1
|
Wanlapakorn N, Sarawanangkoor N, Srimuan D, Thatsanathorn T, Thongmee T, Poovorawan Y. Antibody persistence to diphtheria toxoid, tetanus toxoid, Bordetella pertussis antigens, and Haemophilus influenzae type b following primary and first booster with pentavalent versus hexavalent vaccines. Hum Vaccin Immunother 2024; 20:2352909. [PMID: 38752802 DOI: 10.1080/21645515.2024.2352909] [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: 03/11/2024] [Accepted: 05/05/2024] [Indexed: 07/18/2024] Open
Abstract
Thailand has incorporated the whole-cell (wP) pertussis vaccine into the expanded program on immunization since 1977 and has offered the acellular pertussis (aP) vaccine as an optional vaccine for infants since 2001. We followed healthy children from a clinical trial (ClinicalTrials.gov NCT02408926) in which children were randomly assigned to receive either pentavalent (DTwP-HB-Hib) or hexavalent (DTaP-IPV-HB-Hib) vaccines for their primary series (administered at 2, 4, and 6 months) and first booster vaccination (18 months). Both groups received Tdap-IPV as a second booster at the age of 4 y. Blood samples were collected for evaluation of antibody persistence to diphtheria toxoid (DT), tetanus toxoid (TT), and Bordetella pertussis (B. pertussis) between 2 and 6 y of age annually, and for the immunogenicity study of Tdap-IPV at 1 month after the second booster. Antibody persistence to Haemophilus influenzae type b (Hib) was followed until 3 y of age. A total of 105 hexavalent-vaccinated children and 91 pentavalent-vaccinated children completed this study. Both pentavalent and hexavalent groups demonstrated increased antibody levels against DT, TT, and B. pertussis antigens following the second booster with Tdap-IPV. All children achieved a seroprotective concentration for anti-DT and anti-TT IgG at 1 month post booster. The hexavalent group possessed significantly higher anti-pertactin IgG (adjusted p = .023), whereas the pentavalent group possessed significantly higher anti-pertussis toxin IgG (adjusted p < .001) after the second booster. Despite declining levels post-second booster, a greater number of children sustained protective levels of anti-DT and anti-TT IgG compared to those after the first booster.
Collapse
Affiliation(s)
- Nasamon Wanlapakorn
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nasiri Sarawanangkoor
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Donchida Srimuan
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thaksaporn Thatsanathorn
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thanunrat Thongmee
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| |
Collapse
|
2
|
Shapiro JR, Corrado M, Perry J, Watts TH, Bolotin S. The contributions of T cell-mediated immunity to protection from vaccine-preventable diseases: A primer. Hum Vaccin Immunother 2024; 20:2395679. [PMID: 39205626 PMCID: PMC11364080 DOI: 10.1080/21645515.2024.2395679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
In the face of the ever-present burden of emerging and reemerging infectious diseases, there is a growing need to comprehensively assess individual- and population-level immunity to vaccine-preventable diseases (VPDs). Many of these efforts, however, focus exclusively on antibody-mediated immunity, ignoring the role of T cells. Aimed at clinicians, public health practioners, and others who play central roles in human vaccine research but do not have formal training in immunology, we review how vaccines against infectious diseases elicit T cell responses, what types of vaccines elicit T cell responses, and how T cell responses are measured. We then use examples to demonstrate six ways that T cells contribute to protection from VPD, including directly mediating protection, enabling antibody responses, reducing disease severity, increasing cross-reactivity, improving durability, and protecting special populations. We conclude with a discussion of challenges and solutions to more widespread consideration of T cell responses in clinical vaccinology.
Collapse
Affiliation(s)
- Janna R. Shapiro
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Mario Corrado
- Division of General Internal Medicine, University of Toronto, Toronto, ON, Canada
| | - Julie Perry
- Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Tania H. Watts
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Shelly Bolotin
- Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Health Protection, Public Health Ontario, Toronto, ON, Canada
| |
Collapse
|
3
|
Domenech de Cellès M, Rohani P. Pertussis vaccines, epidemiology and evolution. Nat Rev Microbiol 2024; 22:722-735. [PMID: 38907021 DOI: 10.1038/s41579-024-01064-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2024] [Indexed: 06/23/2024]
Abstract
Pertussis, which is caused by Bordetella pertussis, has plagued humans for at least 800 years, is highly infectious and can be fatal in the unvaccinated, especially very young infants. Although the rollout of whole-cell pertussis (wP) vaccines in the 1940s and 1950s was associated with a drastic drop in incidence, concerns regarding the reactogenicity of wP vaccines led to the development of a new generation of safer, acellular (aP) vaccines that have been adopted mainly in high-income countries. Over the past 20 years, some countries that boast high aP coverage have experienced a resurgence in pertussis, which has led to substantial debate over the basic immunology, epidemiology and evolutionary biology of the bacterium. Controversy surrounds the duration of natural immunity and vaccine-derived immunity, the ability of vaccines to prevent transmission and severe disease, and the impact of evolution on evading vaccine immunity. Resolving these issues is made challenging by incomplete detection of pertussis cases, the absence of a serological marker of immunity, modest sequencing of the bacterial genome and heterogeneity in diagnostic methods of surveillance. In this Review, we lay out the complexities of contemporary pertussis and, where possible, propose a parsimonious explanation for apparently incongruous observations.
Collapse
Affiliation(s)
| | - Pejman Rohani
- Odum School of Ecology, University of Georgia, Athens, GA, USA.
- Center of Ecology of Infectious Diseases, Athens, GA, USA.
- Department of Infectious Diseases, College for Veterinary Medicine, University of Georgia, Athens, GA, USA.
| |
Collapse
|
4
|
Willemsen L, Lee J, Shinde P, Soldevila F, Aoki M, Orfield S, Kojima M, da Silva Antunes R, Sette A, Peters B. Th1 polarization in Bordetella pertussis vaccine responses is maintained through a positive feedback loop. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606623. [PMID: 39149302 PMCID: PMC11326151 DOI: 10.1101/2024.08.05.606623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Outbreaks of Bordetella pertussis (BP), the causative agent of whooping cough, continue despite broad vaccination coverage and have been increasing since vaccination switched from whole-BP (wP) to acellular BP (aP) vaccines. wP vaccination has been associated with more durable protective immunity and an induced Th1 polarized memory T cell response. Here, a multi-omics approach was applied to profile the immune response of 30 wP and 31 aP-primed individuals and identify correlates of T cell polarization before and after Tdap booster vaccination. We found that transcriptional changes indicating an interferon response on day 1 post-booster along with elevated plasma concentrations of IFN-γ and interferon-induced chemokines that peaked at day 1-3 post-booster correlated best with the Th1 polarization of the vaccine-induced memory T cell response on day 28. Our studies suggest that wP-primed individuals maintain their Th1 polarization through this early memory interferon response. This suggests that stimulating the interferon pathway during vaccination could be an effective strategy to elicit a predominant Th1 response in aP-primed individuals that protects better against infection.
Collapse
|
5
|
Da Costa RM, Rooke JL, Wells TJ, Cunningham AF, Henderson IR. Type 5 secretion system antigens as vaccines against Gram-negative bacterial infections. NPJ Vaccines 2024; 9:159. [PMID: 39218947 PMCID: PMC11366766 DOI: 10.1038/s41541-024-00953-6] [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: 02/13/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Infections caused by Gram-negative bacteria are leading causes of mortality worldwide. Due to the rise in antibiotic resistant strains, there is a desperate need for alternative strategies to control infections caused by these organisms. One such approach is the prevention of infection through vaccination. While live attenuated and heat-killed bacterial vaccines are effective, they can lead to adverse reactions. Newer vaccine technologies focus on utilizing polysaccharide or protein subunits for safer and more targeted vaccination approaches. One promising avenue in this regard is the use of proteins released by the Type 5 secretion system (T5SS). This system is the most prevalent secretion system in Gram-negative bacteria. These proteins are compelling vaccine candidates due to their demonstrated protective role in current licensed vaccines. Notably, Pertactin, FHA, and NadA are integral components of licensed vaccines designed to prevent infections caused by Bordetella pertussis or Neisseria meningitidis. In this review, we delve into the significance of incorporating T5SS proteins into licensed vaccines, their contributions to virulence, conserved structural motifs, and the protective immune responses elicited by these proteins.
Collapse
Affiliation(s)
- Rochelle M Da Costa
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Jessica L Rooke
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Timothy J Wells
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Adam F Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Ian R Henderson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
6
|
Lavelle EC, McEntee CP. Vaccine adjuvants: Tailoring innate recognition to send the right message. Immunity 2024; 57:772-789. [PMID: 38599170 DOI: 10.1016/j.immuni.2024.03.015] [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: 01/19/2024] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
Adjuvants play pivotal roles in vaccine development, enhancing immunization efficacy through prolonged retention and sustained release of antigen, lymph node targeting, and regulation of dendritic cell activation. Adjuvant-induced activation of innate immunity is achieved via diverse mechanisms: for example, adjuvants can serve as direct ligands for pathogen recognition receptors or as inducers of cell stress and death, leading to the release of immunostimulatory-damage-associated molecular patterns. Adjuvant systems increasingly stimulate multiple innate pathways to induce greater potency. Increased understanding of the principles dictating adjuvant-induced innate immunity will subsequently lead to programming specific types of adaptive immune responses. This tailored optimization is fundamental to next-generation vaccines capable of inducing robust and sustained adaptive immune memory across different cohorts.
Collapse
Affiliation(s)
- Ed C Lavelle
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| | - Craig P McEntee
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
7
|
Shinde P, Soldevila F, Reyna J, Aoki M, Rasmussen M, Willemsen L, Kojima M, Ha B, Greenbaum JA, Overton JA, Guzman-Orozco H, Nili S, Orfield S, Gygi JP, da Silva Antunes R, Sette A, Grant B, Olsen LR, Konstorum A, Guan L, Ay F, Kleinstein SH, Peters B. A multi-omics systems vaccinology resource to develop and test computational models of immunity. CELL REPORTS METHODS 2024; 4:100731. [PMID: 38490204 PMCID: PMC10985234 DOI: 10.1016/j.crmeth.2024.100731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/04/2024] [Accepted: 02/20/2024] [Indexed: 03/17/2024]
Abstract
Systems vaccinology studies have identified factors affecting individual vaccine responses, but comparing these findings is challenging due to varying study designs. To address this lack of reproducibility, we established a community resource for comparing Bordetella pertussis booster responses and to host annual contests for predicting patients' vaccination outcomes. We report here on our experiences with the "dry-run" prediction contest. We found that, among 20+ models adopted from the literature, the most successful model predicting vaccination outcome was based on age alone. This confirms our concerns about the reproducibility of conclusions between different vaccinology studies. Further, we found that, for newly trained models, handling of baseline information on the target variables was crucial. Overall, multiple co-inertia analysis gave the best results of the tested modeling approaches. Our goal is to engage community in these prediction challenges by making data and models available and opening a public contest in August 2024.
Collapse
Affiliation(s)
- Pramod Shinde
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Ferran Soldevila
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Joaquin Reyna
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA; Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, San Diego, CA, USA
| | - Minori Aoki
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Mikkel Rasmussen
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lisa Willemsen
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Mari Kojima
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Brendan Ha
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Jason A Greenbaum
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - James A Overton
- Knocean Inc., 107 Quebec Avenue, Toronto, Ontario M6P 2T3, Canada
| | - Hector Guzman-Orozco
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Somayeh Nili
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Shelby Orfield
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Jeremy P Gygi
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA
| | - Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Barry Grant
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Lars Rønn Olsen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Anna Konstorum
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Leying Guan
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Ferhat Ay
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Steven H Kleinstein
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Medicine, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
8
|
Rubio-Casillas A, Rodriguez-Quintero CM, Redwan EM, Gupta MN, Uversky VN, Raszek M. Do vaccines increase or decrease susceptibility to diseases other than those they protect against? Vaccine 2024; 42:426-440. [PMID: 38158298 DOI: 10.1016/j.vaccine.2023.12.060] [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: 08/29/2023] [Revised: 11/16/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
Contrary to the long-held belief that the effects of vaccines are specific for the disease they were created; compelling evidence has demonstrated that vaccines can exert positive or deleterious non-specific effects (NSEs). In this review, we compiled research reports from the last 40 years, which were found based on the PubMed search for the epidemiological and immunological studies on the non-specific effects (NSEs) of the most common human vaccines. Analysis of information showed that live vaccines induce positive NSEs, whereas non-live vaccines induce several negative NSEs, including increased female mortality associated with enhanced susceptibility to other infectious diseases, especially in developing countries. These negative NSEs are determined by the vaccination sequence, the antigen concentration in vaccines, the type of vaccine used (live vs. non-live), and also by repeated vaccination. We do not recommend stopping using non-live vaccines, as they have demonstrated to protect against their target disease, so the suggestion is that their detrimental NSEs can be minimized simply by changing the current vaccination sequence. High IgG4 antibody levels generated in response to repeated inoculation with mRNA COVID-19 vaccines could be associated with a higher mortality rate from unrelated diseases and infections by suppressing the immune system. Since most COVID-19 vaccinated countries are reporting high percentages of excess mortality not directly attributable to deaths from such disease, the NSEs of mRNA vaccines on overall mortality should be studied in depth.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan 48900, Jalisco, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan 48900, Jalisco, Mexico.
| | | | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria 21934, Egypt.
| | - Munishwar Nath Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Mikolaj Raszek
- Merogenomics (Genomic Sequencing Consulting), Edmonton, AB T5J 3R8, Canada.
| |
Collapse
|
9
|
Kang KR, Kim JA, Cho GW, Kang HU, Kang HM, Kang JH, Seong BL, Lee SY. Comparative Evaluation of Recombinant and Acellular Pertussis Vaccines in a Murine Model. Vaccines (Basel) 2024; 12:108. [PMID: 38276680 PMCID: PMC10818713 DOI: 10.3390/vaccines12010108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Since the 2000s, sporadic outbreaks of whooping cough have been reported in advanced countries, where the acellular pertussis vaccination rate is relatively high, and in developing countries. Small-scale whooping cough has also continued in many countries, due in part to the waning of immune protection after childhood vaccination, necessitating the development of an improved pertussis vaccine and vaccination program. Currently, two different production platforms are being actively pursued in Korea; one is based on the aP (acellular pertussis) vaccine purified from B. pertussis containing pertussis toxoid (PT), filamentous hemagglutin (FHA) and pertactin (PRN), and the other is based on the recombinant aP (raP), containing genetically detoxified pertussis toxin ADP-ribosyltransferase subunit 1 (PtxS1), FHA, and PRN domain, expressed and purified from recombinant E. coli. aP components were further combined with diphtheria and tetanus vaccine components as a prototype DTaP vaccine by GC Pharma (GC DTaP vaccine). We evaluated and compared the immunogenicity and the protective efficacy of aP and raP vaccines in an experimental murine challenge model: humoral immunity in serum, IgA secretion in nasal lavage, bacterial clearance after challenge, PTx (pertussis toxin) CHO cell neutralization titer, cytokine secretion in spleen single cell, and tissue resident memory CD4+ T cell (CD4+ TRM cell) in lung tissues. In humoral immunogenicity, GC DTaP vaccines showed high titers for PT and PRN and showed similar patterns in nasal lavage and IL-5 cytokine secretions. The GC DTaP vaccine and the control vaccine showed equivalent results in bacterial clearance after challenge, PTx CHO cell neutralization assay, and CD4+ TRM cell. In contrast, the recombinant raP vaccine exhibited strong antibody responses for FHA and PRN, albeit with low antibody level of PT and low titer in PTx CHO neutralization assay, as compared to control and GC DTaP vaccines. The raP vaccine provided a sterile lung bacterial clearance comparable to a commercial control vaccine after the experimental challenge in murine model. Moreover, raP exhibited a strong cytokine response and CD4+ TRM cell in lung tissue, comparable or superior to the experimental and commercial DTaP vaccinated groups. Contingent on improving the biophysical stability and humoral response to PT, the raP vaccine warrants further development as an effective alternative to aP vaccines for the control of a pertussis outbreak.
Collapse
Affiliation(s)
- Kyu-Ri Kang
- The Vaccine Bio Research Institute, Annex to Seoul Saint Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea (J.-H.K.)
| | - Ji-Ahn Kim
- The Vaccine Bio Research Institute, Annex to Seoul Saint Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea (J.-H.K.)
| | - Gyu-Won Cho
- The Vaccine Bio Research Institute, Annex to Seoul Saint Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea (J.-H.K.)
| | - Han-Ul Kang
- The Interdisciplinary Graduate Program in Integrative Biotechnology, Yonsei University, Incheon 21983, Republic of Korea
| | - Hyun-Mi Kang
- The Vaccine Bio Research Institute, Annex to Seoul Saint Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea (J.-H.K.)
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin-Han Kang
- The Vaccine Bio Research Institute, Annex to Seoul Saint Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea (J.-H.K.)
| | - Baik-Lin Seong
- Department of Microbiology and Immunology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Soo-Young Lee
- The Vaccine Bio Research Institute, Annex to Seoul Saint Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea (J.-H.K.)
- Department of Pediatrics, Bucheon St. Mary’s Hospital, The Catholic University of Korea, Bucheon 14647, Republic of Korea
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Olivera I, Pérez CG, Lazarov L, Lopez E, Oddo C, Dibarboure H. Cost minimization analysis of a hexavalent vaccine in Argentina. BMC Health Serv Res 2023; 23:1067. [PMID: 37803345 PMCID: PMC10557326 DOI: 10.1186/s12913-023-10038-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 09/15/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Argentina currently uses a pentavalent vaccine containing diphtheria, tetanus, pertussis (whole cell), Haemophilus influenza type b and hepatitis B antigens, administered concomitantly with the inactivated polio vaccine (IPV) (DTwP-Hib-HB plus IPV) in its childhood vaccination schedule. However, hexavalent vaccines containing acellular pertussis antigens (DTaP-Hib-HB-IPV) and providing protection against the same diseases are also licensed, but are only available with a private prescription or for high-risk pre-term infants in the public health program. We analyzed the cost of switching from the current schedule to the alternative schedule with the hexavalent vaccine in Argentina, assuming similar levels of effectiveness. METHODS The study population was infants ≤ 1 year of age born in Argentina from 2015 to 2019. The analysis considered adverse events, programmatic, logistic, and vaccine costs of both schemes from the societal perspective. The societal costs were disaggregated to summarize costs incurred in the public sector and with vaccination pre-term infants in the public sector. Costs were expressed in 2021 US Dollars (US$). RESULTS Although the cost of vaccines with the alternative scheme would be US$39.8 million (M) more than with the current scheme, these additional costs are in large part offset by fewer adverse event-associated costs and lower programmatic costs such that the overall cost of the alternative scheme would only be an additional US$3.6 M from the societal perspective. The additional cost associated with switching to the alternative scheme in the public sector and with the vaccination of pre-term infants in the public sector would be US$2.1 M and US$84,023, respectively. CONCLUSIONS The switch to an alternative scheme with the hexavalent vaccine in Argentina would result in marginally higher vaccine costs, which are mostly offset by the lower costs associated with improved logistics, fewer separate vaccines, and a reduction in adverse events.
Collapse
Affiliation(s)
- Ignacio Olivera
- Centro de Investigaciones Económicas, CINVE, Salud, Montevideo, Uruguay
| | - Carlos Grau Pérez
- Centro de Investigaciones Económicas, CINVE, Salud, Montevideo, Uruguay
| | - Luis Lazarov
- Centro de Investigaciones Económicas, CINVE, Salud, Montevideo, Uruguay
| | - Eduardo Lopez
- Facultad de Medicina, Universidad del Salvador, Buenos Aires, Argentina
| | - Cristian Oddo
- Sanofi Vaccines, Buenos Aires, Argentina.
- , Cuyo 3512, Martinez, Buenos Aires, B1640GIY, Argentina.
| | | |
Collapse
|
13
|
Dubois V, Chatagnon J, Depessemier M, Locht C. Maternal acellular pertussis vaccination in mice impairs cellular immunity to Bordetella pertussis infection in offspring. JCI Insight 2023; 8:e167210. [PMID: 37581930 PMCID: PMC10561720 DOI: 10.1172/jci.insight.167210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 08/08/2023] [Indexed: 08/17/2023] Open
Abstract
Given the resurgence of pertussis, several countries have introduced maternal tetanus, diphtheria, and acellular pertussis (aP) vaccination during pregnancy to protect young infants against severe pertussis. Although protective against the disease, the effect of maternal aP vaccination on bacterial colonization of the offspring is unknown. Here, we used a mouse model to demonstrate that maternal aP immunization, either before or during pregnancy, protects pups from lung colonization by Bordetella pertussis. However, maternal aP vaccination resulted in significantly prolonged nasal carriage of B. pertussis by inhibiting the natural recruitment of IL-17-producing resident memory T cells and ensuing neutrophil influx in the nasal tissue, especially of those with proinflammatory and cytotoxic properties. Prolonged nasal carriage after aP vaccination is due to IL-4 signaling, as prolonged nasal carriage is abolished in IL-4Rα-/- mice. The effect of maternal aP vaccination can be transferred transplacentally to the offspring or via breastfeeding and is long-lasting, as it persists into adulthood. Maternal aP vaccination may, thus, augment the B. pertussis reservoir.
Collapse
|
14
|
Thorat S, Ogale P, Gautam M, Shaligram U, Gairola S. Development and validation of capillary electrophoresis sodium dodecyl sulfate (CE-SDS) method for purity analysis of pertussis toxin, filamentous haemagglutinin and pertactin antigens. Vaccine 2023; 41:5854-5862. [PMID: 37591705 DOI: 10.1016/j.vaccine.2023.08.010] [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: 03/08/2023] [Revised: 07/18/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
We report here the development and validation of CE-SDS method for purity analysis of Acellular Pertussis vaccine components viz. purified Pertussis toxin (PTx), purified Filamentous haemagglutinin (FHA), and Pertactinantigen (PRN). The method was found to be specific and showed excellent linearity at a concentration range of 15.62 µg/mL-1000 µg/mL for purified PTx, 31.25 µg/mL-1000 µg/mL for purified FHA, and 3.9 µg/mL-1000 µg/mL for PRN antigen. Method reported limit of quantification (LOQ) 31.25 µg/mL, 62.5 µg/mL, and 7.8 µg/mL for purified PTx, FHA, and PRN respectively. Method precision (repeatability and intermediate precision) for purity and molecular weight determination in product matrix was below 10% for all three proteins. Method comparability studies were performed with SDS-PAGE. CE-SDS demonstrated corroborating results with SDS-PAGE for the estimation of purity and molecular weight analysis. However, CE-SDS method exhibited better resolution capabilities for resolving all the sub-unit peaks of PTx and isoforms of purified FHA. CE-SDS method also demonstrated stability indicating potential and thus fits its intended purpose as an effective analytical tool for quality control of acellular pertussis-based vaccines.
Collapse
Affiliation(s)
- Shrikant Thorat
- Serum Institute of India Pvt. Ltd, Hadapsar, Pune 411 028, Maharashtra, India
| | - Pratik Ogale
- Serum Institute of India Pvt. Ltd, Hadapsar, Pune 411 028, Maharashtra, India
| | - Manish Gautam
- Serum Institute of India Pvt. Ltd, Hadapsar, Pune 411 028, Maharashtra, India
| | - Umesh Shaligram
- Serum Institute of India Pvt. Ltd, Hadapsar, Pune 411 028, Maharashtra, India
| | - Sunil Gairola
- Serum Institute of India Pvt. Ltd, Hadapsar, Pune 411 028, Maharashtra, India.
| |
Collapse
|
15
|
da Silva Antunes R, Garrigan E, Quiambao LG, Dhanda SK, Marrama D, Westernberg L, Wang E, Abawi A, Sutherland A, Armstrong SK, Brickman TJ, Sidney J, Frazier A, Merkel TJ, Peters B, Sette A. T cell reactivity to Bordetella pertussis is highly diverse regardless of childhood vaccination. Cell Host Microbe 2023; 31:1404-1416.e4. [PMID: 37490913 PMCID: PMC10528758 DOI: 10.1016/j.chom.2023.06.015] [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: 03/27/2023] [Revised: 05/17/2023] [Accepted: 06/27/2023] [Indexed: 07/27/2023]
Abstract
The incidence of whooping cough due to Bordetella pertussis (BP) infections has increased recently. It is believed that the shift from whole-cell pertussis (wP) vaccines to acellular pertussis (aP) vaccines may be contributing to this rise. While T cells are key in controlling and preventing disease, nearly all knowledge relates to antigens in aP vaccines. A whole-genome mapping of human BP-specific CD4+ T cell responses was performed in healthy vaccinated adults and revealed unexpected broad reactivity to hundreds of antigens. The overall pattern and magnitude of T cell responses to aP and non-aP vaccine antigens are similar regardless of childhood vaccination, suggesting that asymptomatic infections drive the pattern of T cell reactivity in adults. Lastly, lack of Th1/Th2 polarization to non-aP vaccine antigens suggests these antigens have the potential to counteract aP vaccination Th2 bias. These findings enhance our insights into human T cell responses to BP and identify potential targets for next-generation pertussis vaccines.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA.
| | - Emily Garrigan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Lorenzo G Quiambao
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Sandeep Kumar Dhanda
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Daniel Marrama
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Luise Westernberg
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Eric Wang
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Adam Abawi
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Aaron Sutherland
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Sandra K Armstrong
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Timothy J Brickman
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - April Frazier
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Tod J Merkel
- Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| |
Collapse
|
16
|
Wang W, Wang Z, Chen Q, Li M, Jia C, Xu Y, Wu Y, Sun X, Sun H. The Decay of Pertussis Antibodies in Children Aged 0-14 Years in Jiangsu Province, China. Vaccines (Basel) 2023; 11:1336. [PMID: 37631904 PMCID: PMC10458620 DOI: 10.3390/vaccines11081336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
The purpose of this study was to investigate possible influencing factors based on the distribution of the pertussis toxin (PT) and filamentous hemagglutinin (FHA) antibody levels in 0-14-year-old children in Jiangsu Province, China, and to analyze changes in IgG antibody levels after pertussis vaccination in children over time via a restricted cubic spline (RCS)-fitted binary logistic regression model. We collected surveillance data on pertussis through the National Notifiable Disease Reporting System (NNDRS). Serum samples were collected, and PT IgG/FHA IgG antibody levels were determined via an enzyme-linked immunosorbent assay (ELISA). A binary logistic regression model was fitted with an RCS. Peak incidence occurred in children aged 0-1 years from 2007 to 2022, and a second peak emerged in children aged 5 years and older from 2018 onwards which shifted towards older age groups. The geometric mean concentrations (GMC) of the anti-PT IgG antibody and anti-FHA IgG antibody in 1129 patients were 15.13 (13.49-16.76) IU/mL and 22.99 (21.17-24.81) IU/mL, respectively. The seropositivity rates of the anti-PT IgG and anti-FHA IgG antibodies in the group receiving a full vaccination course (four doses) were significantly higher than those of other groups (24.6% vs. 43.3%). The RCS fitting model showed a non-linear relationship between the duration after immunization and the odds ratio (OR) of having PT-IgG and FHA-IgG antibody concentrations ≥20 IU/mL in children with documented immunization histories (1-4 doses) (Poverall < 0. 001; Pnonlinear ≤ 0.001). The children with histories of immunization demonstrated antibody levels that decreased to very low levels around 17 months after the last dose of the vaccine. Therefore, it is recommended that pertussis-containing vaccines be administered as booster immunizations for older children.
Collapse
Affiliation(s)
- Wen Wang
- Department of Rheumatology and Immunology, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian 223800, China
| | - Zhiguo Wang
- Department of Expanded Program on Immunization, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China; (Z.W.)
| | - Qiang Chen
- Department of Expanded Program on Immunization, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China; (Z.W.)
| | - Mei Li
- Department of Expanded Program on Immunization, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China; (Z.W.)
| | - Chengmei Jia
- Department of Expanded Program on Immunization, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China; (Z.W.)
| | - Yan Xu
- Department of Expanded Program on Immunization, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China; (Z.W.)
| | - Yun Wu
- Department of Expanded Program on Immunization, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China; (Z.W.)
| | - Xiang Sun
- Department of Expanded Program on Immunization, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China; (Z.W.)
| | - Hui Sun
- Medical Department, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing 210000, China
| |
Collapse
|
17
|
Uversky VN, Redwan EM, Makis W, Rubio-Casillas A. IgG4 Antibodies Induced by Repeated Vaccination May Generate Immune Tolerance to the SARS-CoV-2 Spike Protein. Vaccines (Basel) 2023; 11:vaccines11050991. [PMID: 37243095 DOI: 10.3390/vaccines11050991] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Less than a year after the global emergence of the coronavirus SARS-CoV-2, a novel vaccine platform based on mRNA technology was introduced to the market. Globally, around 13.38 billion COVID-19 vaccine doses of diverse platforms have been administered. To date, 72.3% of the total population has been injected at least once with a COVID-19 vaccine. As the immunity provided by these vaccines rapidly wanes, their ability to prevent hospitalization and severe disease in individuals with comorbidities has recently been questioned, and increasing evidence has shown that, as with many other vaccines, they do not produce sterilizing immunity, allowing people to suffer frequent re-infections. Additionally, recent investigations have found abnormally high levels of IgG4 in people who were administered two or more injections of the mRNA vaccines. HIV, Malaria, and Pertussis vaccines have also been reported to induce higher-than-normal IgG4 synthesis. Overall, there are three critical factors determining the class switch to IgG4 antibodies: excessive antigen concentration, repeated vaccination, and the type of vaccine used. It has been suggested that an increase in IgG4 levels could have a protecting role by preventing immune over-activation, similar to that occurring during successful allergen-specific immunotherapy by inhibiting IgE-induced effects. However, emerging evidence suggests that the reported increase in IgG4 levels detected after repeated vaccination with the mRNA vaccines may not be a protective mechanism; rather, it constitutes an immune tolerance mechanism to the spike protein that could promote unopposed SARS-CoV2 infection and replication by suppressing natural antiviral responses. Increased IgG4 synthesis due to repeated mRNA vaccination with high antigen concentrations may also cause autoimmune diseases, and promote cancer growth and autoimmune myocarditis in susceptible individuals.
Collapse
Affiliation(s)
- Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria 21934, Egypt
| | - William Makis
- Cross Cancer Institute, Alberta Health Services, 11560 University Avenue, Edmonton, AB T6G 1Z2, Canada
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan 48900, Jalisco, Mexico
- Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan 48900, Jalisco, Mexico
| |
Collapse
|
18
|
Nian X, Liu H, Cai M, Duan K, Yang X. Coping Strategies for Pertussis Resurgence. Vaccines (Basel) 2023; 11:889. [PMID: 37242993 PMCID: PMC10220650 DOI: 10.3390/vaccines11050889] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Pertussis (whooping cough) is a respiratory disease caused primarily by Bordetella pertussis, a Gram-negative bacteria. Pertussis is a relatively contagious infectious disease in people of all ages, mainly affecting newborns and infants under 2 months of age. Pertussis is undergoing a resurgence despite decades of high rates of vaccination. To better cope with the challenge of pertussis resurgence, we evaluated its possible causes and potential countermeasures in the narrative review. Expanded vaccination coverage, optimized vaccination strategies, and the development of a new pertussis vaccine may contribute to the control of pertussis.
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
| | - Hongbo Liu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Mengyao Cai
- 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
| | - 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, Bejing 100029, China
| |
Collapse
|
19
|
da Silva Antunes R, Garrigan E, Quiambao LG, Dhanda SK, Marrama D, Westernberg L, Wang E, Sutherland A, Armstrong SK, Brickman TJ, Sidney J, Frazier A, Merkel T, Peters B, Sette A. Genome-wide characterization of T cell responses to Bordetella pertussis reveals broad reactivity and similar polarization irrespective of childhood vaccination profiles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.24.534182. [PMID: 36993748 PMCID: PMC10055406 DOI: 10.1101/2023.03.24.534182] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The incidence of whooping cough (pertussis), the respiratory disease caused by Bordetella pertussis (BP) has increased in recent years, and it is suspected that the switch from whole-cell pertussis (wP) to acellular pertussis (aP) vaccines may be a contributing factor to the rise in morbidity. While a growing body of evidence indicates that T cells play a role in the control and prevention of symptomatic disease, nearly all data on human BP-specific T cells is related to the four antigens contained in the aP vaccines, and data detailing T cell responses to additional non-aP antigens, are lacking. Here, we derived a full-genome map of human BP-specific CD4+ T cell responses using a high-throughput ex vivo Activation Induced Marker (AIM) assay, to screen a peptide library spanning over 3000 different BP ORFs. First, our data show that BP specific-CD4+ T cells are associated with a large and previously unrecognized breadth of responses, including hundreds of targets. Notably, fifteen distinct non-aP vaccine antigens were associated with reactivity comparable to that of the aP vaccine antigens. Second, the overall pattern and magnitude of CD4+ T cell reactivity to aP and non-aP vaccine antigens was similar regardless of aP vs wP childhood vaccination history, suggesting that the profile of T cell reactivity in adults is not driven by vaccination, but rather is likely driven by subsequent asymptomatic or sub-clinical infections. Finally, while aP vaccine responses were Th1/Th2 polarized as a function of childhood vaccination, CD4+ T cell responses to non-aP BP antigens vaccine responses were not, suggesting that these antigens could be used to avoid the Th2 bias associated with aP vaccination. Overall, these findings enhance our understanding of human T cell responses against BP and suggest potential targets for designing next-generation pertussis vaccines.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, California, USA
| | - Emily Garrigan
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, California, USA
| | - Lorenzo G Quiambao
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, California, USA
| | - Sandeep Kumar Dhanda
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, California, USA
| | - Daniel Marrama
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, California, USA
| | - Luise Westernberg
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, California, USA
| | - Eric Wang
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, California, USA
| | - Aaron Sutherland
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, California, USA
| | - Sandra K Armstrong
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Timothy J Brickman
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, California, USA
| | - April Frazier
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, California, USA
| | - Tod Merkel
- Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, California, USA
- University of California San Diego School of Medicine, La Jolla, San Diego, California, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, California, USA
- University of California San Diego School of Medicine, La Jolla, San Diego, California, USA
| |
Collapse
|
20
|
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
|
21
|
Szwejser-Zawislak E, Wilk MM, Piszczek P, Krawczyk J, Wilczyńska D, Hozbor D. Evaluation of Whole-Cell and Acellular Pertussis Vaccines in the Context of Long-Term Herd Immunity. Vaccines (Basel) 2022; 11:vaccines11010001. [PMID: 36679846 PMCID: PMC9863224 DOI: 10.3390/vaccines11010001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
After the pertussis vaccine had been introduced in the 1940s and was shown to be very successful in reducing the morbidity and mortality associated with the disease, the possibility of improving both vaccine composition and vaccination schedules has become the subject of continuous interest. As a result, we are witnessing a considerable heterogeneity in pertussis vaccination policies, which remains beyond universal consensus. Many pertussis-related deaths still occur in low- and middle-income countries; however, these deaths are attributable to gaps in vaccination coverage and limited access to healthcare in these countries, rather than to the poor efficacy of the first generation of pertussis vaccine consisting in inactivated and detoxified whole cell pathogen (wP). In many, particularly high-income countries, a switch was made in the 1990s to the use of acellular pertussis (aP) vaccine, to reduce the rate of post-vaccination adverse events and thereby achieve a higher percentage of children vaccinated. However the epidemiological data collected over the past few decades, even in those high-income countries, show an increase in pertussis prevalence and morbidity rates, triggering a wide-ranging debate on the causes of pertussis resurgence and the effectiveness of current pertussis prevention strategies, as well as on the efficacy of available pertussis vaccines and immunization schedules. The current article presents a systematic review of scientific reports on the evaluation of the use of whole-cell and acellular pertussis vaccines, in the context of long-term immunity and vaccines efficacy.
Collapse
Affiliation(s)
- Ewa Szwejser-Zawislak
- Institute of Biotechnology of Serums and Vaccines Biomed, Al. Sosnowa 8, 30-224 Krakow, Poland
| | - Mieszko M. Wilk
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Piotr Piszczek
- Institute of Biotechnology of Serums and Vaccines Biomed, Al. Sosnowa 8, 30-224 Krakow, Poland
| | - Justyna Krawczyk
- Institute of Biotechnology of Serums and Vaccines Biomed, Al. Sosnowa 8, 30-224 Krakow, Poland
| | - Daria Wilczyńska
- Institute of Biotechnology of Serums and Vaccines Biomed, Al. Sosnowa 8, 30-224 Krakow, Poland
| | - Daniela Hozbor
- VacSal Laboratory, Institute of Biotechnology and Molecular Biology, Faculty of Sciences, National University of La Plata (UNLP), National Council for Scientific and Technical Research (CONICET), La Plata 1900, Argentina
- Correspondence:
| |
Collapse
|
22
|
Prior exposure to B. pertussis shapes the mucosal antibody response to acellular pertussis booster vaccination. Nat Commun 2022; 13:7429. [PMID: 36460655 PMCID: PMC9716536 DOI: 10.1038/s41467-022-35165-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/21/2022] [Indexed: 12/03/2022] Open
Abstract
Bordetella pertussis (Bp), the causative agent of pertussis, continues to circulate despite widespread vaccination programs. An important question is whether and how (sub)clinical infections shape immune memory to Bp, particularly in populations primed with acellular pertussis vaccines (aP). Here, we examine the prevalence of mucosal antibodies against non-vaccine antigens in aP-primed children and adolescents of the BERT study (NCT03697798), using antibody binding to a Bp mutant strain lacking aP antigens (Bp_mut). Our study identifies increased levels of mucosal IgG and IgA binding to Bp_mut in older aP-primed individuals, suggesting different Bp exposure between aP-primed birth cohorts, in line with pertussis disease incidence data. To examine whether Bp exposure influences vaccination responses, we measured mucosal antibody responses to aP booster vaccination as a secondary study outcome. Although booster vaccination induces significant increases in mucosal antibodies to Bp in both cohorts, the older age group that had higher baseline antibodies to Bp_ mut shows increased persistence of antibodies after vaccination.
Collapse
|
23
|
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
|
24
|
Raihan MO, Espelien BM, Hanson C, McGregor BA, Velaris NA, Alvine TD, Al Golovko S, Bradley DS, Nilles M, Glovko MY, Hur J, Porter JE. Characterization of prostanoids response to Bordetella pertussis antigen BscF and Tdap in LPS-challenged monocytes. Prostaglandins Leukot Essent Fatty Acids 2022; 182:102452. [PMID: 35690004 DOI: 10.1016/j.plefa.2022.102452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 12/29/2022]
Abstract
Prostanoids are potent inflammatory mediators that play a regulatory role in the innate immune activation of the adaptive immune response to determine the duration of protection against infection. We aim to quantify the modulation of prostanoids profiles in lipopolysaccharide (LPS)-stimulated THP-1 cells treated with the novel pertussis antigen BscF. We compared the effect with pertussis antigens present in the current Tdap vaccine to understand the immunomodulatory effect that might contribute to the diminished Tdap vaccine effectiveness. The inflammatory challenge with LPS induced a robust elevation of most prostanoid family members compared to the control treatment. Treatment with BscF and Tdap significantly reduced the LPS-stimulated elevation of prostaglandins (PGs) D2, E2, and F2α, as well as thromboxane (TX) A2 levels. An opposite trend was observed for PGI2, as both antigens accelerated the LPS-stimulated upregulation. Further, we quantified cyclooxygenases (COXs) that catalyze the biosynthesis of prostanoids and found that both antigens significantly reduced LPS-stimulated COX-1 and COX-2, demonstrating that the waning of acellular pertussis vaccines' protective immunity may be due to other downstream enzymes not related to COXs. Our present study validates the potential role of BscF as an adjuvant, resulting in the next-generation pertussis vaccine discovery.
Collapse
Affiliation(s)
- Md Obayed Raihan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - Brenna M Espelien
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - Courtney Hanson
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - Brett A McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - Nathan A Velaris
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - Travis D Alvine
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - Svetlana Al Golovko
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - David S Bradley
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - Matthew Nilles
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - Mikhail Y Glovko
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - James E Porter
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States.
| |
Collapse
|
25
|
Versteegen P, Barkoff AM, Valente Pinto M, van de Kasteele J, Knuutila A, Bibi S, de Rond L, Teräsjärvi J, Sanders K, de Zeeuw-Brouwer ML, Luoto R, ten Hulscher H, Clutterbuck EA, Sanders EAM, Mertsola J, Berbers GAM, He Q, Kelly DF, Buisman AM. Memory B Cell Activation Induced by Pertussis Booster Vaccination in Four Age Groups of Three Countries. Front Immunol 2022; 13:864674. [PMID: 35677044 PMCID: PMC9168128 DOI: 10.3389/fimmu.2022.864674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundImmunogenicity of acellular pertussis (aP) vaccines is conventionally assessed by measuring antibody responses but antibody concentrations wane quickly after vaccination. Memory B cells, however, are critical in sustaining long-term protection and therefore may be an important factor when assessing pertussis immunity after vaccination.AimWe studied pertussis specific memory B cell (re)activation induced by an aP booster vaccination in four different age groups within three countries.Materials and methodsFrom a phase IV longitudinal interventional study, 268 participants across Finland, the Netherlands and the United Kingdom were included and received a 3-component pertussis booster vaccine: children (7-10y, n=53), adolescents (11-15y, n=66), young adults (20-34y, n=74), and older adults (60-70y, n=75). Memory B cells at baseline, day 28, and 1 year post-vaccination were measured by a pertussis toxin (Ptx), filamentous haemagglutinin (FHA), and pertactin (Prn) specific ELISpot assay. Antibody results measured previously were available for comparison. Furthermore, study participants were distributed into groups based on their baseline memory B cell frequencies, vaccine responses were monitored between these groups.ResultsGeometric mean (GM) memory B cell frequencies for pertussis antigens at baseline were low. At 28 days post-vaccination, these frequencies increased within each age group and were still elevated one year post-booster compared to baseline. Highest frequencies at day 28 were found within adolescents (GM: 5, 21, and 13, for Ptx, FHA and Prn, respectively) and lowest within older adults (GM: 2, 9, and 3, respectively). Moderate to strong correlations between memory B cell frequencies at day 28 and antibody concentrations at day 28 and 1 year were observed for Prn. Memory B cell frequencies > 1 per 100,000 PBMCs at baseline were associated with significantly higher memory responses after 28 days and 1 year.ConclusionsAn aP booster vaccine (re)activated memory B cells in all age groups. Still elevated memory B cell frequencies after one year indicates enhanced immunological memory. However, antigen specific memory B cell activation seems weaker in older adults, which might reflect immunosenescence. Furthermore, the presence of circulating memory B cells at baseline positively affects memory B cell responses. This study was registered at www.clinicaltrialsregister.eu: No. 2016-003678-42.
Collapse
Affiliation(s)
- Pauline Versteegen
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | - Alex-Mikael Barkoff
- Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, University of Turku, Turku, Finland
| | - Marta Valente Pinto
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Jan van de Kasteele
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | - Aapo Knuutila
- Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, University of Turku, Turku, Finland
| | - Sagida Bibi
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Lia de Rond
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | - Johanna Teräsjärvi
- Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, University of Turku, Turku, Finland
| | - Katherine Sanders
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Mary-lène de Zeeuw-Brouwer
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | - Raakel Luoto
- Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, University of Turku, Turku, Finland
| | - Hinke ten Hulscher
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | | | - Elisabeth A. M. Sanders
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital, Utrecht, Netherlands
| | - Jussi Mertsola
- Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, University of Turku, Turku, Finland
| | - Guy A. M. Berbers
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
| | - Qiushui He
- Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, University of Turku, Turku, Finland
| | - Dominic F. Kelly
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Anne-Marie Buisman
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, Netherlands
- *Correspondence: Anne-Marie Buisman,
| | | |
Collapse
|
26
|
Long-Term Immunogenicity upon Pertussis Booster Vaccination in Young Adults and Children in Relation to Priming Vaccinations in Infancy. Vaccines (Basel) 2022; 10:vaccines10050693. [PMID: 35632449 PMCID: PMC9146390 DOI: 10.3390/vaccines10050693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 02/01/2023] Open
Abstract
Booster vaccinations for pertussis are advised in many countries during childhood or adulthood. In a phase IV longitudinal interventional study, we assessed long-term immunity following an extra pertussis booster vaccination in children and adults. Children (9 years of age) were primed in infancy with either the Dutch whole cell pertussis (wP) vaccine (n = 49) or acellular pertussis (aP) vaccines (n = 59), and all children received a preschool aP booster. Adults (25–29 years, n = 86) were wP-primed in infancy and did not receive a preschool booster. All were followed-up for approximately 6 years. After the additional booster, antibody responses to pertussis were more heterogeneous but generally higher in adults compared with children, and additional modelling showed that antibody concentrations remained higher for at least a decade. Serologic parameters indicative of recent pertussis infection were more often found in aP-primed children (12%) compared with wP-primed individuals (2%) (p = 0.052). This suggests that the aP booster vaccination in aP-primed children offers less long-term protection against pertussis infection and consequently against transmission. Together, these data show that aP priming in combination with aP boosting may not be sufficient to prevent circulation and transmission, while wP-primed adults may benefit from enhanced long-lasting immunity.
Collapse
|
27
|
Barman S, Soni D, Brook B, Nanishi E, Dowling DJ. Precision Vaccine Development: Cues From Natural Immunity. Front Immunol 2022; 12:662218. [PMID: 35222350 PMCID: PMC8866702 DOI: 10.3389/fimmu.2021.662218] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 12/21/2021] [Indexed: 12/31/2022] Open
Abstract
Traditional vaccine development against infectious diseases has been guided by the overarching aim to generate efficacious vaccines normally indicated by an antibody and/or cellular response that correlates with protection. However, this approach has been shown to be only a partially effective measure, since vaccine- and pathogen-specific immunity may not perfectly overlap. Thus, some vaccine development strategies, normally focused on targeted generation of both antigen specific antibody and T cell responses, resulting in a long-lived heterogenous and stable pool of memory lymphocytes, may benefit from better mimicking the immune response of a natural infection. However, challenges to achieving this goal remain unattended, due to gaps in our understanding of human immunity and full elucidation of infectious pathogenesis. In this review, we describe recent advances in the development of effective vaccines, focusing on how understanding the differences in the immunizing and non-immunizing immune responses to natural infections and corresponding shifts in immune ontogeny are crucial to inform the next generation of infectious disease vaccines.
Collapse
Affiliation(s)
- Soumik Barman
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Dheeraj Soni
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Byron Brook
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Etsuro Nanishi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - David J Dowling
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
28
|
Aibani N, Patel P, Buchanan R, Strom S, Wasan KM, Hancock REW, Gerdts V, Wasan EK. Assessing the In Vivo Effectiveness of Cationic Lipid Nanoparticles with a Triple Adjuvant for Intranasal Vaccination against the Respiratory Pathogen Bordetella pertussis. Mol Pharm 2022; 19:1814-1824. [PMID: 35302764 DOI: 10.1021/acs.molpharmaceut.1c00852] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Continuous outbreaks of pertussis around the world suggest inadequate immune protection in infants and weakened immune responses induced over time by the acellular pertussis vaccine. Vaccine adjuvants provide a means to improve vaccine immunogenicity and support long-term adaptive immunity against pertussis. An acellular pertussis vaccine was prepared with pertactin, pertussis toxin, and fimbriae 2/3 antigens combined with a triple-adjuvant system consisting of innate defense regulator peptide IDR 1002, a Toll-like receptor-3 agonist poly(I:C), and a polyphosphazene in a fixed combination. The vaccine was delivered intranasally in a cationic lipid nanoparticle formulation fabricated by simple admixture and two schema for addition of antigens (LT-A, antigens associated outside of L-TriAdj, and LAT, antigens associated inside of L-TriAdj) to optimize particle size and cationic surface charge. In the former, antigens were associated with the lipidic formulation of the triple adjuvant by electrostatic attraction. In the latter, the antigens resided in the interior of the lipid nanoparticle. Two dose levels of antigens were used with adjuvant comprised of the triple adjuvant with or without the lipid nanoparticle carrier. Formulation of vaccines with the triple adjuvant stimulated systemic and mucosal immune responses. The lipid nanoparticle vaccines favored a Th1 type of response with higher IgG2a and IgA serum antibody titers particularly for pertussis toxin and pertactin formulated at the 5 μg dose level in the admixed formulation. Additionally, the lipid nanoparticle vaccines resulted in high nasal SIgA antibodies and an early (4 weeks post vaccination) response after a single vaccination dose. The LT-A nanoparticles trended toward higher titers of serum antibodies compared to LAT. The cationic lipid-based vaccine nanoparticles formulated with a triple adjuvant showed encouraging results as a potential formulation for intranasally administered pertussis vaccines.
Collapse
Affiliation(s)
- Noorjahan Aibani
- University of Saskatchewan, College of Pharmacy and Nutrition, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Parth Patel
- University of Saskatchewan, College of Pharmacy and Nutrition, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Rachelle Buchanan
- Vaccine and Infectious Disease Organization, University of Saskatchewan, 120 Veterinary Road, Saskatoon, Saskatchewan S7N 5E3, Canada
| | - Stacy Strom
- Vaccine and Infectious Disease Organization, University of Saskatchewan, 120 Veterinary Road, Saskatoon, Saskatchewan S7N 5E3, Canada
| | - Kishor M Wasan
- Department of Urological Sciences, University of British Columbia Faculty of Medicine, Gordon & Leslie Diamond Health Care Centre, Vancouver, British Columbia V5Z 1M9, Canada
| | - Robert E W Hancock
- Centre for Microbial Diseases & Immunity Research, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Volker Gerdts
- Vaccine and Infectious Disease Organization, University of Saskatchewan, 120 Veterinary Road, Saskatoon, Saskatchewan S7N 5E3, Canada
| | - Ellen K Wasan
- University of Saskatchewan, College of Pharmacy and Nutrition, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| |
Collapse
|
29
|
Kroes MM, Miranda-Bedate A, Jacobi RHJ, van Woudenbergh E, den Hartog G, van Putten JPM, de Wit J, Pinelli E. Bordetella pertussis-infected innate immune cells drive the anti-pertussis response of human airway epithelium. Sci Rep 2022; 12:3622. [PMID: 35256671 PMCID: PMC8901624 DOI: 10.1038/s41598-022-07603-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 02/21/2022] [Indexed: 12/13/2022] Open
Abstract
Pertussis is a severe respiratory tract infection caused by Bordetella pertussis. This bacterium infects the ciliated epithelium of the human airways. We investigated the epithelial cell response to B. pertussis infection in primary human airway epithelium (HAE) differentiated at air-liquid interface. Infection of the HAE cells mimicked several hallmarks of B. pertussis infection such as reduced epithelial barrier integrity and abrogation of mucociliary transport. Our data suggests mild immunological activation of HAE by B. pertussis indicated by secretion of IL-6 and CXCL8 and the enrichment of genes involved in bacterial recognition and innate immune processes. We identified IL-1β and IFNγ, present in conditioned media derived from B. pertussis-infected macrophage and NK cells, as essential immunological factors for inducing robust chemokine secretion by HAE in response to B. pertussis. In transwell migration assays, the chemokine-containing supernatants derived from this HAE induced monocyte migration. Our data suggests that the airway epithelium on its own has a limited immunological response to B. pertussis and that for a broad immune response communication with local innate immune cells is necessary. This highlights the importance of intercellular communication in the defense against B. pertussis infection and may assist in the rational design of improved pertussis vaccines.
Collapse
Affiliation(s)
- M M Kroes
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands.,Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - A Miranda-Bedate
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - R H J Jacobi
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - E van Woudenbergh
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands.,Section Paediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - G den Hartog
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - J P M van Putten
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - J de Wit
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - E Pinelli
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands.
| |
Collapse
|
30
|
Bbvac: A Live Vaccine Candidate That Provides Long-Lasting Anamnestic and Th17-Mediated Immunity against the Three Classical Bordetella spp. mSphere 2022; 7:e0089221. [PMID: 35196124 PMCID: PMC8865921 DOI: 10.1128/msphere.00892-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Acute pathogens such as Bordetella pertussis can cause severe disease but are ultimately cleared by the immune response. This has led to the accepted paradigm that convalescent immunity is optimal and therefore broadly accepted as the “gold standard” against which vaccine candidates should be compared. However, successful pathogens like B. pertussis have evolved multiple mechanisms for suppressing and evading host immunity, raising the possibility that disruption of these mechanisms could result in substantially stronger or better immunity. Current acellular B. pertussis vaccines, delivered in a 5-dose regimen, induce only short-term immunity against disease and even less against colonization and transmission. Importantly, they provide modest protection against other Bordetella species that cause substantial human disease. A universal vaccine that protects against the three classical Bordetella spp. could decrease the burden of whooping cough-like disease in humans and other animals. Our recent work demonstrated that Bordetella spp. suppress host inflammatory responses and that disrupting the regulation of immunosuppressive mechanisms can allow the host to generate substantially stronger sterilizing immunity against the three classical Bordetella spp. Here, we identify immune parameters impacted by Bordetella species immunomodulation, including the generation of robust Th17 and B cell memory responses. Disrupting immunomodulation augmented the immune response, providing strong protection against the prototypes of all three classical Bordetella spp. as well as recent clinical isolates. Importantly, the protection in mice lasted for at least 15 months and was associated with recruitment of high numbers of B and T cells in the lungs as well as enhanced Th17 mucosal responses and persistently high titers of antibodies. These findings demonstrate that disrupting bacterial immunomodulatory pathways can generate much stronger and more protective immune responses to infection, with important implications for the development of better vaccines. IMPORTANCE Infectious diseases are a major cause of morbidity and mortality in the United States, accounting for over 40 million hospitalizations since 1998. Therefore, novel vaccine strategies are imperative, which can be improved with a better understanding of the mechanisms that bacteria utilize to suppress host immunity, a key mechanism for establishing colonization. Bordetella spp., the causative agents of whooping cough, suppress host immunity, which allows for persistent colonization. We discovered a regulator of a bacterial immunosuppressive pathway, which, when mutated in Bordetella spp., allows for rapid clearance of infection and subsequent generation of protective immunity for at least 15 months. After infection with the mutant strain, mice exhibited sterilizing immunity against the three classical Bordetella spp., suggesting that the immune response can be both stronger and cross-protective. This work presents a strategy for vaccine development that can be applied to other immunomodulatory pathogens.
Collapse
|
31
|
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: 4.5] [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
|
32
|
Prygiel M, Mosiej E, Górska P, Zasada AA. Diphtheria-tetanus-pertussis vaccine: past, current & future. Future Microbiol 2021; 17:185-197. [PMID: 34856810 DOI: 10.2217/fmb-2021-0167] [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] Open
Abstract
The diphtheria-tetanus-pertussis (DTP) vaccine can prevent diphtheria, tetanus and pertussis. The component antigens of the DTP vaccine had long been monovalent vaccines. The pertussis vaccine was licensed in 1914. The same year, the mixtures of diphtheria toxin and antitoxin were put into use. In 1926, alum-precipitated diphtheria toxoid was registered, and in 1937 adsorbed tetanus toxoid was put on the market. The development of numerous effective DTP vaccines quickly stimulated efforts to combine DTP with other routine vaccines for infants. This overview covers the most important information regarding the invention of DTP vaccines, their modifications and the needs that should be focused on in the future.
Collapse
Affiliation(s)
- Marta Prygiel
- Department of Vaccine & Sera Evaluation, The National Institute of Public Health NIH - National Research Institute, Warsaw, Poland
| | - Ewa Mosiej
- Department of Vaccine & Sera Evaluation, The National Institute of Public Health NIH - National Research Institute, Warsaw, Poland
| | - Paulina Górska
- Department of Vaccine & Sera Evaluation, The National Institute of Public Health NIH - National Research Institute, Warsaw, Poland
| | - Aleksandra A Zasada
- Department of Vaccine & Sera Evaluation, The National Institute of Public Health NIH - National Research Institute, Warsaw, Poland
| |
Collapse
|
33
|
Versteegen P, Berbers GA, Smits G, Sanders EA, van der Klis FR, de Melker HE, van der Maas NA. More than 10 years after introduction of an acellular pertussis vaccine in infancy: a cross-sectional serosurvey of pertussis in the Netherlands. THE LANCET REGIONAL HEALTH. EUROPE 2021; 10:100196. [PMID: 34806066 PMCID: PMC8589709 DOI: 10.1016/j.lanepe.2021.100196] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Background Pertussis is a respiratory disease and still endemic despite high vaccination coverage. In the Dutch national immunisation programme (NIP) whole cell pertussis (wP) priming vaccines for infants were replaced by acellular pertussis (aP) priming vaccines in 2005. Serosurveillance gives the opportunity to objectively monitor effects of changes in the NIP on infection prevalence and vaccine response in the population over time. Methods For this population-based cross-sectional serosurvey a representative sample of Dutch residents (0-89 years) was drawn in 2016/2017. Primary outcome was the percentage of participants with pertussis toxin specific antibody concentrations ≥ 100 IU/ml as an indicator of recent infection, and to identify groups possibly more vulnerable to pertussis infection. Percentages were compared with previous results from 2006/2007. Findings In total 7621 persons were included in the analysis. An increase in recent infections from 3•5% to 5•9% was found in the population from 7 years and older (n=6013) in 2016/2017 compared with 2006/2007. Most noteworthy increase was seen in 12-18-year-olds who were wP primed and aP boosted. Interpretation Infection prevalence is still increasing in the Netherlands inducing a risk of pertussis disease in vulnerable (age) groups. Delaying the preschool booster might prolong the period of protection during primary school and thereby possibly protect younger siblings. Extra boosters might be considered for risk populations like older adults and people with (pulmonary) co-morbidities, since they have higher chances of complications and hospitalisation. An unedited Dutch translation of the abstract is available in Supplementary text 1: Nederlandse samenvatting. Funding The Dutch Ministry of Health, Welfare, and Sport.
Collapse
Affiliation(s)
- Pauline Versteegen
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, 3720 BA, Netherlands
- Corresponding author: Pauline Versteegen, MD, Centre for Infectious Disease Control, National Institute of Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands, +31 6 23 76 44 11
| | - Guy A.M. Berbers
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, 3720 BA, Netherlands
| | - Gaby Smits
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, 3720 BA, Netherlands
| | - Elisabeth A.M. Sanders
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, 3720 BA, Netherlands
- Wilhelmina Children's Hospital, Department of Paediatric Immunology and Infectious Diseases, Lundlaan 6, 3584 EA Utrecht, Netherlands
| | - Fiona R.M. van der Klis
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, 3720 BA, Netherlands
| | - Hester E. de Melker
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, 3720 BA, Netherlands
| | - Nicoline A.T. van der Maas
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Bilthoven, 3720 BA, Netherlands
| |
Collapse
|
34
|
Perez Chacon G, Ramsay J, Brennan-Jones CG, Estcourt MJ, Richmond P, Holt P, Snelling T. Whole-cell pertussis vaccine in early infancy for the prevention of allergy in children. Cochrane Database Syst Rev 2021; 9:CD013682. [PMID: 34693993 PMCID: PMC8543786 DOI: 10.1002/14651858.cd013682.pub2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Atopic diseases are the most common chronic conditions of childhood. The apparent rise in food anaphylaxis in young children over the past three decades is of particular concern, owing to the lack of proven prevention strategies other than the timely introduction of peanut and egg. Due to reported in vitro differences in the immune response of young infants primed with whole-cell pertussis (wP) versus acellular pertussis (aP) vaccine, we systematically appraised and synthesised evidence on the safety and the potential allergy preventive benefits of wP, to inform recommendation for future practice and research. OBJECTIVES To assess the efficacy and safety of wP vaccinations in comparison to aP vaccinations in early infancy for the prevention of atopic diseases in children. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials, Ovid MEDLINE, Embase, and grey literature. The date of the search was 7 September 2020. SELECTION CRITERIA We included randomised controlled trials (RCTs) and non-randomised studies of interventions (NRSIs) that reported the occurrence of atopic diseases, and RCTs only to assess safety outcomes. To be included studies had to have at least six months follow-up, and involve children under 18 years old, who received a first dose of either wP (experimental intervention) or aP (comparator) before six months of age. DATA COLLECTION AND ANALYSIS Two review authors independently screened studies for eligibility, extracted the data, and assessed risk of bias using standard Cochrane methods. We assessed the certainty of the evidence using GRADE. Our primary outcomes were diagnosis of IgE-mediated food allergy and all-cause serious adverse events (SAEs). Secondary outcomes included: diagnosis of not vaccine-associated anaphylaxis or urticaria, diagnosis of asthma, diagnosis of allergic rhinitis, diagnosis of atopic dermatitis and diagnosis of encephalopathy. Due to paucity of RCTs reporting on the atopic outcomes of interest, we assessed a broader outcome domain (cumulative incidence of atopic disease) as specified in our protocol. We summarised effect estimates as risk ratios (RR) and 95% confidence intervals (CI). Where appropriate, we pooled safety data in meta-analyses using fixed-effect Mantel-Haenszel methods, without zero-cell corrections for dichotomous outcomes. MAIN RESULTS We identified four eligible studies reporting on atopic outcomes, representing 7333 children. Based on a single trial, there was uncertain evidence on whether wP vaccines affected the risk of overall atopic disease (RR 0.85, 95% CI 0.62 to 1.17) or asthma only (RR 1.04, 95% CI 0.59 to 1.82; 497 children) by 2.5 years old.Three NRSIs were judged to be at serious or critical risk of bias due to confounding, missing data, or both, and were ineligible for inclusion in a narrative synthesis. We identified 21 eligible studies (137,281 children) that reported the safety outcomes of interest. We judged seven studies to be at high risk of bias and those remaining, at unclear risk. The pooled RR was 0.94 for all-cause SAEs (95% CI 0.78 to 1.15; I2 = 0%; 15 studies, 38,072 children). For every 1000 children primed with a first dose of wP, 11 had an SAE. The corresponding risk with aP was 12 children (95% CI 9 to 13). The 95% CI around the risk difference ranged from three fewer to two more events per 1000 children, and the certainty of the evidence was judged as moderate (downgraded one level for imprecision). No diagnoses of encephalopathy following vaccination were reported (95% CI around the risk difference - 5 to 12 per 100,000 children; seven primary series studies; 115,271 children). The certainty of the evidence was judged as low, since this is a serious condition, and we could not exclude a clinically meaningful difference. AUTHORS' CONCLUSIONS There is very low-certainty evidence that a first dose of wP given early in infancy, compared to a first dose of aP, affects the risk of atopic diseases in children. The incidence of all-cause SAEs in wP and aP vaccinees was low, and no cases of encephalopathy were reported. The certainty of the evidence was judged as moderate for all-cause SAEs, and low for encephalopathy. Future studies should use sensitive and specific endpoints of clinical relevance, and should be conducted in settings with high prevalence of IgE-mediated food allergy. Safety endpoints should prioritise common vaccine reactions, parental acceptability, SAEs and their potential relatedness to the dose administered.
Collapse
Affiliation(s)
- Gladymar Perez Chacon
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
- School of Public Health, Curtin University, Perth, Australia
| | - Jessica Ramsay
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
| | | | - Marie J Estcourt
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
| | - Peter Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
- Division of Paediatrics, The University of Western Australia, Perth, Australia
| | - Patrick Holt
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Tom Snelling
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
| |
Collapse
|
35
|
Pan C, Yue H, Zhu L, Ma GH, Wang HL. Prophylactic vaccine delivery systems against epidemic infectious diseases. Adv Drug Deliv Rev 2021; 176:113867. [PMID: 34280513 PMCID: PMC8285224 DOI: 10.1016/j.addr.2021.113867] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/20/2021] [Accepted: 07/11/2021] [Indexed: 01/04/2023]
Abstract
Prophylactic vaccines have evolved from traditional whole-cell vaccines to safer subunit vaccines. However, subunit vaccines still face problems, such as poor immunogenicity and low efficiency, while traditional adjuvants are usually unable to meet specific response needs. Advanced delivery vectors are important to overcome these barriers; they have favorable safety and effectiveness, tunable properties, precise location, and immunomodulatory capabilities. Nevertheless, there has been no systematic summary of the delivery systems to cover a wide range of infectious pathogens. We herein summarized and compared the delivery systems for major or epidemic infectious diseases caused by bacteria, viruses, fungi, and parasites. We also included the newly licensed vaccines (e.g., COVID-19 vaccines) and those close to licensure. Furthermore, we highlighted advanced delivery systems with high efficiency, cross-protection, or long-term protection against epidemic pathogens, and we put forward prospects and thoughts on the development of future prophylactic vaccines.
Collapse
Affiliation(s)
- Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, PR China
| | - Hua Yue
- 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
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, PR China
| | - Guang-Hui Ma
- 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.
| | - Heng-Liang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, PR China.
| |
Collapse
|
36
|
Elas M, Villatoro N, Pezzoli L. Disproportionality analysis of reported drug adverse events to assess a potential safety signal for pentavalent vaccine in 2019 in El Salvador. Vaccine 2021; 39:4849-4855. [PMID: 34275672 DOI: 10.1016/j.vaccine.2021.07.010] [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: 05/11/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 10/20/2022]
Abstract
Detection and surveillance of vaccine safety hazards is a public health staple. In the post-marketing phase, when vaccines are used in mass, it is crucial to monitor potential signals of adverse reactions that may have been missed in the pre-marketing phase. We analysed spontaneous reports of drug adverse events in El Salvador to assess a potential safety signal related to an increase in febrile seizures following the pentavalent (diphtheria, tetanus, pertussis, hepatitis B, and Haemophilus influenzae Type B) vaccine in 2019. This was a retrospective observational study of adverse event notifications in the national electronic drug safety database from 2011 to 2019. We performed standard disproportionality analysis computing Proportional Reporting Risk (PRR), Reporting Odds Ratio (ROR), Relative Reporting Ratio (RRR), Chi-squared, and Information Component (IC), comparing the pairing of febrile seizures and pentavalent vaccine to all other drugs and adverse events recorded in 2019. The occurrence of febrile seizures following pentavalent vaccination exceeded the WHO expected rate of six cases × 100 000 doses administered from April 2019, with a maximum of 9.2 in September. IC was 4.3, ORR 421.9 (95% Confidence Interval, CI: 123.8-1437.7), PRR 223.5 (95 %CI: 70.2-710.9), RRR was 19.5. The first booster presented the highest rate (14.6 per 100,000 doses) of febrile seizures, more than double than expected. Rates for 2018 remained below expected. Reports of febrile seizures following pentavalent vaccine were also on the increase globally since 2014, with highest rates in 2018 and 2019. There was a disproportion of febrile seizures notifications following pentavalent in El Salvador in 2019, suggesting the existence of a safety signal. This may be due to the change in provider. Further studies should assess the causes of the increase and compute costs and benefits of this vaccination to determine if switching to a less reactogenic vaccine formulation is indicated.
Collapse
Affiliation(s)
- Miguel Elas
- National Center for Pharmacovigilance, Ministry of Health, San Salvador, El Salvador; Master's and Graduate School, Faculty of Medicine, University of El Salvador, San Salvador, El Salvador.
| | - Nora Villatoro
- National Vaccination and Inmunization Program, Ministry of Health, San Salvador, El Salvador.
| | | |
Collapse
|
37
|
Kroes MM, Miranda-Bedate A, Hovingh ES, Jacobi R, Schot C, Pupo E, Raeven RHM, van der Ark AAJ, van Putten JPM, de Wit J, Mariman R, Pinelli E. Naturally circulating pertactin-deficient Bordetella pertussis strains induce distinct gene expression and inflammatory signatures in human dendritic cells. Emerg Microbes Infect 2021; 10:1358-1368. [PMID: 34132167 PMCID: PMC8259873 DOI: 10.1080/22221751.2021.1943537] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Respiratory infections caused by Bordetella pertussis are reemerging despite high pertussis vaccination coverage. Since the introduction of the acellular pertussis vaccine in the late twentieth century, circulating B. pertussis strains increasingly lack expression of the vaccine component pertactin (Prn). In some countries, up to 90% of the circulating B. pertussis strains are deficient in Prn. To better understand the resurgence of pertussis, we investigated the response of human monocyte-derived dendritic cells (moDCs) to naturally circulating Prn-expressing (Prn-Pos) and Prn-deficient (Prn-Neg) B. pertussis strains from 2016 in the Netherlands. Transcriptome analysis of moDC showed enriched IFNα response-associated gene expression after exposure to Prn-Pos B. pertussis strains, whereas the Prn-Neg strains induced enriched expression of interleukin- and TNF-signaling genes, as well as other genes involved in immune activation. Multiplex immune assays confirmed enhanced proinflammatory cytokine secretion by Prn-Neg stimulated moDC. Comparison of the proteomes from the Prn-Pos and Prn-Neg strains revealed, next to the difference in Prn, differential expression of a number of other proteins including several proteins involved in metabolic processes. Our findings indicate that Prn-deficient B. pertussis strains induce a distinct and stronger immune activation of moDCs than the Prn-Pos strains. These findings highlight the role of pathogen adaptation in the resurgence of pertussis as well as the effects that vaccine pressure can have on a bacterial population.
Collapse
Affiliation(s)
- Michiel M Kroes
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands.,Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Alberto Miranda-Bedate
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Elise S Hovingh
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Ronald Jacobi
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Corrie Schot
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Elder Pupo
- Institute for Translational Vaccinology (Intravacc), Bilthoven, Netherlands
| | - René H M Raeven
- Institute for Translational Vaccinology (Intravacc), Bilthoven, Netherlands
| | | | - Jos P M van Putten
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jelle de Wit
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Rob Mariman
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Elena Pinelli
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| |
Collapse
|
38
|
da Silva Antunes R, Soldevila F, Pomaznoy M, Babor M, Bennett J, Tian Y, Khalil N, Qian Y, Mandava A, Scheuermann RH, Cortese M, Pulendran B, Petro CD, Gilkes AP, Purcell LA, Sette A, Peters B. A system-view of Bordetella pertussis booster vaccine responses in adults primed with whole-cell versus acellular vaccine in infancy. JCI Insight 2021; 6:141023. [PMID: 33690224 PMCID: PMC8119213 DOI: 10.1172/jci.insight.141023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 03/03/2021] [Indexed: 01/26/2023] Open
Abstract
The increased incidence of whooping cough worldwide suggests that current vaccination against Bordetella pertussis infection has limitations in quality and duration of protection. The resurgence of infection has been linked to the introduction of acellular vaccines (aP), which have an improved safety profile compared with the previously used whole-cell (wP) vaccines. To determine immunological differences between aP and wP priming in infancy, we performed a systems approach of the immune response to booster vaccination. Transcriptomic, proteomic, cytometric, and serologic profiling revealed multiple shared immune responses with different kinetics across cohorts, including an increase of blood monocyte frequencies and strong antigen-specific IgG responses. Additionally, we found a prominent subset of aP-primed individuals (30%) with a strong differential signature, including higher levels of expression for CCL3, NFKBIA, and ICAM1. Contrary to the wP individuals, this subset displayed increased PT-specific IgE responses after boost and higher antigen-specific IgG4 and IgG3 antibodies against FHA and FIM2/3 at baseline and after boost. Overall, the results show that, while broad immune response patterns to Tdap boost overlap between aP- and wP-primed individuals, a subset of aP-primed individuals present a divergent response. These findings provide candidate targets to study the causes and correlates of waning immunity after aP vaccination.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Ferran Soldevila
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Mikhail Pomaznoy
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Mariana Babor
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Jason Bennett
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Yuan Tian
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Natalie Khalil
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Yu Qian
- J. Craig Venter Institute, La Jolla, California, USA
| | | | - Richard H. Scheuermann
- J. Craig Venter Institute, La Jolla, California, USA
- University of California San Diego School of Medicine, La Jolla, California, USA
| | - Mario Cortese
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Bali Pulendran
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | | | | | | | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
- Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
- University of California San Diego School of Medicine, La Jolla, California, USA
| |
Collapse
|
39
|
Vaure C, Grégoire-Barou V, Courtois V, Chautard E, Dégletagne C, Liu Y. Göttingen Minipigs as a Model to Evaluate Longevity, Functionality, and Memory of Immune Response Induced by Pertussis Vaccines. Front Immunol 2021; 12:613810. [PMID: 33815369 PMCID: PMC8009978 DOI: 10.3389/fimmu.2021.613810] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 02/22/2021] [Indexed: 11/13/2022] Open
Abstract
Evaluation of the short-term and long-term immunological responses in a preclinical model that simulates the targeted age population with a relevant vaccination schedule is essential for human vaccine development. A Göttingen minipig model was assessed, using pertussis vaccines, to demonstrate that vaccine antigen-specific humoral and cellular responses, including IgG titers, functional antibodies, Th polarization and memory B cells can be assessed in a longitudinal study. A vaccination schedule of priming with a whole cell (DTwP) or an acellular (DTaP) pertussis vaccine was applied in neonatal and infant minipigs followed by boosting with a Tdap acellular vaccine. Single cell RNAsequencing was used to explore the long-term maintenance of immune memory cells and their functionality for the first time in this animal model. DTaP but not DTwP vaccination induced pertussis toxin (PT) neutralizing antibodies. The cellular immune response was also characterized by a distinct Th polarization, with a Th-2-biased response for DTaP and a Th-1/Th-17-biased response for DTwP. No difference in the maintenance of pertussis-specific memory B cells was observed in DTaP- or DTwP-primed animals 6 months post Tdap boost. However, an increase in pertussis-specific T cells was still observed in DTaP primed minipigs, together with up-regulation of genes involved in antigen presentation and interferon pathways. Overall, the minipig model reproduced the humoral and cellular immune responses induced in humans by DTwP vs. DTaP priming, followed by Tdap boosting. Our data suggest that the Göttingen minipig is an attractive preclinical model to predict the long-term immunogenicity of human vaccines against Bordetella pertussis and potentially also vaccines against other pathogens.
Collapse
Affiliation(s)
- Céline Vaure
- Research and External Innovation, Sanofi Pasteur, Marcy l'Etoile, France
| | | | - Virginie Courtois
- Research and External Innovation, Sanofi Pasteur, Marcy l'Etoile, France
| | - Emilie Chautard
- Research and External Innovation, Sanofi Pasteur, Marcy l'Etoile, France
| | - Cyril Dégletagne
- Research and External Innovation, Sanofi Pasteur, Marcy l'Etoile, France
| | - Yuanqing Liu
- Research and External Innovation, Sanofi Pasteur, Marcy l'Etoile, France
| |
Collapse
|
40
|
Broset E, Pardo-Seco J, Kanno AI, Aguilo N, Dacosta AI, Rivero-Calle I, Gonzalo-Asensio J, Locht C, Leite LCC, Martin C, Martinón-Torres F. BCG vaccination improves DTaP immune responses in mice and is associated with lower pertussis incidence in ecological epidemiological studies. EBioMedicine 2021; 65:103254. [PMID: 33711798 PMCID: PMC7960937 DOI: 10.1016/j.ebiom.2021.103254] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The Bacillus Calmette-Guérin (BCG), the only vaccine against tuberculosis (TB) currently in use, has shown beneficial effects against unrelated infections and to enhance immune responses to vaccines. However, there is little evidence regarding the influence of BCG vaccination on pertussis. METHODS Here, we studied the ability of BCG to improve the immune responses to diphtheria, tetanus, and acellular (DTaP) or whole-cell pertussis (DTwP) vaccination in a mouse model. We included MTBVAC, an experimental live-attenuated vaccine derived from Mycobacterium tuberculosis, in our studies to explore if it presents similar heterologous immunity as BCG. Furthermore, we explored the potential effect of routine BCG vaccination on pertussis incidence worldwide. FINDINGS We found that both BCG and MTBVAC when administered before DTaP, triggered Th1 immune responses against diphtheria, tetanus, and pertussis in mice. Immunization with DTaP alone failed to trigger a Th1 response, as measured by the production of IFN-γ. Humoral responses against DTaP antigens were also enhanced by previous immunization with BCG or MTBVAC. Furthermore, exploration of human epidemiological data showed that pertussis incidence was 10-fold lower in countries that use DTaP and BCG compared to countries that use only DTaP. INTERPRETATION BCG vaccination may have a beneficial impact on the protection against pertussis conferred by DTaP. Further randomized controlled trials are needed to properly define the impact of BCG on pertussis incidence in a controlled setting. This could be a major finding that would support changes in immunization policies. FUNDING This work was supported by the Ministry of "Economía y Competitividad"; European Commission H2020 program, "Gobierno de Aragón"; CIBERES; "Fundação Butantan"; Instituto de Salud Carlos III and "Fondo FEDER".
Collapse
Affiliation(s)
- Esther Broset
- Grupo de Genetica de Micobacterias, Departamento de Microbiología y Medicina Preventiva, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragón, Zaragoza 50009, Spain; CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.
| | - Jacobo Pardo-Seco
- Translational Pediatrics and Infectious Diseases, Pediatrics Department, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain; GENVIP Research Group (www.genvip.org), Instituto de Investigación Sanitaria de Santiago (SERGAS), University of Santiago de Compostela, Galicia, Spain
| | - Alex I Kanno
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, Av. Vital Brasil 1500, São Paulo 05503-900, Brazil
| | - Nacho Aguilo
- Grupo de Genetica de Micobacterias, Departamento de Microbiología y Medicina Preventiva, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragón, Zaragoza 50009, Spain; CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Isabel Dacosta
- Translational Pediatrics and Infectious Diseases, Pediatrics Department, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain; GENVIP Research Group (www.genvip.org), Instituto de Investigación Sanitaria de Santiago (SERGAS), University of Santiago de Compostela, Galicia, Spain
| | - Irene Rivero-Calle
- Translational Pediatrics and Infectious Diseases, Pediatrics Department, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain; GENVIP Research Group (www.genvip.org), Instituto de Investigación Sanitaria de Santiago (SERGAS), University of Santiago de Compostela, Galicia, Spain
| | - Jesus Gonzalo-Asensio
- Grupo de Genetica de Micobacterias, Departamento de Microbiología y Medicina Preventiva, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragón, Zaragoza 50009, Spain; CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Zaragoza, Spain
| | - Camille Locht
- Center of Infection and Immunity of Lille, Institut Pasteur de Lille, Lille 59019, France; Inserm U1019, Lille 59019, France; CNRS UMR8204, Lille 59019, France; Univ. Lille, Lille 59019, France
| | - Luciana C C Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, Av. Vital Brasil 1500, São Paulo 05503-900, Brazil
| | - Carlos Martin
- Grupo de Genetica de Micobacterias, Departamento de Microbiología y Medicina Preventiva, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragón, Zaragoza 50009, Spain; CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain; Servicio de Microbiología, Hospital Universitario Miguel Servet, IIS Aragón, Zaragoza, Spain
| | - Federico Martinón-Torres
- Translational Pediatrics and Infectious Diseases, Pediatrics Department, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain; GENVIP Research Group (www.genvip.org), Instituto de Investigación Sanitaria de Santiago (SERGAS), University of Santiago de Compostela, Galicia, Spain
| |
Collapse
|
41
|
Versteegen P, Valente Pinto M, Barkoff AM, van Gageldonk PGM, van de Kassteele J, van Houten MA, Sanders EAM, de Groot R, Diavatopoulos DA, Bibi S, Luoto R, He Q, Buisman AM, Kelly DF, Mertsola J, Berbers GAM. Responses to an acellular pertussis booster vaccination in children, adolescents, and young and older adults: A collaborative study in Finland, the Netherlands, and the United Kingdom. EBioMedicine 2021; 65:103247. [PMID: 33647770 PMCID: PMC7920834 DOI: 10.1016/j.ebiom.2021.103247] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 01/08/2023] Open
Abstract
Background Pertussis can lead to serious disease and even death
in infants. Older adults are more vulnerable to complications as well. In
high-income countries, acellular pertussis vaccines are used for priming
vaccination. In the administration of booster vaccinations to different age
groups and target populations there is a substantial between-country variation.
We investigated the effect of age on the response to acellular pertussis booster
vaccination in three European countries. Methods This phase IV longitudinal intervention study
performed in Finland, the Netherlands and the United Kingdom between October
2017 and January 2019 compared the vaccine responses between healthy
participants of four age groups: children (7–10y), adolescents (11–15y), young
adults (20–34y), and older adults (60–70y). All participants received a
three-component acellular pertussis vaccine. Serum IgG and IgA antibody
concentrations to pertussis antigens at day 0, 28, and 1 year were measured with
a multiplex immunoassay, using pertussis toxin concentrations at day 28 as
primary outcome. This trial is registered with ClinicalTrialsRegister.eu
(2016–003,678–42). Findings Children (n = 109), adolescents
(n = 121), young adults
(n = 74), and older adults
(n = 75) showed high IgG antibody concentrations to
pertussis toxin at day 28 with GMCs of 147 (95% CI 120–181), 161 (95% CI
132–196), 103 (95% CI 80–133), and 121 IU/ml (95% CI 94–155), respectively. A
significant increase in GMCs for vaccine antigens in all age groups by 28 days
was found which had decreased by 1 year. Differences in patterns of IgG GMCs at
28 days and 1 year post-vaccination did not have a consistent relationship to
age. In contrast, IgA antibodies for all antigens increased with age at all
timepoints. Interpretation Acellular pertussis booster vaccination induces
significant serum IgG responses to pertussis antigens across the age range which
are not uniformly less in older adults. Acellular boosters could be considered
for older adults to reduce the health and economic burden of
pertussis.
Collapse
Affiliation(s)
- Pauline Versteegen
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Antonie van Leeuwenhoeklaan 9, Bilthoven 3720 BA, Netherlands
| | - Marta Valente Pinto
- University of Oxford, Department of Paediatrics, Oxford Vaccine Group, Oxford OX3 7LE, United Kingdom
| | - Alex M Barkoff
- University of Turku, Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, Turku 20500, Finland
| | - Pieter G M van Gageldonk
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Antonie van Leeuwenhoeklaan 9, Bilthoven 3720 BA, Netherlands
| | - Jan van de Kassteele
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Antonie van Leeuwenhoeklaan 9, Bilthoven 3720 BA, Netherlands
| | | | - Elisabeth A M Sanders
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Antonie van Leeuwenhoeklaan 9, Bilthoven 3720 BA, Netherlands; Wilhelmina Children's Hospital, Department of Paediatric Immunology and Infectious Diseases, Lundlaan 6, 3584 EA Utrecht, Netherlands
| | - Ronald de Groot
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Nijmegen 6525 GA, Netherlands
| | - Dimitri A Diavatopoulos
- Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Nijmegen 6525 GA, Netherlands
| | - Sagida Bibi
- University of Oxford, Department of Paediatrics, Oxford Vaccine Group, Oxford OX3 7LE, United Kingdom
| | - Raakel Luoto
- University of Turku, Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, Turku 20500, Finland
| | - Qiushui He
- University of Turku, Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, Turku 20500, Finland
| | - Anne-Marie Buisman
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Antonie van Leeuwenhoeklaan 9, Bilthoven 3720 BA, Netherlands
| | - Dominic F Kelly
- University of Oxford, Department of Paediatrics, Oxford Vaccine Group, Oxford OX3 7LE, United Kingdom; Oxford University Hospitals NHS Foundation Trust, Headington, Oxford OX3 9DU, United Kingdom
| | - Jussi Mertsola
- University of Turku, Institute of Biomedicine, Microbiology, Virology and Immunology, and Turku University Hospital, Turku 20500, Finland
| | - Guy A M Berbers
- National Institute for Public Health and the Environment, Centre for Infectious Disease Control, Antonie van Leeuwenhoeklaan 9, Bilthoven 3720 BA, Netherlands.
| |
Collapse
|
42
|
McAlister SM, van den Biggelaar AHJ, Woodman TL, Hutton H, Thornton RB, Richmond PC. An observational study of antibody responses to a primary or subsequent pertussis booster vaccination in Australian healthcare workers. Vaccine 2021; 39:1642-1651. [PMID: 33589299 DOI: 10.1016/j.vaccine.2021.01.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 11/23/2020] [Accepted: 01/16/2021] [Indexed: 11/29/2022]
Abstract
Adult pertussis vaccination is increasingly recommended to control pertussis in the community. However, there is little data on the duration and kinetics of immunity to pertussis boosters in adults. We compared IgG responses to vaccination with a tetanus, low-dose diphtheria, low-dose acellular pertussis (Tdap) booster at 1 week, 1 month and 1 year post-vaccination in whole-cell (wP)-primed Australian paediatric healthcare workers who had received an adult Tdap booster 5-12 years previously, to those who received their first Tdap booster. Tdap vaccination was well tolerated in both groups. Previously boosted adults had significantly higher pre-vaccination IgG concentrations for all vaccine-antigens, and more were seropositive for pertussis toxin (PT)-specific IgG (≥ 5 IU/mL) (69.5%; 95% confidence interval (CI) 59.5-79.5) than adults in the naïve group (45.2%; 95% CI 32.8-57.5). Tdap vaccination significantly increased IgG responses 1 month post-vaccination in both groups. This increase was more rapid in previously boosted than in naïve adults, with geometric mean fold-increases in PT-IgG at 1 week post vaccination of 3.6 (95% CI 2.9-4.3) and 2.6 (95% CI 2.2-3.2), respectively. Antibody waning between 1 month and 1 year post-vaccination was similar between groups for IgG specific to PT and filamentous haemagglutinin (FHA), but was faster for IgG against pertactin (PRN) in the naïve group (GMC ratio 0.36; 95% CI 0.31-0.42) than the previously boosted group (GMC ratio 0.45; 95% CI 0.39-0.50). At baseline, all but one adult had protective IgG titres against tetanus toxin (TT) (≥ 0.1 IU/mL), and 75.6% in the previously boosted and 61.3% in the naïve group had protective IgG titres against diphtheria toxoid (DT) of ≥ 0.1 IU/mL. This study shows that pertussis immune memory is maintained up to 12 years after Tdap vaccination in wP-primed Australian adults. There was no evidence that pertussis immune responses waned faster after a booster dose. These findings support current recommendations of repeating Tdap booster vaccination in paediatric healthcare workers at least every 10 years. Clinical trials registry: ACTRN12615001262594.
Collapse
Affiliation(s)
- Sonia M McAlister
- Vaccine Trials Group, Wesfarmers Centre of Vaccines & Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; Division of Paediatrics, School of Medicine, The University of Western Australia, Perth, Western Australia, Australia.
| | - Anita H J van den Biggelaar
- Vaccine Trials Group, Wesfarmers Centre of Vaccines & Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; Division of Paediatrics, School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Tabitha L Woodman
- Vaccine Trials Group, Wesfarmers Centre of Vaccines & Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; Division of Paediatrics, School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Heidi Hutton
- Vaccine Trials Group, Wesfarmers Centre of Vaccines & Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Ruth B Thornton
- Vaccine Trials Group, Wesfarmers Centre of Vaccines & Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; Division of Paediatrics, School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Peter C Richmond
- Vaccine Trials Group, Wesfarmers Centre of Vaccines & Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; Division of Paediatrics, School of Medicine, The University of Western Australia, Perth, Western Australia, Australia; Departments of Immunology and General Paediatrics, Perth Children's Hospital, Perth, Western Australia, Australia
| |
Collapse
|
43
|
Dubois V, Chatagnon J, Thiriard A, Bauderlique-Le Roy H, Debrie AS, Coutte L, Locht C. Suppression of mucosal Th17 memory responses by acellular pertussis vaccines enhances nasal Bordetella pertussis carriage. NPJ Vaccines 2021; 6:6. [PMID: 33420041 PMCID: PMC7794405 DOI: 10.1038/s41541-020-00270-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Pertussis has made a spectacular rebound in countries that have switched from whole-cell (wPV) to acellular pertussis vaccines (aPV). Here, we show that, unlike wPV, aPV, while protective against lung colonization by Bordetella pertussis (Bp), did not protect BALB/c mice from nasal colonization, but instead substantially prolonged nasal carriage. aPV prevented the natural induction of nasal interleukin-17 (IL-17)-producing and interferon-γ (IFN-γ)-producing CD103+ CD44+ CD69+ CD4+-resident memory T (TRM) cells. IL-17-deficient, but not IFN-γ-deficient, mice failed to clear nasal Bp, indicating a key role of IL-17+ TRM cells in the control of nasal infection. These cells appeared essential for neutrophil recruitment, crucial for clearance of Bp tightly bound to the nasal epithelium. Transfer of IL-17+ TRM cells from Bp-infected mice to IL-17-deficient mice resulted in neutrophil recruitment and protection against nasal colonization. Thus, aPV may have augmented the Bp reservoir by inhibiting natural TRM cell induction and neutrophil recruitment, thereby contributing to the pertussis resurgence.
Collapse
Affiliation(s)
- Violaine Dubois
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Center for Infection and Immunity of Lille, Univ. Lille, 59000, Lille, France.
| | - Jonathan Chatagnon
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Center for Infection and Immunity of Lille, Univ. Lille, 59000, Lille, France
| | - Anaïs Thiriard
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Center for Infection and Immunity of Lille, Univ. Lille, 59000, Lille, France
| | - Hélène Bauderlique-Le Roy
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US41-UMS 2014-PLBS, Univ. Lille, 59000, Lille, France
| | - Anne-Sophie Debrie
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Center for Infection and Immunity of Lille, Univ. Lille, 59000, Lille, France
| | - Loïc Coutte
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Center for Infection and Immunity of Lille, Univ. Lille, 59000, Lille, France
| | - Camille Locht
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Center for Infection and Immunity of Lille, Univ. Lille, 59000, Lille, France
| |
Collapse
|
44
|
Novák J, Jurnečka D, Linhartová I, Holubová J, Staněk O, Štipl D, Dienstbier A, Večerek B, Azevedo N, Provazník J, Beneš V, Šebo P. A Mutation Upstream of the rplN-rpsD Ribosomal Operon Downregulates Bordetella pertussis Virulence Factor Production without Compromising Bacterial Survival within Human Macrophages. mSystems 2020; 5:e00612-20. [PMID: 33293402 PMCID: PMC7742992 DOI: 10.1128/msystems.00612-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/06/2020] [Indexed: 11/20/2022] Open
Abstract
The BvgS/BvgA two-component system controls expression of ∼550 genes of Bordetella pertussis, of which, ∼245 virulence-related genes are positively regulated by the BvgS-phosphorylated transcriptional regulator protein BvgA (BvgA∼P). We found that a single G-to-T nucleotide transversion in the 5'-untranslated region (5'-UTR) of the rplN gene enhanced transcription of the ribosomal protein operon and of the rpoA gene and provoked global dysregulation of B. pertussis genome expression. This comprised overproduction of the alpha subunit (RpoA) of the DNA-dependent RNA polymerase, downregulated BvgA and BvgS protein production, and impaired production and secretion of virulence factors by the mutant. Nonetheless, the mutant survived like the parental bacteria for >2 weeks inside infected primary human macrophages and persisted within infected mouse lungs for a longer period than wild-type B. pertussis These observations suggest that downregulation of virulence factor production by bacteria internalized into host cells may enable persistence of the whooping cough agent in the airways.IMPORTANCE We show that a spontaneous mutation that upregulates transcription of an operon encoding ribosomal proteins and causes overproduction of the downstream-encoded α subunit (RpoA) of RNA polymerase causes global effects on gene expression levels and proteome composition of Bordetella pertussis Nevertheless, the resulting important downregulation of the BvgAS-controlled expression of virulence factors of the whooping cough agent did not compromise its capacity to persist for prolonged periods inside primary human macrophage cells, and it even enhanced its capacity to persist in infected mouse lungs. These observations suggest that the modulation of BvgAS-controlled expression of virulence factors may occur also during natural infections of human airways by Bordetella pertussis and may possibly account for long-term persistence of the pathogen within infected cells of the airways.
Collapse
Affiliation(s)
- Jakub Novák
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - David Jurnečka
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Irena Linhartová
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jana Holubová
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Ondřej Staněk
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Daniel Štipl
- Laboratory of Post-Transcriptional Control of Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Ana Dienstbier
- Laboratory of Post-Transcriptional Control of Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Branislav Večerek
- Laboratory of Post-Transcriptional Control of Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Nayara Azevedo
- Genomics Core Facility, European Molecular Biology Laboratory, Services and Technology Unit, Heidelberg, Germany
| | - Jan Provazník
- Genomics Core Facility, European Molecular Biology Laboratory, Services and Technology Unit, Heidelberg, Germany
| | - Vladimír Beneš
- Genomics Core Facility, European Molecular Biology Laboratory, Services and Technology Unit, Heidelberg, Germany
| | - Peter Šebo
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
45
|
Cole LE, Zhang J, Pacheco KM, Lhéritier P, Anosova NG, Piolat J, Zheng L, Reveneau N. Immunological Distinctions between Acellular and Whole-Cell Pertussis Immunizations of Baboons Persist for at Least One Year after Acellular Vaccine Boosting. Vaccines (Basel) 2020; 8:vaccines8040729. [PMID: 33276673 PMCID: PMC7761625 DOI: 10.3390/vaccines8040729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/19/2020] [Accepted: 11/26/2020] [Indexed: 01/17/2023] Open
Abstract
While both whole-cell (wP) and acellular pertussis (aP) vaccines have been highly effective at reducing the global pertussis disease burden, there are concerns that compared to wP vaccination, the immune responses to aP vaccination may wane more rapidly. To gain insights into the vaccine elicited immune responses, pre-adult baboons were immunized with either aP or wP vaccines, boosted with an aP vaccine, and observed over a nearly two-year period. Priming with a wP vaccine elicited a more Th17-biased response than priming with aP, whereas priming with an aP vaccine led to a more Th2-biased response than priming with wP. These differences were maintained after aP vaccine boost immunizations. Compared to aP, animals primed with a wP vaccine exhibited greater numbers of pertussis specific memory B cells. While aP and wP vaccine priming initially elicited similar levels of anti-pertussis toxin antibody, titers declined more rapidly in aP vaccine primed animals leading to a 4-fold difference. Both wP and aP vaccine immunization could induce serum bactericidal activity (SBA); however, only one wP vaccine immunization was required to elicit SBA while multiple aP vaccine immunizations were required to elicit lower, less durable SBA titers. In conclusion, when compared to aP vaccine, priming with wP vaccine elicits distinct cellular and humoral immune responses that persist after aP vaccine boosting.
Collapse
Affiliation(s)
- Leah E. Cole
- Sanofi Pasteur, Cambridge, MA 02139, USA; (J.Z.); (K.M.P.); (N.G.A.)
- Correspondence: (L.E.C.); (N.R.); Tel.: +1-617-866-4473 (L.E.C.); +33-4-37-66-8510 (N.R.)
| | - Jinrong Zhang
- Sanofi Pasteur, Cambridge, MA 02139, USA; (J.Z.); (K.M.P.); (N.G.A.)
| | - Kristl M. Pacheco
- Sanofi Pasteur, Cambridge, MA 02139, USA; (J.Z.); (K.M.P.); (N.G.A.)
| | | | | | - Julie Piolat
- Sanofi Pasteur, 69280 Marcy L’Etoile, France; (P.L.); (J.P.)
| | | | - Nathalie Reveneau
- Sanofi Pasteur, 69280 Marcy L’Etoile, France; (P.L.); (J.P.)
- Correspondence: (L.E.C.); (N.R.); Tel.: +1-617-866-4473 (L.E.C.); +33-4-37-66-8510 (N.R.)
| |
Collapse
|
46
|
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: 22] [Impact Index Per Article: 5.5] [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
|
47
|
Reche PA. Potential Cross-Reactive Immunity to SARS-CoV-2 From Common Human Pathogens and Vaccines. Front Immunol 2020; 11:586984. [PMID: 33178220 PMCID: PMC7596387 DOI: 10.3389/fimmu.2020.586984] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/25/2020] [Indexed: 12/16/2022] Open
Abstract
The recently emerged SARS-CoV-2 causing the ongoing COVID-19 pandemic is particularly virulent in the elderly while children are largely spared. Here, we explored the potential role of cross-reactive immunity acquired from pediatric vaccinations and exposure to common human pathogens in the protection and pathology of COVID-19. To that end, we sought for peptide matches to SARS-CoV-2 (identity ≥ 80%, in at least eight residues) in the proteomes of 25 human pathogens and in vaccine antigens, and subsequently predicted their T and B cell reactivity to identify potential cross-reactive epitopes. We found that viruses subject to pediatric vaccinations do not contain cross-reactive epitopes with SARS-CoV-2, precluding that they can provide any general protection against COVID-19. Likewise, common viruses including rhinovirus, respiratory syncytial virus, influenza virus, and several herpesviruses are also poor or null sources of cross-reactive immunity to SARS-CoV-2, discarding that immunological memory against these viruses can have any general protective or pathological role in COVID-19. In contrast, we found combination vaccines for treating diphtheria, tetanus, and pertussis infectious diseases (DTP vaccine) to be significant sources of potential cross-reactive immunity to SARS-CoV-2. DTP cross-reactive epitopes with SARS-CoV-2 include numerous CD8 and CD4 T cell epitopes with broad population protection coverage and potentially neutralizing B cell epitopes in SARS-CoV-2 Spike protein. Worldwide, children receive several DTP vaccinations, including three-four doses the first year of life and one at 4-6 years of age. Moreover, a low antigenic Tdap dose is also given at ages 9-14. Thereby, children may well be protected from SARS-CoV-2 through cross-reactive immunity elicited by DTP vaccinations, supporting testing in the general population to prevent COVID-19.
Collapse
Affiliation(s)
- Pedro A. Reche
- Department of Immunology & O2, Faculty of Medicine, University Complutense of Madrid, Madrid, Spain
| |
Collapse
|
48
|
da Silva Antunes R, Quiambao LG, Soldevila F, Sutherland A, Peters B, Sette A. Lack of evidence supporting a role of IFN-β and TGF-β in differential polarization of Bordetella pertussis specific-T cell responses. Cytokine 2020; 137:155313. [PMID: 33002739 DOI: 10.1016/j.cyto.2020.155313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 10/23/2022]
Abstract
Bordetella Pertussis (BP) vaccine-induced immunity is waning worldwide despite excellent vaccine coverage. Replacement of the whole-cell inactivated vaccine (wP) by an acellular subunit vaccine (aP) is thought to play a major role and to be associated with the recurrence of whooping cough. Previously, we detected that the polarization towards a Th2 and Th1/Th17 response in aP and wP vaccinees, respectively, persists upon aP boosting in adolescents and adults. Additionally, IL-9 and TGF-β were found to be up-regulated in aP-primed donors and network analysis further identified IFN-β as a potential upstream regulator of IL-17 and IL-9. Based on these findings, we hypothesized that IFN-β produced following aP vaccination may lead to increased IL-9 and decreased IL-17 production. Also, due to the well characterized role of TGF-β in both Th17 and Th9 differentiation, we put forth that TGF-β addition to BP-stimulated CD4 + T cells might modulate IL-17 and IL-9 production. To test this hypothesis, we stimulated in vitro cultures of PBMC or isolated naive CD4 + T cells from aP vs wP donors with a pool of BP epitopes and assessed the effect of IFN-β or TGF-β in proliferative responses as well as in the cytokine secretion of IL-4, IL-9, IL-17, and IFN-γ. IFN-β reduced BP-specific proliferation in PBMC as well as cytokine production but increased IL-9, IL-4, and IFN-γ cytokines in naïve CD4 + T cells. These effects were independent of the childhood vaccination received by the donors. Similarly, TGF-β reduced BP-specific proliferation in PBMC but induced proliferation in naïve CD4 + T cells. However, stimulation was associated with a generalized inhibition of cytokine production regardless of the original aP or wP vaccination received by the donors. Our study suggests that key T cell functions such as cytokine secretion are under the control of antigen stimulation and environmental cues but molecular pathways different than the ones investigated here might underlie the long-lasting differential cytokine production associated with aP- vs wP-priming in childhood vaccination.
Collapse
Affiliation(s)
| | - Lorenzo G Quiambao
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Ferran Soldevila
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Aaron Sutherland
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States; School of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States; School of Medicine, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
49
|
Lesne E, Cavell BE, Freire-Martin I, Persaud R, Alexander F, Taylor S, Matheson M, van Els CACM, Gorringe A. Acellular Pertussis Vaccines Induce Anti-pertactin Bactericidal Antibodies Which Drives the Emergence of Pertactin-Negative Strains. Front Microbiol 2020; 11:2108. [PMID: 32983069 PMCID: PMC7481377 DOI: 10.3389/fmicb.2020.02108] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022] Open
Abstract
Despite high vaccination coverage, Bordetella pertussis the causative agent of whooping cough is still a health concern worldwide. A resurgence of pertussis cases has been reported, particularly in countries using acellular vaccines with waning immunity and pathogen adaptation thought to be responsible. A better understanding of protective immune responses is needed for the development of improved vaccines. In our study, B. pertussis strain B1917 variants presenting a single gene deletion were generated to analyze the role of vaccine components or candidate vaccine antigens as targets for bactericidal antibodies generated after acellular vaccination or natural infection. Our results show that acellular vaccination generates bactericidal antibodies that are only directed against pertactin. Serum bactericidal assay performed with convalescent samples show that disease induces bactericidal antibodies against Prn but against other antigen(s) as well. Four candidate vaccine antigens (CyaA, Vag8, BrkA, and TcfA) have been studied but were not targets for complement-mediated bactericidal antibodies after natural infection. We confirm that Vag8 and BrkA are involved in complement resistance and would be targeted by blocking antibodies. Our study suggests that the emergence and the widespread circulation of Prn-deficient strains is driven by acellular vaccination and the generation of bactericidal antibodies targeting Prn.
Collapse
Affiliation(s)
- Elodie Lesne
- Public Health England, Porton Down, United Kingdom
| | | | | | - Ruby Persaud
- Public Health England, Porton Down, United Kingdom
| | | | | | | | - Cécile A. C. M. van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | | |
Collapse
|
50
|
Perez Chacon G, Estcourt M, Ramsay J, Brennan-Jones CG, Richmond P, Holt P, Snelling T. Whole-cell pertussis vaccine in early infancy for the prevention of allergy. Hippokratia 2020. [DOI: 10.1002/14651858.cd013682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Gladymar Perez Chacon
- Wesfarmers Centre of Vaccines and Infectious Diseases; Telethon Kids Institute; Perth Australia
- School of Public Health; Curtin University; Perth Australia
| | - Marie Estcourt
- Sydney School of Public Health, Faculty of Medicine and Health; University of Sydney; Camperdown Australia
| | - Jessica Ramsay
- Wesfarmers Centre of Vaccines and Infectious Diseases; Telethon Kids Institute; Perth Australia
| | | | - Peter Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases; Telethon Kids Institute; Perth Australia
- Division of Paediatrics; The University of Western Australia; Perth Australia
| | - Patrick Holt
- Telethon Kids Institute; The University of Western Australia; Perth Australia
| | - Tom Snelling
- Wesfarmers Centre of Vaccines and Infectious Diseases; Telethon Kids Institute; Perth Australia
- Sydney School of Public Health, Faculty of Medicine and Health; University of Sydney; Camperdown Australia
| |
Collapse
|