lyon, France
- CIC Inserm 1408 Vaccinology, Saint-Etienne, France
| |
Collapse
|
7
|
Kehagia E, Papakyriakopoulou P, Valsami G. Advances in intranasal vaccine delivery: A promising non-invasive route of immunization. Vaccine 2023:S0264-410X(23)00529-7. [PMID: 37179163 PMCID: PMC10173027 DOI: 10.1016/j.vaccine.2023.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 04/25/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023]
Abstract
The importance of vaccination has been proven particularly significant the last three years, as it is revealed to be the most efficient weapon for the prevention of several infections including SARS-COV-2. Parenteral vaccination is the most applicable method of immunization, for the prevention of systematic and respiratory infections, or central nervous system disorders, involving T and B cells to a whole-body immune response. However, the mucosal vaccines, such as nasal vaccines, can additionally activate the immune cells localized on the mucosal tissue of the upper and lower respiratory tract. This dual stimulation of the immune system, along with their needle-free administration favors the development of novel nasal vaccines to produce long-lasting immunity. In recent years, the nanoparticulate systems have been extensively involved in the formulation of nasal vaccines as polymeric, polysaccharide and lipid ones, as well as in the form of proteosomes, lipopeptides and virosomes. Advanced delivery nanosystems have been designed and evaluated as carriers or adjuvants for nasal vaccination. To this end, several nanoparticulate vaccines are undergone clinical trials as promising candidates for nasal immunization, while nasal vaccines against influenza type A and B and hepatitis B have been approved by health authorities. This comprehensive literature review aims to summarize the critical aspects of these formulations and highlight their potential for the future establishment of nasal vaccination. Both preclinical (in vitro and in vivo) and clinical studies are incorporated, summarized, and critically discussed, as well as the limitations of nasal immunization.
Collapse
Affiliation(s)
- Eleni Kehagia
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15784, Greece
| | - Paraskevi Papakyriakopoulou
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15784, Greece.
| | - Georgia Valsami
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15784, Greece
| |
Collapse
|
8
|
Yu M, Charles A, Cagigi A, Christ W, Österberg B, Falck-Jones S, Azizmohammadi L, Åhlberg E, Falck-Jones R, Svensson J, Nie M, Warnqvist A, Hellgren F, Lenart K, Arcoverde Cerveira R, Ols S, Lindgren G, Lin A, Maecker H, Bell M, Johansson N, Albert J, Sundling C, Czarnewski P, Klingström J, Färnert A, Loré K, Smed-Sörensen A. Delayed generation of functional virus-specific circulating T follicular helper cells correlates with severe COVID-19. Nat Commun 2023; 14:2164. [PMID: 37061513 PMCID: PMC10105364 DOI: 10.1038/s41467-023-37835-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 04/03/2023] [Indexed: 04/17/2023] Open
Abstract
Effective humoral immune responses require well-orchestrated B and T follicular helper (Tfh) cell interactions. Whether these interactions are impaired and associated with COVID-19 disease severity is unclear. Here, longitudinal blood samples across COVID-19 disease severity are analysed. We find that during acute infection SARS-CoV-2-specific circulating Tfh (cTfh) cells expand with disease severity. SARS-CoV-2-specific cTfh cell frequencies correlate with plasmablast frequencies and SARS-CoV-2 antibody titers, avidity and neutralization. Furthermore, cTfh cells but not other memory CD4 T cells, from severe patients better induce plasmablast differentiation and antibody production compared to cTfh cells from mild patients. However, virus-specific cTfh cell development is delayed in patients that display or later develop severe disease compared to those with mild disease, which correlates with delayed induction of high-avidity neutralizing antibodies. Our study suggests that impaired generation of functional virus-specific cTfh cells delays high-quality antibody production at an early stage, potentially enabling progression to severe disease.
Collapse
Affiliation(s)
- Meng Yu
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Afandi Charles
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Alberto Cagigi
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Wanda Christ
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Björn Österberg
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sara Falck-Jones
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lida Azizmohammadi
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Eric Åhlberg
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ryan Falck-Jones
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Julia Svensson
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mu Nie
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Warnqvist
- Division of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fredrika Hellgren
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Klara Lenart
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Rodrigo Arcoverde Cerveira
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sebastian Ols
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Gustaf Lindgren
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ang Lin
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Holden Maecker
- The Human Immune Monitoring Center, Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Max Bell
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Niclas Johansson
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Jan Albert
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Clinical Microbiology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Paulo Czarnewski
- Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Jonas Klingström
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anna Färnert
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
9
|
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
|
10
|
Diks AM, de Graaf H, Teodosio C, Groenland RJ, de Mooij B, Ibrahim M, Hill AR, Read RC, van Dongen JJ, Berkowska MA. Distinct early cellular kinetics in participants protected against colonization upon Bordetella pertussis challenge. J Clin Invest 2023; 133:163121. [PMID: 36649086 PMCID: PMC9974097 DOI: 10.1172/jci163121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUNDTo date, only limited data are available on the mechanisms of protection against colonization with Bordetella pertussis in humans.METHODSIn this study, the cellular responses to B. pertussis challenge were monitored longitudinally using high-dimensional EuroFlow-based flow cytometry, allowing quantitative detection of more than 250 different immune cell subsets in the blood of 15 healthy donors.RESULTSParticipants who were protected against colonization showed different early cellular responses compared with colonized participants. Especially prominent for colonization-protected participants were the early expansion of CD36- nonclassical monocytes on day 1 (D1), natural killer cells (D3), follicular T helper cells (D1-D3), and plasma cells (D3). Plasma cell expansion on D3 correlated negatively with the CFU load on D7 and D9 after challenge. Increased plasma cell maturation on D11-D14 was found in participants with seroconversion.CONCLUSIONThese early cellular immune responses following experimental infection can now be further characterized and potentially linked to an efficient mucosal immune response, preventing colonization. Ultimately, their presence may be used to evaluate whether new B. pertussis vaccine candidates are protective against B. pertussis colonization, e.g., by bacterial challenge after vaccination.TRIAL REGISTRATIONClinicalTrials.gov NCT03751514.FUNDINGInnovative Medicines Initiative 2 Joint Undertaking and the EuroFlow Consortium.
Collapse
Affiliation(s)
- Annieck M Diks
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Hans de Graaf
- Faculty of Medicine and.,NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Cristina Teodosio
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands.,Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC, USAL-CSIC-FICUS) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Rick J Groenland
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Bas de Mooij
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Muktar Ibrahim
- NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Alison R Hill
- NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Robert C Read
- NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Jacques Jm van Dongen
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands.,Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC, USAL-CSIC-FICUS) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | | | | |
Collapse
|
11
|
Locht C. Pasteurian Contributions to the Study of Bordetella pertussis Toxins. Toxins (Basel) 2023; 15:toxins15030176. [PMID: 36977067 PMCID: PMC10054083 DOI: 10.3390/toxins15030176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
As a tribute to Louis Pasteur on the occasion of the 200th anniversary of his birth, this article summarizes the main contributions of scientists from Pasteur Institutes to the current knowledge of toxins produced by Bordetella pertussis. The article therefore focuses on publications authored by researchers from Pasteur Institutes and is not intended as a systematic review of B. pertussis toxins. Besides identifying B. pertussis as the causative agent of whooping cough, Pasteurians have made several major contributions with respect to the structure–function relationship of the Bordetella lipo-oligosaccharide, adenylyl cyclase toxin and pertussis toxin. In addition to contributing to the understanding of these toxins’ mechanisms at the molecular and cellular levels and their role in pathogenesis, scientists at Pasteur Institutes have also exploited potential applications of the gathered knowledge of these toxins. These applications range from the development of novel tools to study protein–protein interactions over the design of novel antigen delivery tools, such as prophylactic or therapeutic vaccine candidates against cancer and viral infection, to the development of a live attenuated nasal pertussis vaccine. This scientific journey from basic science to applications in the field of human health matches perfectly with the overall scientific objectives outlined by Louis Pasteur himself.
Collapse
Affiliation(s)
- Camille Locht
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Centre for Infection and Immunity of Lille, F-59000 Lille, France
| |
Collapse
|
12
|
Liu J, Yan XD, Li XQ, Du YH, Zhu LL, Ye TT, Cao ZY, Dong ZW, Li ST, Xu X, Bai W, Li D, Zhang JW, Wang SJ, Li SH, Sun J, Yin XZ. Chrysanthemum sporopollenin: A novel vaccine delivery system for nasal mucosal immunity. Front Immunol 2023; 14:1132129. [PMID: 36845130 PMCID: PMC9947463 DOI: 10.3389/fimmu.2023.1132129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/27/2023] [Indexed: 02/11/2023] Open
Abstract
Objective Mucosal immunization was an effective defender against pathogens. Nasal vaccines could activate both systemic and mucosal immunity to trigger protective immune responses. However, due to the weak immunogenicity of nasal vaccines and the lack of appropriate antigen carriers, very few nasal vaccines have been clinically approved for human use, which was a major barrier to the development of nasal vaccines. Plant-derived adjuvants are promising candidates for vaccine delivery systems due to their relatively safe immunogenic properties. In particular, the distinctive structure of pollen was beneficial to the stability and retention of antigen in the nasal mucosa. Methods Herein, a novel wild-type chrysanthemum sporopollenin vaccine delivery system loaded with a w/o/w emulsion containing squalane and protein antigen was fabricated. The unique internal cavities and the rigid external walls within the sporopollenin skeleton construction could preserve and stabilize the inner proteins. The external morphological characteristics were suitable for nasal mucosal administration with high adhesion and retention. Results Secretory IgA antibodies in the nasal mucosa can be induced by the w/o/w emulsion with the chrysanthemum sporopollenin vaccine delivery system. Moreover, the nasal adjuvants produce a stronger humoral response (IgA and IgG) compared to squalene emulsion adjuvant. Mucosal adjuvant benefited primarily from prolongation of antigens in the nasal cavity, improvement of antigen penetration in the submucosa and promotion of CD8+ T cells in spleen. Disccusion Based on effective delivering both the adjuvant and the antigen, the increase of protein antigen stability and the realization of mucosal retention, the chrysanthemum sporopollenin vaccine delivery system has the potential to be a promising adjuvant platform. This work provide a novel idea for the fabrication of protein-mucosal delivery vaccine.
Collapse
Affiliation(s)
- Jun Liu
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
- Ji-Wen Zhang laboratory, Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong New Area, Shanghai, China
| | - Xiao-Dan Yan
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Xian-Qiang Li
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Yu-Hao Du
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Li-Li Zhu
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Tian-Tian Ye
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Ze-Ying Cao
- Ji-Wen Zhang laboratory, Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong New Area, Shanghai, China
| | - Zhe-Wen Dong
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Shu-Tao Li
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Xue Xu
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Wei Bai
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Dan Li
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Ji-Wen Zhang
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
- Ji-Wen Zhang laboratory, Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong New Area, Shanghai, China
| | - Shu-Jun Wang
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Shan-Hu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing, China
| | - Jin Sun
- Pharmacy laboratory, Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Xian-Zhen Yin
- Ji-Wen Zhang laboratory, Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong New Area, Shanghai, China
- Lingang Laboratory, Shanghai, China
| |
Collapse
|
13
|
Molina Estupiñan JL, Aradottir Pind AA, Foroutan Pajoohian P, Jonsdottir I, Bjarnarson SP. The adjuvants dmLT and mmCT enhance humoral immune responses to a pneumococcal conjugate vaccine after both parenteral or mucosal immunization of neonatal mice. Front Immunol 2023; 13:1078904. [PMID: 36741402 PMCID: PMC9896006 DOI: 10.3389/fimmu.2022.1078904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/12/2022] [Indexed: 01/21/2023] Open
Abstract
Immaturity of the neonatal immune system contributes to increased susceptibility to infectious diseases and poor vaccine responses. Therefore, better strategies for early life vaccination are needed. Adjuvants can enhance the magnitude and duration of immune responses. In this study we assessed the effects of the adjuvants dmLT and mmCT and different immunization routes, subcutaneous (s.c.) and intranasal (i.n.), on neonatal immune response to a pneumococcal conjugate vaccine Pn1-CRM197. Pn1-specific antibody (Ab) levels of neonatal mice immunized with Pn1-CRM197 alone were low. The adjuvants enhanced IgG Ab responses up to 8 weeks after immunization, more after s.c. than i.n. immunization. On the contrary, i.n. immunization with either adjuvant enhanced serum and salivary IgA levels more than s.c. immunization. In addition, both dmLT and mmCT enhanced germinal center formation and accordingly, dmLT and mmCT enhanced the induction and persistence of Pn1-specific IgG+ Ab-secreting cells (ASCs) in spleen and bone marrow (BM), irrespective of the immunization route. Furthermore, i.n. immunization enhanced Pn1-specific IgA+ ASCs in BM more than s.c. immunizatiofimmu.2022.1078904n. However, a higher i.n. dose of the Pn1-CRM197 was needed to achieve IgG response comparable to that elicited by s.c. immunization with either adjuvant. We conclude that dmLT and mmCT enhance both induction and persistence of the neonatal immune response to the vaccine Pn1-CRM197, following mucosal or parenteral immunization. This indicates that dmLT and mmCT are promising adjuvants for developing safe and effective early life vaccination strategies.
Collapse
Affiliation(s)
- Jenny Lorena Molina Estupiñan
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Audur Anna Aradottir Pind
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Poorya Foroutan Pajoohian
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Ingileif Jonsdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Stefania P. Bjarnarson
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland,*Correspondence: Stefania P. Bjarnarson,
| |
Collapse
|
14
|
Blanchard-Rohner G. Novel approaches to reactivate pertussis immunity. Expert Rev Vaccines 2022; 21:1787-1797. [PMID: 36400443 DOI: 10.1080/14760584.2022.2149499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Whole cell and acellular pertussis vaccines have been very effective in decreasing the deaths of neonates and infants from Bordetella pertussis. Despite high vaccine coverage worldwide, pertussis remains one of the most common vaccine-preventable diseases, thus suggesting that new pertussis vaccination strategies are needed. Several candidates are currently under development, such as acellular pertussis vaccines that use genetically detoxified pertussis toxin, acellular pertussis vaccines delivered with new adjuvants or new delivery systems, or an intranasally delivered, live attenuated vaccine. AREAS COVERED This review discusses the different possibilities for improving current pertussis vaccines and the present state of knowledge on the pertussis vaccine candidates under development. EXPERT OPINION Until there is a safe, effective, and affordable alternative to the two types of existing vaccines, we should maintain sufficient childhood coverage and increase the vaccination of pregnant women, adolescents, and young adults.
Collapse
Affiliation(s)
- Geraldine Blanchard-Rohner
- Center of Vaccinology, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Unit of Immunology and Vaccinology, Division of General Pediatrics, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| |
Collapse
|
15
|
Valeri V, Sochon A, Cousu C, Chappert P, Lecoeuche D, Blanc P, Weill JC, Reynaud CA. The whole-cell pertussis vaccine imposes a broad effector B cell response in mouse heterologous prime-boost settings. JCI Insight 2022; 7:157034. [PMID: 36136586 PMCID: PMC9675447 DOI: 10.1172/jci.insight.157034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 09/16/2022] [Indexed: 12/15/2022] Open
Abstract
ÍSince the introduction of new generation pertussis vaccines, resurgence of pertussis has been observed in many developed countries. Former whole-cell pertussis (wP) vaccines are able to protect against disease and transmission but have been replaced in several industrialized countries because of their reactogenicity and adverse effects. Current acellular pertussis (aP) vaccines, made of purified proteins of Bordetella pertussis, are efficient at preventing disease but fail to induce long-term protection from infection. While the systemic and mucosal T cell immunity induced by the 2 types of vaccines has been well described, much less is known concerning B cell responses. Taking advantage of an inducible activation-induced cytidine deaminase fate-mapping mouse model, we compared effector and memory B cells induced by the 2 classes of vaccines and showed that a stronger and broader memory B cell and plasma cell response was achieved by a wP prime. We also observed that homologous or heterologous vaccine combinations that include at least 1 wP administration, even as a booster dose, were sufficient to induce this broad effector response, thus highlighting its dominant imprint on the B cell profile. Finally, we describe the settlement of memory B cell populations in the lung following subcutaneous wP prime vaccination.
Collapse
Affiliation(s)
- Viviana Valeri
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMR 8253, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Akhésa Sochon
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMR 8253, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Clara Cousu
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMR 8253, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Pascal Chappert
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMR 8253, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Damiana Lecoeuche
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMR 8253, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Jean-Claude Weill
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMR 8253, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Claude-Agnès Reynaud
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMR 8253, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
16
|
Nian X, Zhang J, Huang S, Duan K, Li X, Yang X. Development of Nasal Vaccines and the Associated Challenges. Pharmaceutics 2022; 14:1983. [PMID: 36297419 PMCID: PMC9609876 DOI: 10.3390/pharmaceutics14101983] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 02/02/2024] Open
Abstract
Viruses, bacteria, fungi, and several other pathogenic microorganisms usually infect the host via the surface cells of respiratory mucosa. Nasal vaccination could provide a strong mucosal and systemic immunity to combat these infections. The intranasal route of vaccination offers the advantage of easy accessibility over the injection administration. Therefore, nasal immunization is considered a promising strategy for disease prevention, particularly in the case of infectious diseases of the respiratory system. The development of a nasal vaccine, particularly the strategies of adjuvant and antigens design and optimization, enabling rapid induction of protective mucosal and systemic responses against the disease. In recent times, the development of efficacious nasal vaccines with an adequate safety profile has progressed rapidly, with effective handling and overcoming of the challenges encountered during the process. In this context, the present report summarizes the most recent findings regarding the strategies used for developing nasal vaccines as an efficient alternative to conventional vaccines.
Collapse
Affiliation(s)
- Xuanxuan Nian
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Jiayou Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Shihe Huang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Kai Duan
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Xinguo Li
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Xiaoming Yang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- China National Biotech Group Company Limited, Beijing 100029, China
| |
Collapse
|
17
|
Lin X, Sheng Y, Zhang X, Li Z, Yang Y, Wu J, Su Z, Ma G, Zhang S. Oil-in-ionic liquid nanoemulsion-based intranasal delivery system for influenza split-virus vaccine. J Control Release 2022; 346:380-391. [PMID: 35483639 DOI: 10.1016/j.jconrel.2022.04.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 01/12/2023]
Abstract
Effective antigen delivery and immune stimulation in nasal mucosa determine the success of mucosal immunity. Here, an oil-in-ionic liquid (o/IL) nanoemulsion formulated with choline and niacin IL ([Cho][Nic]), squalene, and Tween 80 surfactant is explored as a vaccine delivery system for intranasal mucosal immunization. Compared to the o/w emulsion counterpart without the ILs, the o/IL manoemulsion showed a reduced and more uniform size of approximately 168 nm and significantly improved stability. Studies in mice model showed that when was used as an intranasal vaccine delivery system for influenza split-virus antigens, the antigens in the o/IL nanoemulsion induced strong mucosal immune responses with secretory IgA titers 25- and 5.8-fold higher than those of naked and commercial MF59-adjuvanted antigens, respectively. The o/IL nanoemulsion system also induced stronger systemic humoral responses. The excellent mucosal adjuvant effects of the o/IL nanoemulsion mainly benefited from the prolonged retention of antigens in the nasal cavity, enhanced antigen permeation into the submucosa, and the consequently promoted proliferation of CD11b cells and CD4+ T cells in nasal mucosa-associated lymphoid tissue. Moreover, when used as an injection adjuvant, the o/IL nanoemulsion also induced stronger humoral immune responses than MF59. Thus, the [Cho][Nic]-based o/IL nanoemulsion vaccine delivery system can serve as a promising adjuvant platform.
Collapse
Affiliation(s)
- Xuan Lin
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Yanan Sheng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xuan Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Zhengjun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Yanli Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Jie Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Zhiguo Su
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Songping Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China.
| |
Collapse
|
18
|
Age and Primary Vaccination Background Influence the Plasma Cell Response to Pertussis Booster Vaccination. Vaccines (Basel) 2022; 10:vaccines10020136. [PMID: 35214595 PMCID: PMC8878388 DOI: 10.3390/vaccines10020136] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023] Open
Abstract
Pertussis is a vaccine-preventable disease caused by the bacterium Bordetella pertussis. Over the past years, the incidence and mortality of pertussis increased significantly. A possible cause is the switch from whole-cell to acellular pertussis vaccines, although other factors may also contribute. Here, we applied high-dimensional flow cytometry to investigate changes in B cells in individuals of different ages and distinct priming backgrounds upon administration of an acellular pertussis booster vaccine. Participants were divided over four age cohorts. We compared longitudinal kinetics within each cohort and between the different cohorts. Changes in the B-cell compartment were correlated to numbers of vaccine-specific B- and plasma cells and serum Ig levels. Expansion and maturation of plasma cells 7 days postvaccination was the most prominent cellular change in all age groups and was most pronounced for more mature IgG1+ plasma cells. Plasma cell responses were stronger in individuals primed with whole-cell vaccine than in individuals primed with acellular vaccine. Moreover, IgG1+ and IgA1+ plasma cell expansion correlated with FHA-, Prn-, or PT- specific serum IgG or IgA levels. Our study indicates plasma cells as a potential early cellular marker of an immune response and contributes to understanding differences in immune responses between age groups and primary vaccination backgrounds.
Collapse
|
19
|
Mucosal Immunization with DTaP Confers Protection against Bordetella pertussis Infection and Cough in Sprague-Dawley Rats. Infect Immun 2021; 89:e0034621. [PMID: 34516235 PMCID: PMC8594602 DOI: 10.1128/iai.00346-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pertussis is a respiratory disease caused by the Gram-negative pathogen, Bordetella pertussis. The transition from a whole-cell pertussis vaccine (wP and DTP) to an acellular pertussis vaccine (aP, DTaP, and Tdap) correlates with an increase in pertussis cases, despite widespread vaccine implementation and coverage, and it is now appreciated that the protection provided by aP rapidly wanes. To recapitulate the localized immunity observed from natural infection, mucosal vaccination with aP was explored using the coughing rat model of pertussis. Overall, our goal was to evaluate the route of vaccination in the coughing rat model of pertussis. Immunity induced by both oral gavage and intranasal vaccination of aP in B. pertussis challenged rats over a 9-day infection was compared to intramuscular wP (IM-wP)- and IM-aP-immunized rats that were used as positive controls. Our data demonstrate that mucosal immunization of aP resulted in the production of anti-B. pertussis IgG antibody titers similar to IM-wP- and IM-aP-vaccinated controls postchallenge. IN-aP also induced anti-B. pertussis IgA antibodies in the nasal cavity. Immunization with IM-wP, IM-aP, IN-aP, and OG-aP immunization protected against B. pertussis-induced cough, whereas OG-aP immunization did not protect against respiratory distress. Mucosal immunization by both intranasal and oral gavage administration protected against acute inflammation and decreased bacterial burden in the lung compared to mock-vaccinated challenge rats. The data presented in this study suggest that mucosal vaccination with aP can induce a mucosal immune response and provide protection against B. pertussis challenge. This study highlights the potential benefits and uses of the coughing rat model of pertussis; however, further questions regarding waning immunity still require additional investigation.
Collapse
|
20
|
Saso A, Kampmann B, Roetynck S. Vaccine-Induced Cellular Immunity against Bordetella pertussis: Harnessing Lessons from Animal and Human Studies to Improve Design and Testing of Novel Pertussis Vaccines. Vaccines (Basel) 2021; 9:877. [PMID: 34452002 PMCID: PMC8402596 DOI: 10.3390/vaccines9080877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022] Open
Abstract
Pertussis ('whooping cough') is a severe respiratory tract infection that primarily affects young children and unimmunised infants. Despite widespread vaccine coverage, it remains one of the least well-controlled vaccine-preventable diseases, with a recent resurgence even in highly vaccinated populations. Although the exact underlying reasons are still not clear, emerging evidence suggests that a key factor is the replacement of the whole-cell (wP) by the acellular pertussis (aP) vaccine, which is less reactogenic but may induce suboptimal and waning immunity. Differences between vaccines are hypothesised to be cell-mediated, with polarisation of Th1/Th2/Th17 responses determined by the composition of the pertussis vaccine given in infancy. Moreover, aP vaccines elicit strong antibody responses but fail to protect against nasal colonisation and/or transmission, in animal models, thereby potentially leading to inadequate herd immunity. Our review summarises current knowledge on vaccine-induced cellular immune responses, based on mucosal and systemic data collected within experimental animal and human vaccine studies. In addition, we describe key factors that may influence cell-mediated immunity and how antigen-specific responses are measured quantitatively and qualitatively, at both cellular and molecular levels. Finally, we discuss how we can harness this emerging knowledge and novel tools to inform the design and testing of the next generation of improved infant pertussis vaccines.
Collapse
Affiliation(s)
- Anja Saso
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Beate Kampmann
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Sophie Roetynck
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| |
Collapse
|
21
|
Abstract
Mucosal vaccines offer the potential to trigger robust protective immune responses at the predominant sites of pathogen infection. In principle, the induction of adaptive immunity at mucosal sites, involving secretory antibody responses and tissue-resident T cells, has the capacity to prevent an infection from becoming established in the first place, rather than only curtailing infection and protecting against the development of disease symptoms. Although numerous effective mucosal vaccines are in use, the major advances seen with injectable vaccines (including adjuvanted subunit antigens, RNA and DNA vaccines) have not yet been translated into licensed mucosal vaccines, which currently comprise solely live attenuated and inactivated whole-cell preparations. The identification of safe and effective mucosal adjuvants allied to innovative antigen discovery and delivery strategies is key to advancing mucosal vaccines. Significant progress has been made in resolving the mechanisms that regulate innate and adaptive mucosal immunity and in understanding the crosstalk between mucosal sites, and this provides valuable pointers to inform mucosal adjuvant design. In particular, increased knowledge on mucosal antigen-presenting cells, innate lymphoid cell populations and resident memory cells at mucosal sites highlights attractive targets for vaccine design. Exploiting these insights will allow new vaccine technologies to be leveraged to facilitate rational mucosal vaccine design for pathogens including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and for cancer.
Collapse
|
22
|
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
|
23
|
Camp JV, Wilson RL, Singletary M, Blanchard JL, Aldovini A, Kaminski RW, Oaks EV, Kozlowski PA. Invaplex functions as an intranasal adjuvant for subunit and DNA vaccines co-delivered in the nasal cavity of nonhuman primates. Vaccine X 2021; 8:100105. [PMID: 34258576 PMCID: PMC8255935 DOI: 10.1016/j.jvacx.2021.100105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/04/2021] [Accepted: 06/11/2021] [Indexed: 01/15/2023] Open
Abstract
Development of intranasal vaccines for HIV-1 and other mucosal pathogens has been hampered by the lack of adjuvants that can be given safely to humans. We have found that an intranasal Shigella vaccine (Invaplex) which is well tolerated in humans can also function as an adjuvant for intranasal protein and DNA vaccines in mice. To determine whether Invaplex could potentially adjuvant similar vaccines in humans, we simultaneously administered a simian immunodeficiency virus (SIV) envelope (Env) protein and DNA encoding simian-human immunodeficiency virus (SHIV) with or without Invaplex in the nasal cavity of female rhesus macaques. Animals were intranasally boosted with adenoviral vectors expressing SIV env or gag,pol to evaluate memory responses. Anti-SIV antibodies in sera and nasal, genital tract and rectal secretions were quantitated by ELISA. Intracellular cytokine staining was used to measure Th1-type T cells in blood. Macaques given DNA/protein immunizations with 0.5 mg Invaplex developed greater serum IgG, nasal IgA and cervicovaginal IgA responses to SIV Env and SHIV Gag,Pol proteins when compared to non-adjuvanted controls. Rectal IgA responses to Env were only briefly elevated and not observed to Gag,Pol. Invaplex increased frequencies of IFNγ-producing CD4 and CD8 T cells to the Env protein, but not T cell responses induced by the DNA. Ad-SIV boosting increased Env-specific polyfunctional T cells and Env- and Gag,Pol-specific antibodies in serum and all secretions. The data suggest that Invaplex could be highly effective as an adjuvant for intranasal protein vaccines in humans, especially those intended to prevent infections in the genital or respiratory tract.
Collapse
Key Words
- Ad, adenovirus
- CVS, cervicovaginal secretions
- Env, envelope
- HIV/AIDS
- ICS, intracellular cytokine staining
- IM, intramuscular
- IN, intranasal
- IgA
- Mucosal adjuvant
- NHP, nonhuman primates
- NS, nasal secretions
- RS, rectal secretions
- Reproductive
- Respiratory tract
- S-IgA, secretory IgA
- Th, T helper
Collapse
Affiliation(s)
- Jeremy V Camp
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Robert L Wilson
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Morgan Singletary
- Tulane National Primate Research Center, Division of Veterinary Medicine, Covington, LA 70433, USA
| | - James L Blanchard
- Tulane National Primate Research Center, Division of Veterinary Medicine, Covington, LA 70433, USA
| | - Anna Aldovini
- Departments of Medicine and Pediatrics, Children's Hospital and Harvard, Boston, MA 02115, USA
| | - Robert W Kaminski
- Department of Subunit Enteric Vaccines and Immunology, Division of Bacterial and Rickettsial Diseases, The Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Edwin V Oaks
- Department of Subunit Enteric Vaccines and Immunology, Division of Bacterial and Rickettsial Diseases, The Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
24
|
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
|
25
|
Swietnicki W. Secretory System Components as Potential Prophylactic Targets for Bacterial Pathogens. Biomolecules 2021; 11:892. [PMID: 34203937 PMCID: PMC8232601 DOI: 10.3390/biom11060892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 01/18/2023] Open
Abstract
Bacterial secretory systems are essential for virulence in human pathogens. The systems have become a target of alternative antibacterial strategies based on small molecules and antibodies. Strategies to use components of the systems to design prophylactics have been less publicized despite vaccines being the preferred solution to dealing with bacterial infections. In the current review, strategies to design vaccines against selected pathogens are presented and connected to the biology of the system. The examples are given for Y. pestis, S. enterica, B. anthracis, S. flexneri, and other human pathogens, and discussed in terms of effectiveness and long-term protection.
Collapse
Affiliation(s)
- Wieslaw Swietnicki
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. R. Weigla 12, 53-114 Wroclaw, Poland
| |
Collapse
|
26
|
Diks AM, Khatri I, Oosten LE, de Mooij B, Groenland RJ, Teodosio C, Perez-Andres M, Orfao A, Berbers GAM, Zwaginga JJ, van Dongen JJM, Berkowska MA. Highly Sensitive Flow Cytometry Allows Monitoring of Changes in Circulating Immune Cells in Blood After Tdap Booster Vaccination. Front Immunol 2021; 12:666953. [PMID: 34177905 PMCID: PMC8223751 DOI: 10.3389/fimmu.2021.666953] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022] Open
Abstract
Antigen-specific serum immunoglobulin (Ag-specific Ig) levels are broadly used as correlates of protection. However, in several disease and vaccination models these fail to predict immunity. In these models, in-depth knowledge of cellular processes associated with protective versus poor responses may bring added value. We applied high-throughput multicolor flow cytometry to track over-time changes in circulating immune cells in 10 individuals following pertussis booster vaccination (Tdap, Boostrix®, GlaxoSmithKline). Next, we applied correlation network analysis to extensively investigate how changes in individual cell populations correlate with each other and with Ag-specific Ig levels. We further determined the most informative cell subsets and analysis time points for future studies. Expansion and maturation of total IgG1 plasma cells, which peaked at day 7 post-vaccination, was the most prominent cellular change. Although these cells preceded the increase in Ag-specific serum Ig levels, they did not correlate with the increase of Ig levels. In contrast, strong correlation was observed between Ag-specific IgGs and maximum expansion of total IgG1 and IgA1 memory B cells at days 7 to 28. Changes in circulating T cells were limited, implying the need for a more sensitive approach. Early changes in innate immune cells, i.e. expansion of neutrophils, and expansion and maturation of monocytes up to day 5, most likely reflected their responses to local damage and adjuvant. Here we show that simultaneous monitoring of multiple circulating immune subsets in blood by flow cytometry is feasible. B cells seem to be the best candidates for vaccine monitoring.
Collapse
Affiliation(s)
- Annieck M. Diks
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Indu Khatri
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, Netherlands
| | | | - Bas de Mooij
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Rick J. Groenland
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Cristina Teodosio
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Martin Perez-Andres
- Cancer Research Centre (IBMCC, USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Alberto Orfao
- Cancer Research Centre (IBMCC, USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Guy A. M. Berbers
- Center for Infectious Disease Control, National Institute of Public Health and the Environment, Bilthoven, Netherlands
| | - Jaap Jan Zwaginga
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | | |
Collapse
|
27
|
Yang R, Deng Y, Huang B, Huang L, Lin A, Li Y, Wang W, Liu J, Lu S, Zhan Z, Wang Y, A R, Wang W, Niu P, Zhao L, Li S, Ma X, Zhang L, Zhang Y, Yao W, Liang X, Zhao J, Liu Z, Peng X, Li H, Tan W. A core-shell structured COVID-19 mRNA vaccine with favorable biodistribution pattern and promising immunity. Signal Transduct Target Ther 2021; 6:213. [PMID: 34059617 PMCID: PMC8165147 DOI: 10.1038/s41392-021-00634-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/17/2021] [Accepted: 05/05/2021] [Indexed: 12/28/2022] Open
Abstract
Although inoculation of COVID-19 vaccines has rolled out globally, there is still a critical need for safe and effective vaccines to ensure fair and equitable supply for all countries. Here, we report on the development of a highly efficacious mRNA vaccine, SW0123 that is composed of sequence-modified mRNA encoding the full-length SARS-CoV-2 Spike protein packaged in core-shell structured lipopolyplex (LPP) nanoparticles. SW0123 is easy to produce using a large-scale microfluidics-based apparatus. The unique core-shell structured nanoparticle facilitates vaccine uptake and demonstrates a high colloidal stability, and a desirable biodistribution pattern with low liver targeting effect upon intramuscular administration. Extensive evaluations in mice and nonhuman primates revealed strong immunogenicity of SW0123, represented by induction of Th1-polarized T cell responses and high levels of antibodies that were capable of neutralizing not only the wild-type SARS-CoV-2, but also a panel of variants including D614G and N501Y variants. In addition, SW0123 conferred effective protection in both mice and non-human primates upon SARS-CoV-2 challenge. Taken together, SW0123 is a promising vaccine candidate that holds prospects for further evaluation in humans.
Collapse
Affiliation(s)
- Ren Yang
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yao Deng
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Baoying Huang
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lei Huang
- Stemirna Therapeutics, Shanghai, China
| | - Ang Lin
- Stemirna Therapeutics, Shanghai, China
| | - Yuhua Li
- National Institute for Food and Drug Control, Beijing, China
| | - Wenling Wang
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jingjing Liu
- National Institute for Food and Drug Control, Beijing, China
| | - Shuaiyao Lu
- National Kunming High-level Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China
| | - Zhenzhen Zhan
- Shanghai East Hospital, Tongji University, Shanghai, China
| | - Yufei Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences and National Center for Nanoscience and Technology of China, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ruhan A
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wen Wang
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Peihua Niu
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Li Zhao
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | | | | | | | | | | | - Xingjie Liang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences and National Center for Nanoscience and Technology of China, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhongmin Liu
- Shanghai East Hospital, Tongji University, Shanghai, China
| | - Xiaozhong Peng
- National Kunming High-level Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Yunnan, China.
| | | | - Wenjie Tan
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China.
| |
Collapse
|
28
|
Solans L, Debrie AS, Coutte L, Locht C. Construction and evaluation of a pertactin-deficient live attenuated pertussis vaccine candidate BPZE1 derivative. Vaccine 2021; 39:2843-2849. [PMID: 33896662 DOI: 10.1016/j.vaccine.2021.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/22/2021] [Accepted: 04/08/2021] [Indexed: 10/21/2022]
Abstract
Pertussis, mainly caused by Bordetella pertussis, is a severe respiratory disease that can be fatal, especially in young infants. Vaccines, massively implemented since the middle of the last century, have substantially reduced the pertussis incidence, but have not been able to fully control the disease. One of the shortcomings of current pertussis vaccines is their inability to prevent infection by and transmission of B. pertussis, in contrast to immunity following natural infection. We have developed the live attenuated nasal vaccine BPZE1 and have shown that it prevents both disease and B. pertussis infection in preclinical models. This vaccine is now in clinical development. However, the initial clinical studies have suggested that vaccine take is hampered by pre-existing antibodies to pertactin. Here, we have constructed a pertactin-deficient BPZE1 derivative called BPZE1P in order to overcome this limitation. BPZE1P colonized the murine respiratory tract as efficiently as BPZE1 and induced antibodies at levels similar to those elicited by BPZE1. In the presence of pre-existing antibodies induced by acellular pertussis vaccination, BPZE1P colonized the mouse respiratory tract more efficiently than BPZE1. Both vaccines protected equally well the murine lungs and noses from challenge with laboratory and clinical strains of B. pertussis, including pertactin-deficient strains, against which current acellular pertussis vaccines are less efficient. BPZE1P may thus be an interesting alternative to BPZE1 to overcome vaccine take limitations due to pre-existing antibodies to pertactin.
Collapse
Affiliation(s)
- Luis Solans
- 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
| | - Anne-Sophie Debrie
- 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
| | - Loïc Coutte
- 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
| | - 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
|
29
|
Abstract
INTRODUCTION Pertussis, caused by Bordetella pertussis, remains a major public health problem, despite high vaccination coverage. Furthermore, the disease incidence has increased recently, especially in countries that have switched from whole-cell to acellular pertussis vaccines. AREAS COVERED Here, we provide a state-of-the art summary of the reasons for the pertussis resurgence and discuss potential solutions using current vaccines and challenges for the development of novel vaccines. PubMed was searched for publications with the terms pertussis and vaccines. Many new vaccine candidates are proposed but most have not reached clinical development. Most of them induce strong systemic immune responses and protection in mice. However, since B. pertussis is a mucosal pathogen, albeit with systemic effects, local immunity may be crucial to prevent B. pertussis infection and transmission. Recent efforts have focused on vaccine candidates able to induce immunity in the nasal cavity, and one of them is currently in clinical development. EXPERT COMMENTARY New pertussis vaccines are needed to durably control the disease and circulation of B. pertussis. A major challenge is to prove efficacy against disease in randomized controlled trials, while it is feasible to provide evidence for prevention of infection, since asymptomatic carriage of B. pertussis is wide spread.
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, Lille, France
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
Jahnmatz M, Richert L, al-Tawil N, Storsaeter J, Colin C, Bauduin C, Thalen M, Solovay K, Rubin K, Mielcarek N, Thorstensson R, Locht C, Dager L, Ekholm N, Gustafsson M, Linde Å, Lång C, Nastase M, Reinholdsson IL, Sigurdardottir E, Wahlberg A, Zarea I, Aktas T, Andersson I, Hanson Pihlainen E, Ljungman M, Ringman M, Tecleab T, Wehlin L, Allais F, Assuied A, Chêne G, Gilbert C, Jean D, Le Marec F, Moinot L, Reboud P, Rousseau E, Roy C, Schwimmer C, Taïeb L, Wallet C, Derocle G, Gueguen S, Lévy-Marchal C, Esperou H, Debrie AS, Raze D, Coutte L, Diallo A, Mercier N. Safety and immunogenicity of the live attenuated intranasal pertussis vaccine BPZE1: a phase 1b, double-blind, randomised, placebo-controlled dose-escalation study. THE LANCET. INFECTIOUS DISEASES 2020; 20:1290-1301. [DOI: 10.1016/s1473-3099(20)30274-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/10/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022]
|
33
|
Chasaide CN, Mills KH. Next-Generation Pertussis Vaccines Based on the Induction of Protective T Cells in the Respiratory Tract. Vaccines (Basel) 2020; 8:E621. [PMID: 33096737 PMCID: PMC7711671 DOI: 10.3390/vaccines8040621] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022] Open
Abstract
Immunization with current acellular pertussis (aP) vaccines protects against severe pertussis, but immunity wanes rapidly after vaccination and these vaccines do not prevent nasal colonization with Bordetella pertussis. Studies in mouse and baboon models have demonstrated that Th1 and Th17 responses are integral to protective immunity induced by previous infection with B. pertussis and immunization with whole cell pertussis (wP) vaccines. Mucosal Th17 cells, IL-17 and secretory IgA (sIgA) are particularly important in generating sustained sterilizing immunity in the nasal cavity. Current aP vaccines induce potent IgG and Th2-skewed T cell responses but are less effective at generating Th1 and Th17 responses and fail to prime respiratory tissue-resident memory T (TRM) cells, that maintain long-term immunity at mucosal sites. In contrast, a live attenuated pertussis vaccine, pertussis outer membrane vesicle (OMV) vaccines or aP vaccines formulated with novel adjuvants do induce cellular immune responses in the respiratory tract, especially when delivered by the intranasal route. An increased understanding of the mechanisms of sustained protective immunity, especially the role of respiratory TRM cells, will facilitate the development of next generation pertussis vaccines that not only protect against pertussis disease, but prevent nasal colonization and transmission of B. pertussis.
Collapse
Affiliation(s)
| | - Kingston H.G. Mills
- School of Biochemistry and Immunology, Trinity College Dublin, 2, D02 PN40 Dublin, Ireland;
| |
Collapse
|
34
|
Damron FH, Barbier M, Dubey P, Edwards KM, Gu XX, Klein NP, Lu K, Mills KHG, Pasetti MF, Read RC, Rohani P, Sebo P, Harvill ET. Overcoming Waning Immunity in Pertussis Vaccines: Workshop of the National Institute of Allergy and Infectious Diseases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:877-882. [PMID: 32769142 PMCID: PMC7454230 DOI: 10.4049/jimmunol.2000676] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 01/21/2023]
Abstract
Despite high vaccine coverage in many parts of the world, pertussis is resurging in a number of areas in which acellular vaccines are the primary vaccine administered to infants and young children. This is attributed in part to the suboptimal and short-lived immunity elicited by acellular pertussis vaccines and to their inability to prevent nasal colonization and transmission of the etiologic agent Bordetella pertussis In response to this escalating public health concern, the National Institute of Allergy and Infectious Diseases held the workshop "Overcoming Waning Immunity in Pertussis Vaccines" in September 2019 to identify issues and possible solutions for the defects in immunity stimulated by acellular pertussis vaccines. Discussions covered aspects of the current problem, gaps in knowledge and possible paths forward. This review summarizes presentations and discussions of some of the key points that were raised by the workshop.
Collapse
Affiliation(s)
- F Heath Damron
- Department of Microbiology, Immunology, and Cell Biology and Vaccine Development Center at West Virginia University Health Sciences Center, West Virginia University, Morgantown, WV 26506
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology and Vaccine Development Center at West Virginia University Health Sciences Center, West Virginia University, Morgantown, WV 26506
| | - Purnima Dubey
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| | - Kathryn M Edwards
- Division of Infectious Diseases, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Xin-Xing Gu
- Division of Microbiology and Infectious Diseases, National Institutes of Health, Washington, D.C. 20852
| | - Nicola P Klein
- Kaiser Permanente Vaccine Study Center, Kaiser Permanente Northern California, Oakland, CA 94611
| | - Kristina Lu
- Division of Microbiology and Infectious Diseases, National Institutes of Health, Washington, D.C. 20852
| | - Kingston H G Mills
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin D02 PN40, Ireland
| | - Marcela F Pasetti
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Robert C Read
- National Institute for Health Research Southampton Biomedical Research Centre, University Hospital Southampton and Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Pejman Rohani
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
- Odum School of Ecology, University of Georgia, Athens, GA 30602; and
| | - Peter Sebo
- Institute of Microbiology of the Czech Academy of Sciences, 142 20 Prague 4, Czech Republic
| | - Eric T Harvill
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602;
| |
Collapse
|
35
|
Boyle M. Livening up pertussis vaccine development. Sci Transl Med 2020. [DOI: 10.1126/scitranslmed.aba9014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Vaccination with a
Bordetella pertussis
live attenuated vaccine induces a beneficial T
H
1 cell bias and broadly targeting functional antibodies in humans.
Collapse
Affiliation(s)
- Michelle Boyle
- Immunology Department, QIMR-Berghofer, Herston, Queensland 4006, Australia
| |
Collapse
|