1
|
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
|
2
|
Szinger D, Berki T, Drenjančević I, Samardzic S, Zelić M, Sikora M, Požgain A, Markovics Á, Farkas N, Németh P, Böröcz K. Raising Epidemiological Awareness: Assessment of Measles/MMR Susceptibility in Highly Vaccinated Clusters within the Hungarian and Croatian Population-A Sero-Surveillance Analysis. Vaccines (Basel) 2024; 12:486. [PMID: 38793737 PMCID: PMC11125914 DOI: 10.3390/vaccines12050486] [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: 04/09/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Perceptions of the complete eradication of vaccine-preventable diseases such as measles, mumps, and rubella (MMR) may foster complacency and compromise vaccination efforts. Decreased measles vaccination rates during the COVID-19 pandemic have heightened the risk of outbreaks, even in adequately vaccinated populations. To address this, we have aligned with ECDC recommendations, leveraging previous cross-border sero-epidemiological assessments between Pécs, Hungary, and Osijek, Croatia, to identify latent risk groups and uncover potential parallels between our nations. Testing 2680 Hungarian and 1764 Croatian serum samples for anti-MMR IgG via ELISAs revealed anti-measles seropositivity ratios below expectations in Croatian cohorts aged ~20-30 (75.7%), ~30-40 (77.5%) and ~40-50 years (73.3%). Similarly, Hungarian samples also showed suboptimal seropositivity ratios in the ~30-40 (80.9%) and ~40-50 (87.3%) age groups. Considering mumps- and rubella-associated seropositivity trends, in both examined populations, individuals aged ~30-50 years exhibited the highest vulnerability. Additionally, we noted congruent seropositivity trends across both countries, despite distinct immunization and epidemiological contexts. Therefore, we propose expanding research to encompass the intricate dynamics of vaccination, including waning long-term immunity. This understanding could facilitate targeted interventions and bolster public awareness. Our findings underscore persistent challenges in attaining robust immunity against measles despite vaccination endeavors.
Collapse
Affiliation(s)
- Dávid Szinger
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary; (D.S.); (T.B.); (P.N.)
| | - Timea Berki
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary; (D.S.); (T.B.); (P.N.)
| | - Ines Drenjančević
- Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
- Scientific Centre for Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Senka Samardzic
- Department of Public Health, Teaching Institute of Public Health for The Osijek-Baranja County, 31000 Osijek, Croatia; (S.S.); (M.Z.); (M.S.); (A.P.)
| | - Marija Zelić
- Department of Public Health, Teaching Institute of Public Health for The Osijek-Baranja County, 31000 Osijek, Croatia; (S.S.); (M.Z.); (M.S.); (A.P.)
| | - Magdalena Sikora
- Department of Public Health, Teaching Institute of Public Health for The Osijek-Baranja County, 31000 Osijek, Croatia; (S.S.); (M.Z.); (M.S.); (A.P.)
| | - Arlen Požgain
- Department of Public Health, Teaching Institute of Public Health for The Osijek-Baranja County, 31000 Osijek, Croatia; (S.S.); (M.Z.); (M.S.); (A.P.)
- Department of Microbiology, Parasitology and Clinical Laboratory Diagnostics, Medical Faculty of Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Ákos Markovics
- Department of General and Physical Chemistry, Faculty of Natural Sciences, University of Pécs, 7622 Pécs, Hungary;
| | - Nelli Farkas
- Department of Bioanalysis, Medical School, University of Pécs, Szigeti u. 12, 7643 Pécs, Hungary;
| | - Péter Németh
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary; (D.S.); (T.B.); (P.N.)
| | - Katalin Böröcz
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary; (D.S.); (T.B.); (P.N.)
| |
Collapse
|
3
|
Essa S, Safar HA, Raghupathy R. Cytokine responses to major human Cytomegalovirus antigens in mouse model. Cytokine 2024; 176:156546. [PMID: 38359558 DOI: 10.1016/j.cyto.2024.156546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/30/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Human cytomegalovirus (CMV) continues to be a source of severe complications in immunologically immature and immunocompromised hosts. Effective CMV vaccines that help diminish CMV disease in transplant patients and avoid congenital infection are essential. Though the exact roles of defense mechanisms are unidentified, virus-specific antibodies and cytokine responses are known to be involved in controlling CMV infections. Identifying the CMV antigens that trigger these protective immune responses will help us choose the most suitable CMV-related proteins for future vaccines. CMV envelope glycoprotein B (UL55/gB), matrix proteins (UL83/pp65, UL99/pp28, UL32/pp150), and assembly protein UL80a/pp38 are known to be targets for antiviral immune responses. We immunized mice intraperitoneally with these five CMV-related proteins for their ability to induce specific antibody responses and cytokine production in a mouse model. We observed a significant CMV-antigen-specific antibody response to UL80a/pp38 and UL83/pp65 (E/C>2.0). Mice immunized with UL80a/pp38 had significantly higher concentrations of GM-CSF, IFN-γ, IL-2, IL-4, IL-5, and IL-17A (p<0.05). Mice immunized with UL83/pp65 showed significantly higher concentrations of GM-CSF, IFN-γ, IL-2 IL-4, IL-10, IL-12, IL-17A, and TNF-α. Ratios of Th1 to Th2 cytokines revealed a Th1 cytokine bias in mice immunized with UL80a/pp38, UL83/pp65, UL32/pp150, and UL55/gB. We suggest that stimulation with multiple CMV-related proteins, which include UL80a/pp38, UL83/pp65, UL32/pp150, and UL55/gB antigens, will allow both humoral and cellular immune responses to be efficiently activated, thus serving as appropriate CMV antigens for future novel vaccines and immune-based therapeutic design.
Collapse
Affiliation(s)
- Sahar Essa
- Department of Microbiology, College of Medicine, Kuwait University, Kuwait City, Kuwait.
| | - Hussain A Safar
- OMICS Research Unit, Health Science Center, Kuwait University, Kuwait City, Kuwait.
| | - Raj Raghupathy
- Department of Microbiology, College of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
4
|
Adhikari J, Heffernan J, Edeling M, Fernandez E, Jethva PN, Diamond MS, Fremont DH, Gross ML. Epitope Mapping of Japanese Encephalitis Virus Neutralizing Antibodies by Native Mass Spectrometry and Hydrogen/Deuterium Exchange. Biomolecules 2024; 14:374. [PMID: 38540792 PMCID: PMC10967844 DOI: 10.3390/biom14030374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 06/27/2024] Open
Abstract
Japanese encephalitis virus (JEV) remains a global public health concern due to its epidemiological distribution and the existence of multiple strains. Neutralizing antibodies against this infection have shown efficacy in in vivo studies. Thus, elucidation of the epitopes of neutralizing antibodies can aid in the design and development of effective vaccines against different strains of JEV. Here, we describe a combination of native mass spectrometry (native-MS) and hydrogen/deuterium exchange mass spectrometry (HDX-MS) to complete screening of eight mouse monoclonal antibodies (MAbs) against JEV E-DIII to identify epitope regions. Native-MS was used as a first pass to identify the antibodies that formed a complex with the target antigen, and it revealed that seven of the eight monoclonal antibodies underwent binding. Native mass spectra of a MAb (JEV-27) known to be non-binding showed broad native-MS peaks and poor signal, suggesting the protein is a mixture or that there are impurities in the sample. We followed native-MS with HDX-MS to locate the binding sites for several of the complex-forming antibodies. This combination of two mass spectrometry-based approaches should be generally applicable and particularly suitable for screening of antigen-antibody and other protein-protein interactions when other traditional approaches give unclear results or are difficult, unavailable, or need to be validated.
Collapse
Affiliation(s)
- Jagat Adhikari
- Department of Chemistry, Washington University in St. Louis, Saint Louis, MO 63130, USA; (J.A.); (P.N.J.)
| | - James Heffernan
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63130, USA; (J.H.); (M.E.); (E.F.); (M.S.D.); (D.H.F.)
| | - Melissa Edeling
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63130, USA; (J.H.); (M.E.); (E.F.); (M.S.D.); (D.H.F.)
| | - Estefania Fernandez
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63130, USA; (J.H.); (M.E.); (E.F.); (M.S.D.); (D.H.F.)
| | - Prashant N. Jethva
- Department of Chemistry, Washington University in St. Louis, Saint Louis, MO 63130, USA; (J.A.); (P.N.J.)
| | - Michael S. Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63130, USA; (J.H.); (M.E.); (E.F.); (M.S.D.); (D.H.F.)
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63130, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63130, USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63130, USA
| | - Daved H. Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63130, USA; (J.H.); (M.E.); (E.F.); (M.S.D.); (D.H.F.)
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63130, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO 63130, USA
| | - Michael L. Gross
- Department of Chemistry, Washington University in St. Louis, Saint Louis, MO 63130, USA; (J.A.); (P.N.J.)
| |
Collapse
|
5
|
van Schaik EJ, Fratzke AP, Gregory AE, Dumaine JE, Samuel JE. Vaccine development: obligate intracellular bacteria new tools, old pathogens: the current state of vaccines against obligate intracellular bacteria. Front Cell Infect Microbiol 2024; 14:1282183. [PMID: 38567021 PMCID: PMC10985213 DOI: 10.3389/fcimb.2024.1282183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Obligate intracellular bacteria have remained those for which effective vaccines are unavailable, mostly because protection does not solely rely on an antibody response. Effective antibody-based vaccines, however, have been developed against extracellular bacteria pathogens or toxins. Additionally, obligate intracellular bacteria have evolved many mechanisms to subvert the immune response, making vaccine development complex. Much of what we know about protective immunity for these pathogens has been determined using infection-resolved cases and animal models that mimic disease. These studies have laid the groundwork for antigen discovery, which, combined with recent advances in vaccinology, should allow for the development of safe and efficacious vaccines. Successful vaccines against obligate intracellular bacteria should elicit potent T cell memory responses, in addition to humoral responses. Furthermore, they ought to be designed to specifically induce strong cytotoxic CD8+ T cell responses for protective immunity. This review will describe what we know about the potentially protective immune responses to this group of bacteria. Additionally, we will argue that the novel delivery platforms used during the Sars-CoV-2 pandemic should be excellent candidates to produce protective immunity once antigens are discovered. We will then look more specifically into the vaccine development for Rickettsiaceae, Coxiella burnetti, and Anaplasmataceae from infancy until today. We have not included Chlamydia trachomatis in this review because of the many vaccine related reviews that have been written in recent years.
Collapse
Affiliation(s)
- E J van Schaik
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Medical Research and Education Building, Bryan, TX, United States
| | - A P Fratzke
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Medical Research and Education Building, Bryan, TX, United States
- Charles River Laboratories, Reno, NV, United States
| | - A E Gregory
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Medical Research and Education Building, Bryan, TX, United States
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Jennifer E Dumaine
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Medical Research and Education Building, Bryan, TX, United States
| | - J E Samuel
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Medical Research and Education Building, Bryan, TX, United States
- Department of Veterinary Pathobiology, School of Veterinary Medicine, Texas A&M University (TAMU), College Station, TX, United States
| |
Collapse
|
6
|
deCamp AC, Corcoran MM, Fulp WJ, Willis JR, Cottrell CA, Bader DLV, Kalyuzhniy O, Leggat DJ, Cohen KW, Hyrien O, Menis S, Finak G, Ballweber-Fleming L, Srikanth A, Plyler JR, Rahaman F, Lombardo A, Philiponis V, Whaley RE, Seese A, Brand J, Ruppel AM, Hoyland W, Mahoney CR, Cagigi A, Taylor A, Brown DM, Ambrozak DR, Sincomb T, Mullen TM, Maenza J, Kolokythas O, Khati N, Bethony J, Roederer M, Diemert D, Koup RA, Laufer DS, McElrath JM, McDermott AB, Karlsson Hedestam GB, Schief WR. Human immunoglobulin gene allelic variation impacts germline-targeting vaccine priming. NPJ Vaccines 2024; 9:58. [PMID: 38467663 PMCID: PMC11384754 DOI: 10.1038/s41541-024-00811-5] [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: 04/25/2023] [Accepted: 01/26/2024] [Indexed: 03/13/2024] Open
Abstract
Vaccine priming immunogens that activate germline precursors for broadly neutralizing antibodies (bnAbs) have promise for development of precision vaccines against major human pathogens. In a clinical trial of the eOD-GT8 60mer germline-targeting immunogen, higher frequencies of vaccine-induced VRC01-class bnAb-precursor B cells were observed in the high dose compared to the low dose group. Through immunoglobulin heavy chain variable (IGHV) genotyping, statistical modeling, quantification of IGHV1-2 allele usage and B cell frequencies in the naive repertoire for each trial participant, and antibody affinity analyses, we found that the difference between dose groups in VRC01-class response frequency was best explained by IGHV1-2 genotype rather than dose and was most likely due to differences in IGHV1-2 B cell frequencies for different genotypes. The results demonstrate the need to define population-level immunoglobulin allelic variations when designing germline-targeting immunogens and evaluating them in clinical trials.
Collapse
Grants
- INV-008352 Bill & Melinda Gates Foundation
- INV-007371 Bill & Melinda Gates Foundation
- INV-008017 Bill & Melinda Gates Foundation
- UM1 AI069481 NIAID NIH HHS
- UM1 AI068635 NIAID NIH HHS
- U19 AI128914 NIAID NIH HHS
- P01 AI094419 NIAID NIH HHS
- UM1 AI144462 NIAID NIH HHS
- INV-007522 Bill & Melinda Gates Foundation
- INV-008813 Bill & Melinda Gates Foundation
- INV-007375 Bill & Melinda Gates Foundation
- UM1 AI068618 NIAID NIH HHS
- INV-032929 Bill & Melinda Gates Foundation
- Funding: This work was supported by the Bill and Melinda Gates Foundation Collaboration for AIDS Vaccine Discovery (CCVIMC INV-007371 to R.A.K., A.B.M., and M.J.M.; VISC INV-008017 and INV-032929 to A.C.D.; VxPDC INV-008352 and INV-007375 to IAVI; and NAC INV-007522 and INV-008813 to W.R.S.), IAVI (including IAVI 167627819 to M.J.M. and other support to W.R.S.), the IAVI Neutralizing Antibody Center (NAC) to W.R.S., National Institute of Allergy and Infectious Diseases (NIAID) P01 AI094419 (HIVRAD Optimizing HIV immunogen-BCR interactions for vaccine development") (to W.R.S.), UM1 Al100663 (Scripps Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery) and UM1 AI144462 (Scripps Consortium for HIV/AIDS Vaccine Development) (to W.R.S. and M.J.M.); and UM1AI069481 (Seattle-Lausanne CTU), U19AI128914 (HIPC), and UM1AI068618 (HVTN LC) to M.J.M.; by the Ragon Institute of MGH, MIT, and Harvard (to W.R.S.) and by the Swedish Research Council (grant #2017-00968) to GKH.
Collapse
Affiliation(s)
- Allan C deCamp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA.
| | - Martin M Corcoran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - William J Fulp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Jordan R Willis
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Center for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Christopher A Cottrell
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Center for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Daniel L V Bader
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Center for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Oleksandr Kalyuzhniy
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Center for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - David J Leggat
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kristen W Cohen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Sergey Menis
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Center for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Greg Finak
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Lamar Ballweber-Fleming
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Abhinaya Srikanth
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jason R Plyler
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Farhad Rahaman
- IAVI, 125 Broad Street, 9th floor, New York, NY, 10004, USA
| | | | | | - Rachael E Whaley
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Aaron Seese
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Joshua Brand
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alexis M Ruppel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wesley Hoyland
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Celia R Mahoney
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Alberto Cagigi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alison Taylor
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David M Brown
- The Foundation for the National Institutes of Health, North Bethesda, MD, USA
| | - David R Ambrozak
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Troy Sincomb
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Center for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Tina-Marie Mullen
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Center for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Janine Maenza
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Orpheus Kolokythas
- Department of Radiology, University of Washington, Seattle, WA, 98195, USA
| | - Nadia Khati
- Department of Radiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Jeffrey Bethony
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David Diemert
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dagna S Laufer
- IAVI, 125 Broad Street, 9th floor, New York, NY, 10004, USA
| | - Juliana M McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - William R Schief
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- Center for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02139, USA.
- Moderna Inc., Cambridge, MA, 02139, USA.
| |
Collapse
|
7
|
De Weerdt L, Thiriard A, Leuridan E, Marchant A, Maertens K. Immunogenicity at delivery after Tdap vaccination in successive pregnancies. Front Immunol 2024; 15:1360201. [PMID: 38464513 PMCID: PMC10920275 DOI: 10.3389/fimmu.2024.1360201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
Background Tetanus, diphtheria, acellular pertussis (Tdap) vaccination is recommended to be administered in every pregnancy. Although the safety of this strategy has been confirmed, the immunogenicity of Tdap vaccination in two successive pregnancies has not yet been described. This study investigated Tdap-specific immunity levels and transplacental transfer in two successive pregnancies after repeated Tdap-vaccination. Methods Women enrolled in prior studies on Tdap vaccination during pregnancy were invited to participate in a follow-up study if they became pregnant again. Women who received a Tdap vaccine in both pregnancies were considered for this analysis. Tdap-specific total IgG and IgG subclasses were measured with a multiplex immunoassay. Results In total, 27 participants with a mean interval between deliveries of 2.4 years were included in the analysis. In maternal serum, Tdap-specific total IgG levels were comparable at both deliveries whereas in cord serum, all Tdap-specific total IgG antibody levels were reduced at the second compared to the first delivery. This was largely reflected in the IgG1 levels in maternal and cord serum. Transplacental transfer ratios of total IgG and IgG1 were also mostly reduced in the second compared to the first pregnancy. Conclusion This study reports for the first time Tdap-specific total IgG and IgG subclass levels and transfer ratios after repeated Tdap vaccination in successive pregnancies. We found reduced transfer of most Tdap-specific IgG and IgG1 antibodies in the successive pregnancy. As pertussis-specific antibodies wane quickly, Tdap vaccination in each pregnancy remains beneficial. However, more research is needed to understand the impact of closely spaced booster doses during pregnancy on early infant protection against pertussis.
Collapse
Affiliation(s)
- Louise De Weerdt
- Centre for the Evaluation of Vaccination, Vaccine and Infectious Diseases Institute, University of Antwerp, Antwerp, Belgium
| | - Anaïs Thiriard
- European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium
| | - Elke Leuridan
- Centre for the Evaluation of Vaccination, Vaccine and Infectious Diseases Institute, University of Antwerp, Antwerp, Belgium
| | - Arnaud Marchant
- European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium
| | - Kirsten Maertens
- Centre for the Evaluation of Vaccination, Vaccine and Infectious Diseases Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
8
|
Knuutila A, Dalby T, Ahvenainen N, Barkoff AM, Jørgensen CS, Fuursted K, Mertsola J, He Q. Antibody avidity to pertussis toxin after acellular pertussis vaccination and infection. Emerg Microbes Infect 2023; 12:e2174782. [PMID: 36715361 PMCID: PMC9936998 DOI: 10.1080/22221751.2023.2174782] [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] [Indexed: 01/31/2023]
Abstract
Pertussis toxin (PT) is a unique virulence factor of Bordetella pertussis, and therefore a key component of acellular pertussis vaccines. Although immunity after infection seems to persist longer than after vaccination, the exact mechanisms are not fully known. In this study the overall binding strength (avidity) of anti-PT IgG antibodies was compared after acellular booster vaccination and infection, as a parameter to evaluate long-lasting protection.Danish and Finnish serum samples from a total of 134 serologically confirmed patients and 112 children who received acellular booster vaccines were included in this study. The concentration of anti-PT IgG was first determined by ELISA, followed by two separate ELISAs to evaluate antibody avidity: either with a dilution series of urea as a bond-breaking agent of antibody and antigen binding and a constant anti-PT IgG concentration between the samples or with a constant dilution ratio of sera and detergent. In addition to urea, the use of diethylamine and ammonium thiocyanate as disruptive agents were first compared between each other.A strong Spearman correlation (R > 0.801) was noted between avidity and concentration of anti-PT IgG antibodies if a constant serum dilution method was used, and avidity was noted to be higher in patients in comparison to vaccinees in Denmark, but not in Finland. However, no correlation between antibody concentration and avidity was found if a constant anti-PT IgG concentration was used (R = -0.157). With this method, avidity after vaccination was significantly higher in comparison to that after infection in both Danish and Finnish subjects (p < 0.01). A shorter time since the latest booster vaccination was found to affect avidity positively on the next PT-antigen exposure with either vaccination or infection.
Collapse
Affiliation(s)
- Aapo Knuutila
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Tine Dalby
- Statens Serum Institut, Copenhagen, Denmark
| | | | | | | | | | - Jussi Mertsola
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Qiushui He
- Institute of Biomedicine, University of Turku, Turku, Finland,InFLAMES Research Flagship Center, University of Turku, Turku, Finland, Qiushui He
| |
Collapse
|
9
|
Febriani Y, Mansour T, Sadarangani M, Ulanova M, Amaral K, Halperin SA, De Serres G, Racine É, Brousseau N. Tdap vaccine in pregnancy and immunogenicity of pertussis and pneumococcal vaccines in children: What is the impact of different immunization schedules? Vaccine 2023; 41:6745-6753. [PMID: 37816653 DOI: 10.1016/j.vaccine.2023.09.063] [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: 02/28/2023] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND In 2019, the 3 + 1 schedule for children's vaccination (2-4-6-18 months old) was changed for a reduced 2 + 1 schedule (2-4-12 months old) in Quebec, Canada. We compared the post-booster anti-pertussis and anti-pneumococcus IgG antibody concentrations among children of Tdap-vaccinated and unvaccinated mothers for different vaccine schedules and vaccine formulations. METHODS We conducted an observational cohort study. An invitation letter to potential participants was provided during a routine vaccination visit. Children's blood samples were analyzed post-booster at 13 (2 + 1 schedule) or 19 (3 + 1 schedule) months of age for antibodies against pertussis antigens (pertussis toxin (PT), filamentous hemagglutinin (FHA) and pertactin (PRN)) and pneumococcal antigens (serotypes 4, 18C, 19A, and 19F). IgG concentrations among children of Tdap-vaccinated and unvaccinated mothers for each vaccination schedule were compared using geometric mean concentrations (GMCs) and GMC ratios (GMRs), adjusting for potentially immune-response-influencing factors (aGMR). Serotype-specific pneumococcal seroprotection rates were also compared. RESULTS A total of 360 children were included for pertussis analysis and 248 for pneumococcal analysis. For the 2 + 1 schedule, 13-month-old children of Tdap-vaccinated mothers had lower GMCs against PT, FHA, and PRN, with aGMR (95 %CI) of 0.77 (0.65-0.90), 0.66 (0.55-0.79), 0.72 (0.52-0.99), respectively. For the 3 + 1 schedule, at 19 months old, the interference appeared to be attenuated (higher aGMR values). GMCs against PT were slightly higher in the 3 + 1 than the 2 + 1 schedule: 126.5 IU/ml vs 91.6 IU/ml; aGMR = 1.27. GMCs against PT, FHA and PRN were slightly higher among children who received Infanrix hexa® compared to those who received Pediacel® at 12 months old. For pneumococcal antibodies, at 13 months old, there was no strong evidence of immune interference in children of Tdap-vaccinated mothers. CONCLUSION Infant vaccination schedule may influence immune interference associated with maternal Tdap vaccination. More studies are needed to assess the clinical impact of this interference on children's protection.
Collapse
Affiliation(s)
- Yossi Febriani
- Centre Hospitalier Universitaire (CHU) de Québec-Université Laval Research Center, Quebec City, Quebec, Canada
| | - Thowiba Mansour
- Centre Hospitalier Universitaire (CHU) de Québec-Université Laval Research Center, Quebec City, Quebec, Canada
| | - Manish Sadarangani
- BC Children's Hospital Research Institute, Vaccine Evaluation Center, Vancouver, British Columbia, Canada; University of British Columbia, Department of Pediatrics, Vancouver, British Columbia, Canada
| | | | - Kyle Amaral
- BC Children's Hospital Research Institute, Vaccine Evaluation Center, Vancouver, British Columbia, Canada; University of British Columbia, Department of Medicine, Vancouver, British Columbia, Canada
| | - Scott A Halperin
- Canadian Center for Vaccinology, Departments of Pediatrics and Microbiology and Immunology, Dalhousie University and IWK Health, Halifax, Nova Scotia, Canada
| | - Gaston De Serres
- Centre Hospitalier Universitaire (CHU) de Québec-Université Laval Research Center, Quebec City, Quebec, Canada; Institut national de santé publique du Québec, Biological and Occupational Risks, Quebec City, Quebec, Canada; Laval University, Department of Social and Preventive Medicine, Faculty of Medicine, Quebec City, Quebec, Canada
| | - Étienne Racine
- Institut national de santé publique du Québec, Biological and Occupational Risks, Quebec City, Quebec, Canada
| | - Nicholas Brousseau
- Centre Hospitalier Universitaire (CHU) de Québec-Université Laval Research Center, Quebec City, Quebec, Canada; Institut national de santé publique du Québec, Biological and Occupational Risks, Quebec City, Quebec, Canada; Laval University, Department of Social and Preventive Medicine, Faculty of Medicine, Quebec City, Quebec, Canada.
| |
Collapse
|
10
|
Szinger D, Berki T, Németh P, Erdo-Bonyar S, Simon D, Drenjančević I, Samardzic S, Zelić M, Sikora M, Požgain A, Böröcz K. Following Natural Autoantibodies: Further Immunoserological Evidence Regarding Their Silent Plasticity and Engagement in Immune Activation. Int J Mol Sci 2023; 24:14961. [PMID: 37834409 PMCID: PMC10573785 DOI: 10.3390/ijms241914961] [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: 08/29/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
Contradictory reports are available on vaccine-associated hyperstimulation of the immune system, provoking the formation of pathological autoantibodies. Despite being interconnected within the same network, the role of the quieter, yet important non-pathological and natural autoantibodies (nAAbs) is less defined. We hypothesize that upon a prompt immunological trigger, physiological nAAbs also exhibit a moderate plasticity. We investigated their inducibility through aged and recent antigenic triggers. Anti-viral antibodies (anti-MMR n = 1739 and anti-SARS-CoV-2 IgG n = 330) and nAAbs (anti-citrate synthase IgG, IgM n = 1739) were measured by in-house and commercial ELISAs using Croatian (Osijek) anonymous samples with documented vaccination backgrounds. The results were subsequently compared for statistical evaluation. Interestingly, the IgM isotype nAAb showed a statistically significant connection with anti-MMR IgG seropositivity (p < 0.001 in all cases), while IgG isotype nAAb levels were elevated in association with anti-SARS CoV-2 specific seropositivity (p = 0.019) and in heterogeneous vaccine regimen recipients (unvaccinated controls vector/mRNA vaccines p = 0.002). Increasing evidence supports the interplay between immune activation and the dynamic expansion of nAAbs. Consequently, further questions may emerge regarding the ability of nAAbs silently shaping the effectiveness of immunization. We suggest re-evaluating the impact of nAAbs on the complex functioning of the immunological network.
Collapse
Affiliation(s)
- David Szinger
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary; (D.S.)
| | - Timea Berki
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary; (D.S.)
| | - Péter Németh
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary; (D.S.)
| | - Szabina Erdo-Bonyar
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary; (D.S.)
| | - Diana Simon
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary; (D.S.)
| | - Ines Drenjančević
- Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
- Scientific Centre for Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Senka Samardzic
- Department of Public Health, Teaching Institute of Public Health for The Osijek-Baranja County, 31000 Osijek, Croatia
| | - Marija Zelić
- Department of Public Health, Teaching Institute of Public Health for The Osijek-Baranja County, 31000 Osijek, Croatia
| | - Magdalena Sikora
- Department of Public Health, Teaching Institute of Public Health for The Osijek-Baranja County, 31000 Osijek, Croatia
| | - Arlen Požgain
- Department of Public Health, Teaching Institute of Public Health for The Osijek-Baranja County, 31000 Osijek, Croatia
- Department of Microbiology, Parasitology, and Clinical Laboratory Diagnostics, Medical Faculty of Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Katalin Böröcz
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary; (D.S.)
| |
Collapse
|
11
|
Cheuvart B, Callegaro A, Rosillon D, Meyer N, Guignard A. Effectiveness of maternal immunisation with a three-component acellular pertussis vaccine at preventing pertussis in infants in the United States: Post-hoc analysis of a case-control study using Bayesian dynamic borrowing. Vaccine 2023; 41:5805-5812. [PMID: 37635001 DOI: 10.1016/j.vaccine.2023.07.057] [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: 03/24/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Immunisation during pregnancy with a tetanus toxoid, reduced diphtheria toxoid and acellular pertussis (Tdap) vaccine can protect infants against pertussis between birth and paediatric vaccination. We aimed to estimate the vaccine effectiveness (VE) of third-trimester pregnancy immunisation with the three-component acellular pertussis (Td3ap) vaccine at preventing pertussis in infants <2 months in the United States (US), to support a label update. METHODS We performed a post-hoc sub-analysis of a case-control study conducted in six US Emerging Infections Program Network states between 2011 and 2014. Our analysis included only cases and controls whose mothers were either vaccinated with Td3ap or did not receive any Tdap vaccine. The association between Td3ap maternal immunisation and pertussis in infants was assessed for US data using a frequentist method with conditional logistic regression. A robustified analysis was conducted using Bayesian dynamic borrowing of non-US data, considering a mixing-weighted prior of 90% for historical non-US VE data, and of 10% for a vague prior. VE was estimated as (1-odds ratio) × 100%. Sensitivity analyses accounting for the impact of each non-US study, different mixing weights and missing/ambiguous data were performed. RESULTS We included 108 cases and 183 controls. Based on US data, the estimated VE of third-trimester maternal immunisation with Td3ap at preventing pertussis in infants <2 months was 78.0% (95% confidence interval: -38.0; 96.5). VE estimated by Bayesian dynamic borrowing of non-US data (with a 90% weight for historical data) was 83.4% (95% credible interval: 55.7; 92.5); sensitivity analyses produced similar VE estimates. CONCLUSIONS Effectiveness of third-trimester pregnancy immunisation with Td3ap at preventing infant pertussis in the US is very likely to be ≥ 50% and is most likely ∼ 80%. Bayesian dynamic borrowing of non-US VE data allowed overcoming the limited power (due to small sample size) of a brand-specific sub-analysis by considering additional evidence.
Collapse
Affiliation(s)
| | | | | | - Nadia Meyer
- GSK, Avenue Fleming 20, 1300 Wavre, Belgium.
| | | |
Collapse
|
12
|
Wang Z, Ma W, Fu X, Qi Y, Zhao Y, Zhang S. Development and applications of mRNA treatment based on lipid nanoparticles. Biotechnol Adv 2023; 65:108130. [PMID: 36933868 DOI: 10.1016/j.biotechadv.2023.108130] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/06/2022] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Nucleic acid-based therapies such as messenger RNA have the potential to revolutionize modern medicine and enhance the performance of existing pharmaceuticals. The key challenges of mRNA-based therapies are delivering the mRNA safely and effectively to the target tissues and cells and controlling its release from the delivery vehicle. Lipid nanoparticles (LNPs) have been widely studied as drug carriers and are considered to be state-of-the-art technology for nucleic acid delivery. In this review, we begin by presenting the advantages and mechanisms of action of mRNA therapeutics. Then we discuss the design of LNP platforms based on ionizable lipids and the applications of mRNA-LNP vaccines for prevention of infectious diseases and for treatment of cancer and various genetic diseases. Finally, we describe the challenges and future prospects of mRNA-LNP therapeutics.
Collapse
Affiliation(s)
- Zhe Wang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Wanting Ma
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Xingxing Fu
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Yanfei Qi
- Centenary Institute, The University of Sydney, Sydney, NSW 2050, Australia
| | - Yinan Zhao
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| |
Collapse
|
13
|
deCamp AC, Corcoran MM, Fulp WJ, Willis JR, Cottrell CA, Bader DLV, Kalyuzhniy O, Leggat DJ, Cohen KW, Hyrien O, Menis S, Finak G, Ballweber-Fleming L, Srikanth A, Plyler JR, Rahaman F, Lombardo A, Philiponis V, Whaley RE, Seese A, Brand J, Ruppel AM, Hoyland W, Mahoney CR, Cagigi A, Taylor A, Brown DM, Ambrozak DR, Sincomb T, Mullen TM, Maenza J, Kolokythas O, Khati N, Bethony J, Roederer M, Diemert D, Koup RA, Laufer DS, McElrath JM, McDermott AB, Hedestam GBK, Schief WR. Human immunoglobulin gene allelic variation impacts germline-targeting vaccine priming. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.10.23287126. [PMID: 36993183 PMCID: PMC10055468 DOI: 10.1101/2023.03.10.23287126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Vaccine priming immunogens that activate germline precursors for broadly neutralizing antibodies (bnAbs) have promise for development of precision vaccines against major human pathogens. In a clinical trial of the eOD-GT8 60mer germline-targeting immunogen, higher frequencies of vaccine-induced VRC01-class bnAb-precursor B cells were observed in the high dose compared to the low dose group. Through immunoglobulin heavy chain variable (IGHV) genotyping, statistical modeling, quantification of IGHV1-2 allele usage and B cell frequencies in the naive repertoire for each trial participant, and antibody affinity analyses, we found that the difference between dose groups in VRC01-class response frequency was best explained by IGHV1-2 genotype rather than dose and was most likely due to differences in IGHV1-2 B cell frequencies for different genotypes. The results demonstrate the need to define population-level immunoglobulin allelic variations when designing germline-targeting immunogens and evaluating them in clinical trials. One-Sentence Summary Human genetic variation can modulate the strength of vaccine-induced broadly neutralizing antibody precursor B cell responses.
Collapse
|
14
|
Kick AR, Grete AF, Crisci E, Almond GW, Käser T. Testable Candidate Immune Correlates of Protection for Porcine Reproductive and Respiratory Syndrome Virus Vaccination. Vaccines (Basel) 2023; 11:vaccines11030594. [PMID: 36992179 DOI: 10.3390/vaccines11030594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/26/2023] [Accepted: 02/26/2023] [Indexed: 03/08/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is an on-going problem for the worldwide pig industry. Commercial and experimental vaccinations often demonstrate reduced pathology and improved growth performance; however, specific immune correlates of protection (CoP) for PRRSV vaccination have not been quantified or even definitively postulated: proposing CoP for evaluation during vaccination and challenge studies will benefit our collective efforts towards achieving protective immunity. Applying the breadth of work on human diseases and CoP to PRRSV research, we advocate four hypotheses for peer review and evaluation as appropriate testable CoP: (i) effective class-switching to systemic IgG and mucosal IgA neutralizing antibodies is required for protective immunity; (ii) vaccination should induce virus-specific peripheral blood CD4+ T-cell proliferation and IFN-γ production with central memory and effector memory phenotypes; cytotoxic T-lymphocytes (CTL) proliferation and IFN-γ production with a CCR7- phenotype that should migrate to the lung; (iii) nursery, finishing, and adult pigs will have different CoP; (iv) neutralizing antibodies provide protection and are rather strain specific; T cells confer disease prevention/reduction and possess greater heterologous recognition. We believe proposing these four CoP for PRRSV can direct future vaccine design and improve vaccine candidate evaluation.
Collapse
Affiliation(s)
- Andrew R Kick
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Department of Chemistry & Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Alicyn F Grete
- Department of Chemistry & Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Elisa Crisci
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - Glen W Almond
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - Tobias Käser
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| |
Collapse
|
15
|
Frost H, Excler JL, Sriskandan S, Fulurija A. Correlates of immunity to Group A Streptococcus: a pathway to vaccine development. NPJ Vaccines 2023; 8:1. [PMID: 36650164 PMCID: PMC9844947 DOI: 10.1038/s41541-022-00593-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/06/2022] [Indexed: 01/19/2023] Open
Abstract
Understanding immunity in humans to Group A Streptococcus (Strep A) is critical for the development of successful vaccines to prevent the morbidity and mortality attributed to Strep A infections. Despite decades of effort, no licensed vaccine against Strep A exists and immune correlates of protection are lacking; a major impediment to vaccine development. In the absence of a vaccine, we can take cues from the development of natural immunity to Strep A in humans to identify immune correlates of protection. The age stratification of incidence of acute Strep A infections, peaking in young children and waning in early adulthood, coincides with the development of specific immune responses. Therefore, understanding the immune mechanisms involved in natural protection from acute Strep A infection is critical to identifying immune correlates to inform vaccine development. This perspective summarises the findings from natural infection studies, existing assays of immunity to Strep A, and highlights the gaps in knowledge to guide the development of Strep A vaccines and associated correlates of protection.
Collapse
Affiliation(s)
- Hannah Frost
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | | | - Shiranee Sriskandan
- Department of Infectious Disease, Imperial College London, London, UK.
- MRC Centre for Molecular Bacteriology & Infection, Imperial College London, London, UK.
| | - Alma Fulurija
- Telethon Kid's Institute, Perth, WA, Australia.
- The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
16
|
Escudero-Pérez B, Lawrence P, Castillo-Olivares J. Immune correlates of protection for SARS-CoV-2, Ebola and Nipah virus infection. Front Immunol 2023; 14:1156758. [PMID: 37153606 PMCID: PMC10158532 DOI: 10.3389/fimmu.2023.1156758] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/20/2023] [Indexed: 05/09/2023] Open
Abstract
Correlates of protection (CoP) are biological parameters that predict a certain level of protection against an infectious disease. Well-established correlates of protection facilitate the development and licensing of vaccines by assessing protective efficacy without the need to expose clinical trial participants to the infectious agent against which the vaccine aims to protect. Despite the fact that viruses have many features in common, correlates of protection can vary considerably amongst the same virus family and even amongst a same virus depending on the infection phase that is under consideration. Moreover, the complex interplay between the various immune cell populations that interact during infection and the high degree of genetic variation of certain pathogens, renders the identification of immune correlates of protection difficult. Some emerging and re-emerging viruses of high consequence for public health such as SARS-CoV-2, Nipah virus (NiV) and Ebola virus (EBOV) are especially challenging with regards to the identification of CoP since these pathogens have been shown to dysregulate the immune response during infection. Whereas, virus neutralising antibodies and polyfunctional T-cell responses have been shown to correlate with certain levels of protection against SARS-CoV-2, EBOV and NiV, other effector mechanisms of immunity play important roles in shaping the immune response against these pathogens, which in turn might serve as alternative correlates of protection. This review describes the different components of the adaptive and innate immune system that are activated during SARS-CoV-2, EBOV and NiV infections and that may contribute to protection and virus clearance. Overall, we highlight the immune signatures that are associated with protection against these pathogens in humans and could be used as CoP.
Collapse
Affiliation(s)
- Beatriz Escudero-Pérez
- WHO Collaborating Centre for Arbovirus and Haemorrhagic Fever Reference and Research, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Reims, Braunschweig, Germany
- *Correspondence: Beatriz Escudero-Pérez, ; Javier Castillo-Olivares,
| | - Philip Lawrence
- CONFLUENCE: Sciences et Humanités (EA 1598), Université Catholique de Lyon (UCLy), Lyon, France
| | - Javier Castillo-Olivares
- Laboratory of Viral Zoonotics, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Beatriz Escudero-Pérez, ; Javier Castillo-Olivares,
| |
Collapse
|
17
|
Single cell multi-omic reference atlases of non-human primate immune tissues reveals CD102 as a biomarker for long-lived plasma cells. Commun Biol 2022; 5:1399. [PMID: 36543997 PMCID: PMC9770566 DOI: 10.1038/s42003-022-04216-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 11/02/2022] [Indexed: 12/24/2022] Open
Abstract
In response to infection or immunization, antibodies are produced that provide protection against re-exposure with the same pathogen. These antibodies can persist at high titers for decades and are maintained by bone marrow-resident long-lived plasma cells (LLPC). However, the durability of antibody responses to immunization varies amongst vaccines. It is unknown what factors contribute to the differential longevity of serum antibody responses and whether heterogeneity in LLPC contributes to this phenomenon. While LLPC differentiation has been studied extensively in mice, little is known about this population in humans or non-human primates (NHP). Here, we use multi-omic single-cell profiling to identify and characterize the LLPC compartment in NHP. We identify LLPC biomarkers including the marker CD102 and show that CD102 in combination with CD31 identifies LLPC in NHP bone marrow. Additionally, we find that CD102 is expressed by LLPC in mouse and humans. These results further our understanding of the LLPC compartment in NHP, identify biomarkers of LLPC, and provide tissue-specific single cell references for future studies.
Collapse
|
18
|
Britto C, Alter G. The next frontier in vaccine design: blending immune correlates of protection into rational vaccine design. Curr Opin Immunol 2022; 78:102234. [PMID: 35973352 PMCID: PMC9612370 DOI: 10.1016/j.coi.2022.102234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/27/2022] [Accepted: 07/13/2022] [Indexed: 02/02/2023]
Abstract
Despite the extraordinary speed and success in SARS-Cov-2 vaccine development, the emergence of variants of concern perplexed the vaccine development community. Neutralizing antibodies waned antibodies waned and were evaded by viral variants, despite the preservation of protection against severe disease and death across vaccinated populations. Similar to other vaccine design efforts, the lack of mechanistic correlates of immunity against Coronavirus Disease 2019, raised questions related to the need for vaccine redesign and boosting. Hence, our limited understanding of mechanistic correlates of immunity - across pathogens - remains a major obstacle in vaccine development. The identification and incorporation of mechanistic correlates of immunity are key to the accelerated design of highly impactful globally relevant vaccines. Systems-biology tools can be applied strategically to define a complete understanding of mechanistic correlates of immunity. Embedding immunological dissection and target immune profile identification, beyond canonical antibody binding and neutralization, may accelerate the design and success of durable protective vaccines.
Collapse
Affiliation(s)
- Carl Britto
- Department of Pediatrics, Boston Children's Hospital, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
19
|
Surface Modification of Biodegradable Microparticles with the Novel Host-Derived Immunostimulant CPDI-02 Significantly Increases Short-Term and Long-Term Mucosal and Systemic Antibodies against Encapsulated Protein Antigen in Young Naïve Mice after Respiratory Immunization. Pharmaceutics 2022; 14:pharmaceutics14091843. [PMID: 36145590 PMCID: PMC9502690 DOI: 10.3390/pharmaceutics14091843] [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: 08/05/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
Generating long-lived mucosal and systemic antibodies through respiratory immunization with protective antigens encapsulated in nanoscale biodegradable particles could potentially decrease or eliminate the incidence of many infectious diseases, but requires the incorporation of a suitable mucosal immunostimulant. We previously found that respiratory immunization with a model protein antigen (LPS-free OVA) encapsulated in PLGA 50:50 nanoparticles (~380 nm diameter) surface-modified with complement peptide-derived immunostimulant 02 (CPDI-02; formerly EP67) through 2 kDa PEG linkers increases mucosal and systemic OVA-specific memory T-cells with long-lived surface phenotypes in young, naïve female C57BL/6 mice. Here, we determined if respiratory immunization with LPS-free OVA encapsulated in similar PLGA 50:50 microparticles (~1 μm diameter) surface-modified with CPDI-02 (CPDI-02-MP) increases long-term OVA-specific mucosal and systemic antibodies. We found that, compared to MP surface-modified with inactive, scrambled scCPDI-02 (scCPDI-02-MP), intranasal administration of CPDI-02-MP in 50 μL sterile PBS greatly increased titers of short-term (14 days post-immunization) and long-term (90 days post-immunization) antibodies against encapsulated LPS-free OVA in nasal lavage fluids, bronchoalveolar lavage fluids, and sera of young, naïve female C57BL/6 mice with minimal lung inflammation. Thus, surface modification of ~1 μm biodegradable microparticles with CPDI-02 is likely to increase long-term mucosal and systemic antibodies against encapsulated protein antigen after respiratory and possibly other routes of mucosal immunization.
Collapse
|
20
|
Immunomodulation and exposure to per- and polyfluoroalkyl substances: an overview of the current evidence from animal and human studies. Arch Toxicol 2022; 96:2261-2285. [PMID: 35695909 DOI: 10.1007/s00204-022-03303-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) have been widely used and represent a class of environmental persistent chemicals. An association of a reduction of vaccination efficacy with PFAS serum levels in humans was used by the European Food Safety Authority as a key effect for PFAS risk assessment. The data support for using this association is reviewed by a critical analysis of the respective human epidemiology and the available animal studies on the immunomodulation of PFAS. Based on an analysis of the available human epidemiology, the overall level of evidence regarding associations between PFAS serum levels and reduced antibody response remains weak. Absence of an association between an increase in clinical infections and PFAS serum levels and the limited understanding of the importance of antibody levels as an isolated data point further support this conclusion. Animal toxicity studies with PFAS focusing on immunomodulation also provide only limited support for immunomodulation as an important endpoint in PFAS toxicity. While immunomodulation is observed after PFAS administration, generally at blood concentrations several orders of magnitude above those seen in environmentally exposed humans, the relevance of these observation is hampered by the high doses required to influence immune endpoints, the limited number of endpoints assessed, and inconsistent results. The limitations of the current database on associations of human PFAS exposures outlined here indicate that more evidence is required to select immunomodulation as a critical endpoint for human PFAS risk assessment.
Collapse
|
21
|
Lipsit S, Facciuolo A, Scruten E, Griebel P, Napper S. Plasma Cytokines and Birth Weight as Biomarkers of Vaccine-Induced Humoral Responses in Piglets. Front Vet Sci 2022; 9:922992. [PMID: 35903142 PMCID: PMC9325413 DOI: 10.3389/fvets.2022.922992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022] Open
Abstract
Failure to mount an effective immune response to vaccination leaves individuals at risk for infection and can compromise herd immunity. Vaccine unresponsiveness can range from poor responses “low responders” to a failure to seroconvert “non-responders.” Biomarkers of vaccine unresponsiveness, particularly those measured at the time of vaccination, could facilitate more strategic vaccination programs. We previously reported that pro-inflammatory cytokine signaling within peripheral blood mononuclear cells, elevated plasma interferon-gamma (IFNγ), and low birth weight correlated with vaccine-induced serum IgG titers in piglets that were below the threshold of detectable seroconversion (vaccine non-responders). These observations suggested that plasma IFNγ concentration and birth weight might serve as pre-vaccination biomarkers of vaccine unresponsiveness. To test this hypothesis, piglets (n = 67) from a different production facility were vaccinated with the same commercial Mycoplasma hyopneumoniae bacterin (RespiSure-One) to determine if there was a consistent and significant association between vaccine-induced serum IgG titers and either plasma cytokine concentrations or birth weight. All piglets seroconverted following vaccination with significantly less variability in vaccine-induced serum IgG titers than observed in the previous vaccine trial. Piglets exhibited highly variable birth weights and plasma cytokine concentrations prior to vaccination, but there were no significant associations (p > 0.05) between these variables and vaccine-induced serum IgG titers. There were significant (p < 0.001) differences in plasma IFNγ concentrations among individual litters (n = 6), and plasma IFNγ concentrations decreased in all pigs from birth to 63-days of age. One of the six litters (n = 11 piglets) exhibited significantly elevated plasma IFNγ concentrations during the first 3 weeks of life (p < 0.001) and at the time of vaccination (p < 0.01). This litter, however, had similar vaccine-induced serum IgG titers when compared to the other piglets in this study. Collectively the two studies indicate that while plasma cytokines and birth weight can be associated with vaccine non-responsiveness, their temporal and individual variation, as well as the complexity of the vaccine responsiveness phenotype, make them inconsistent biomarkers for predicting the less extreme phenotype of vaccine low responders.
Collapse
Affiliation(s)
- Sean Lipsit
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Antonio Facciuolo
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada
| | - Erin Scruten
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada
| | - Philip Griebel
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada
- School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada
| | - Scott Napper
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
- *Correspondence: Scott Napper
| |
Collapse
|
22
|
Alexandre M, Marlin R, Prague M, Coleon S, Kahlaoui N, Cardinaud S, Naninck T, Delache B, Surenaud M, Galhaut M, Dereuddre-Bosquet N, Cavarelli M, Maisonnasse P, Centlivre M, Lacabaratz C, Wiedemann A, Zurawski S, Zurawski G, Schwartz O, Sanders RW, Le Grand R, Levy Y, Thiébaut R. Modelling the response to vaccine in non-human primates to define SARS-CoV-2 mechanistic correlates of protection. eLife 2022; 11:75427. [PMID: 35801637 PMCID: PMC9282856 DOI: 10.7554/elife.75427] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 06/22/2022] [Indexed: 11/29/2022] Open
Abstract
The definition of correlates of protection is critical for the development of next-generation SARS-CoV-2 vaccine platforms. Here, we propose a model-based approach for identifying mechanistic correlates of protection based on mathematical modelling of viral dynamics and data mining of immunological markers. The application to three different studies in non-human primates evaluating SARS-CoV-2 vaccines based on CD40-targeting, two-component spike nanoparticle and mRNA 1273 identifies and quantifies two main mechanisms that are a decrease of rate of cell infection and an increase in clearance of infected cells. Inhibition of RBD binding to ACE2 appears to be a robust mechanistic correlate of protection across the three vaccine platforms although not capturing the whole biological vaccine effect. The model shows that RBD/ACE2 binding inhibition represents a strong mechanism of protection which required significant reduction in blocking potency to effectively compromise the control of viral replication.
Collapse
Affiliation(s)
- Marie Alexandre
- Department of Public Health, Inserm Bordeaux Population Health Research Centre, University of Bordeaux, Inria SISTM, UMR 1219, Bordeaux, France
| | - Romain Marlin
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Mélanie Prague
- Department of Public Health, Inserm Bordeaux Population Health Research Centre, University of Bordeaux, Inria SISTM, UMR 1219, Bordeaux, France
| | - Severin Coleon
- Vaccine Research Institute, Inserm U955, Créteil, France
| | - Nidhal Kahlaoui
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | | | - Thibaut Naninck
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Benoit Delache
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | | | - Mathilde Galhaut
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Nathalie Dereuddre-Bosquet
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Mariangela Cavarelli
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Pauline Maisonnasse
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | | | | | | | - Sandra Zurawski
- Baylor Scott and White Research Institute, Dallas, United States
| | - Gerard Zurawski
- Baylor Scott and White Research Institute, Dallas, United States
| | | | - Rogier W Sanders
- Department of Medical Microbiology, University of Amsterdam, Amsterdam, Netherlands
| | - Roger Le Grand
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Yves Levy
- Vaccine Research Institute, Inserm U955, Créteil, France
| | - Rodolphe Thiébaut
- Department of Public Health, Inserm Bordeaux Population Health Research Centre, University of Bordeaux, Inria SISTM, UMR 1219, Bordeaux, France
| |
Collapse
|
23
|
Ekezie W, Awwad S, Krauchenberg A, Karara N, Dembiński Ł, Grossman Z, del Torso S, Dornbusch HJ, Neves A, Copley S, Mazur A, Hadjipanayis A, Grechukha Y, Nohynek H, Damnjanović K, Lazić M, Papaevangelou V, Lapii F, Stein-Zamir C, Rath B. Access to Vaccination among Disadvantaged, Isolated and Difficult-to-Reach Communities in the WHO European Region: A Systematic Review. Vaccines (Basel) 2022; 10:1038. [PMID: 35891201 PMCID: PMC9324407 DOI: 10.3390/vaccines10071038] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/30/2022] [Accepted: 06/14/2022] [Indexed: 01/27/2023] Open
Abstract
Vaccination has a significant impact on morbidity and mortality. High vaccination coverage rates are required to achieve herd protection against vaccine-preventable diseases. However, limited vaccine access and hesitancy among specific communities represent significant obstacles to this goal. This review provides an overview of critical factors associated with vaccination among disadvantaged groups in World Health Organisation European countries. Initial searches yielded 18,109 publications from four databases, and 104 studies from 19 out of 53 countries reporting 22 vaccine-preventable diseases were included. Nine groups representing the populations of interest were identified, and most of the studies focused on asylum seekers, refugees, migrants and deprived communities. Recall of previous vaccinations received was poor, and serology was conducted in some cases to confirm protection for those who received prior vaccinations. Vaccination coverage was lower among study populations compared to the general population or national average. Factors that influenced uptake, which presented differently at different population levels, included health service accessibility, language and vaccine literacy, including risk perception, disease severity and vaccination benefits. Strategies that could be implemented in vaccination policy and programs were also identified. Overall, interventions specific to target communities are vital to improving uptake. More innovative strategies need to be deployed to improve vaccination coverage among disadvantaged groups.
Collapse
Affiliation(s)
- Winifred Ekezie
- Vienna Vaccine Safety Initiative e.V., 10437 Berlin, Germany; (W.E.); (S.A.)
- Department of Epidemiology and Public Health, University of Nottingham, Nottingham NG5 1PB, UK
- College of Life Sciences, University of Leicester, Leicester LE5 4PW, UK
- ImmuHubs Consortium, Coordinating Entity: Vienna Vaccine Safety Initiative e.V., 10437 Berlin, Germany;
| | - Samy Awwad
- Vienna Vaccine Safety Initiative e.V., 10437 Berlin, Germany; (W.E.); (S.A.)
- ImmuHubs Consortium, Coordinating Entity: Vienna Vaccine Safety Initiative e.V., 10437 Berlin, Germany;
- Stanford University, Palo Alto, CA 94305, USA
| | - Arja Krauchenberg
- ImmuHubs Consortium, Coordinating Entity: Vienna Vaccine Safety Initiative e.V., 10437 Berlin, Germany;
- European Parents Association, 1000 Brussels, Belgium
| | - Nora Karara
- Young European Academy of Paediatrics, 1000 Brussels, Belgium;
- Evangelical Hospital Queen Elisabeth Herzberge, 10365 Berlin, Germany
| | - Łukasz Dembiński
- European Academy of Paediatrics, 1000 Brussels, Belgium; (Ł.D.); (Z.G.); (S.d.T.); (H.J.D.); (A.N.); (S.C.); (A.M.); (A.H.); (Y.G.); (V.P.); (F.L.)
- Department of Pediatric Gastroenterology and Nutrition, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Zachi Grossman
- European Academy of Paediatrics, 1000 Brussels, Belgium; (Ł.D.); (Z.G.); (S.d.T.); (H.J.D.); (A.N.); (S.C.); (A.M.); (A.H.); (Y.G.); (V.P.); (F.L.)
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - Stefano del Torso
- European Academy of Paediatrics, 1000 Brussels, Belgium; (Ł.D.); (Z.G.); (S.d.T.); (H.J.D.); (A.N.); (S.C.); (A.M.); (A.H.); (Y.G.); (V.P.); (F.L.)
| | - Hans Juergen Dornbusch
- European Academy of Paediatrics, 1000 Brussels, Belgium; (Ł.D.); (Z.G.); (S.d.T.); (H.J.D.); (A.N.); (S.C.); (A.M.); (A.H.); (Y.G.); (V.P.); (F.L.)
| | - Ana Neves
- European Academy of Paediatrics, 1000 Brussels, Belgium; (Ł.D.); (Z.G.); (S.d.T.); (H.J.D.); (A.N.); (S.C.); (A.M.); (A.H.); (Y.G.); (V.P.); (F.L.)
| | - Sian Copley
- European Academy of Paediatrics, 1000 Brussels, Belgium; (Ł.D.); (Z.G.); (S.d.T.); (H.J.D.); (A.N.); (S.C.); (A.M.); (A.H.); (Y.G.); (V.P.); (F.L.)
| | - Artur Mazur
- European Academy of Paediatrics, 1000 Brussels, Belgium; (Ł.D.); (Z.G.); (S.d.T.); (H.J.D.); (A.N.); (S.C.); (A.M.); (A.H.); (Y.G.); (V.P.); (F.L.)
| | - Adamos Hadjipanayis
- European Academy of Paediatrics, 1000 Brussels, Belgium; (Ł.D.); (Z.G.); (S.d.T.); (H.J.D.); (A.N.); (S.C.); (A.M.); (A.H.); (Y.G.); (V.P.); (F.L.)
| | - Yevgenii Grechukha
- European Academy of Paediatrics, 1000 Brussels, Belgium; (Ł.D.); (Z.G.); (S.d.T.); (H.J.D.); (A.N.); (S.C.); (A.M.); (A.H.); (Y.G.); (V.P.); (F.L.)
| | - Hanna Nohynek
- Finnish Institute for Health and Welfare, FI-00271 Helsinki, Finland;
| | - Kaja Damnjanović
- Faculty of Philosophy, University of Belgrade, 11000 Beograd, Serbia;
| | - Milica Lazić
- Faculty of Philosophy, University of Novi Sad, 21000 Novi Sad, Serbia;
| | - Vana Papaevangelou
- European Academy of Paediatrics, 1000 Brussels, Belgium; (Ł.D.); (Z.G.); (S.d.T.); (H.J.D.); (A.N.); (S.C.); (A.M.); (A.H.); (Y.G.); (V.P.); (F.L.)
| | - Fedir Lapii
- European Academy of Paediatrics, 1000 Brussels, Belgium; (Ł.D.); (Z.G.); (S.d.T.); (H.J.D.); (A.N.); (S.C.); (A.M.); (A.H.); (Y.G.); (V.P.); (F.L.)
| | | | - Barbara Rath
- Vienna Vaccine Safety Initiative e.V., 10437 Berlin, Germany; (W.E.); (S.A.)
- Department of Epidemiology and Public Health, University of Nottingham, Nottingham NG5 1PB, UK
- ImmuHubs Consortium, Coordinating Entity: Vienna Vaccine Safety Initiative e.V., 10437 Berlin, Germany;
| | | |
Collapse
|
24
|
Schenk J, Abrams S, Litzroth A, Cornelissen L, Grammens T, Theeten H, Hens N. Identifying immunity gaps for measles using Belgian serial serology data. Vaccine 2022; 40:3676-3683. [PMID: 35589453 PMCID: PMC9108896 DOI: 10.1016/j.vaccine.2022.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/21/2022] [Accepted: 05/04/2022] [Indexed: 11/29/2022]
Abstract
Vaccine-preventable diseases, such as measles, have been re-emerging in countries with moderate to high vaccine uptake. It is increasingly important to identify and close immunity gaps and increase coverage of routine childhood vaccinations, including two doses of the measles-mumps-rubella vaccine (MMR). Here, we present a simple cohort model relying on a Bayesian approach to evaluate the evolution of measles seroprevalence in Belgium using the three most recent cross-sectional serological survey data collections (2002, 2006 and 2013) and information regarding vaccine properties. We find measles seroprevalence profiles to be similar for the different regions in Belgium. These profiles exhibit a drop in seroprevalence in birth cohorts that were offered vaccination at suboptimal coverages in the first years after routine vaccination has been started up. This immunity gap is observed across all cross-sectional survey years, although it is more pronounced in survey year 2013. At present, the COVID-19 pandemic could negatively impact the immunization coverage worldwide, thereby increasing the need for additional immunization programs in groups of children that are impacted by this. Therefore, it is now even more important to identify existing immunity gaps and to sustain and reach vaccine-derived measles immunity goals.
Collapse
Affiliation(s)
- Julie Schenk
- Data Science Institute, Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-BioStat), UHasselt, Diepenbeek, Belgium.
| | - Steven Abrams
- Data Science Institute, Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-BioStat), UHasselt, Diepenbeek, Belgium,Global Health Institute (GHI), Family Medicine and Population Health (FAMPOP), University of Antwerp, Wilrijk, Belgium
| | - Amber Litzroth
- Service of Epidemiology of Infectious Diseases, Scientific Directorate of Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - Laura Cornelissen
- Service of Epidemiology of Infectious Diseases, Scientific Directorate of Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - Tine Grammens
- Service of Epidemiology of Infectious Diseases, Scientific Directorate of Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - Heidi Theeten
- Centre for Evaluation of Vaccination (CEV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium,Public Health and Surveillance Department, Zorg en Gezondheid Vlaanderen, Belgium
| | - Niel Hens
- Data Science Institute, Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-BioStat), UHasselt, Diepenbeek, Belgium,Centre for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
25
|
Sluder AE, Raju Paul S, Moise L, Dold C, Richard G, Silva-Reyes L, Baeten LA, Scholzen A, Reeves PM, Pollard AJ, Garritsen A, Bowen RA, De Groot AS, Rollier C, Poznansky MC. Evaluation of a Human T Cell-Targeted Multi-Epitope Vaccine for Q Fever in Animal Models of Coxiella burnetii Immunity. Front Immunol 2022; 13:901372. [PMID: 35651616 PMCID: PMC9149306 DOI: 10.3389/fimmu.2022.901372] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022] Open
Abstract
T cell-mediated immunity plays a central role in the control and clearance of intracellular Coxiella burnetii infection, which can cause Q fever. Therefore, we aimed to develop a novel T cell-targeted vaccine that induces pathogen-specific cell-mediated immunity to protect against Q fever in humans while avoiding the reactogenicity of the current inactivated whole cell vaccine. Human HLA class II T cell epitopes from C. burnetii were previously identified and selected by immunoinformatic predictions of HLA binding, conservation in multiple C. burnetii isolates, and low potential for cross-reactivity with the human proteome or microbiome. Epitopes were selected for vaccine inclusion based on long-lived human T cell recall responses to corresponding peptides in individuals that had been naturally exposed to the bacterium during a 2007-2010 Q fever outbreak in the Netherlands. Multiple viral vector-based candidate vaccines were generated that express concatemers of selected epitope sequences arranged to minimize potential junctional neo-epitopes. The vaccine candidates caused no antigen-specific reactogenicity in a sensitized guinea pig model. A subset of the vaccine epitope peptides elicited antigenic recall responses in splenocytes from C57BL/6 mice previously infected with C. burnetii. However, immunogenicity of the vaccine candidates in C57BL/6 mice was dominated by a single epitope and this was insufficient to confer protection against an infection challenge, highlighting the limitations of assessing human-targeted vaccine candidates in murine models. The viral vector-based vaccine candidates induced antigen-specific T cell responses to a broader array of epitopes in cynomolgus macaques, establishing a foundation for future vaccine efficacy studies in this large animal model of C. burnetii infection.
Collapse
Affiliation(s)
- Ann E Sluder
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
| | - Susan Raju Paul
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
| | | | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, The National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | | | - Laura Silva-Reyes
- Oxford Vaccine Group, Department of Paediatrics, The National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Laurie A Baeten
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | | | - Patrick M Reeves
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, The National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | | | - Richard A Bowen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | | | - Christine Rollier
- Oxford Vaccine Group, Department of Paediatrics, The National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
26
|
Affiliation(s)
- Joshua T Schiffer
- Vaccine and Infectious Diseases Division, University of Washington, Seattle, WA, USA.
- Clinical and Research Division Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA.
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA.
| |
Collapse
|
27
|
Silva RP, DiVenere AM, Amengor D, Maynard JA. Antibodies binding diverse pertactin epitopes protect mice from B. pertussis infection. J Biol Chem 2022; 298:101715. [PMID: 35151691 PMCID: PMC8931430 DOI: 10.1016/j.jbc.2022.101715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 11/27/2022] Open
Abstract
Infection by the bacterium Bordetella pertussis continues to cause considerable morbidity and mortality worldwide. Many current acellular pertussis vaccines include the antigen pertactin, which has presumptive adhesive and immunomodulatory activities, but is rapidly lost from clinical isolates after the introduction of these vaccines. To better understand the contributions of pertactin antibodies to protection and pertactin's role in pathogenesis, we isolated and characterized recombinant antibodies binding four distinct epitopes on pertactin. We demonstrate that four of these antibodies bind epitopes that are conserved across all three classical Bordetella strains, and competition assays further showed that antibodies binding these epitopes are also elicited by B. pertussis infection of baboons. Surprisingly, we found that representative antibodies binding each epitope protected mice against experimental B. pertussis infection. A cocktail of antibodies from each epitope group protected mice against a subsequent lethal dose of B. pertussis and greatly reduced lung colonization levels after sublethal challenge. Each antibody reduced B. pertussis lung colonization levels up to 100-fold when administered individually, which was significantly reduced when antibody effector functions were impaired, with no antibody mediating antibody-dependent complement-induced lysis. These data suggest that antibodies binding multiple pertactin epitopes protect primarily by the same bactericidal mechanism, which overshadows contributions from blockade of other pertactin functions. These antibodies expand the available tools to further dissect pertactin's role in infection and understand the impact of antipertactin antibodies on bacterial fitness.
Collapse
|
28
|
Clinical Development of mRNA Vaccines: Challenges and Opportunities. Curr Top Microbiol Immunol 2022; 440:167-186. [PMID: 35906319 DOI: 10.1007/82_2022_259] [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: 01/06/2023]
Abstract
The emergence of safe and effective mRNA platform-based COVID-19 vaccines from the recent pandemic has changed the face of vaccine development. Compared with conventional technologies used historically, mRNA-based vaccines offer a rapid flexible and robust approach to preventing disease caused by transient viral strains such as SAR2-CoV-2 variants of concern and seasonal influenza. Adaptations in the formulation of the mRNA delivery systems such as with lipid nanoparticle delivery (LNP) used in mRNA-1273 and BNT16b2b have enabled this technology to flourish under the urgent collective response and collaborative regulatory understanding derived from COVID-19 vaccine development. The application of mRNA-based therapeutics in other areas holds potential promise including combination vaccines that might deliver protections against multiple infectious diseases. Future studies and further advances in mRNA-based technologies will provide insight into the clinical efficacy and real-world effectiveness of vaccines as well as provisions with respect to the impact of reactogenicity profiles. Overall, the success of mRNA-based COVID-19 vaccines has helped unlock a platform likely to result in many more candidate vaccines entering clinical evaluation to address the unmet medical needs of other diseases including viral respiratory diseases, herpesviruses, and historically challenging vaccine targets such as HIV.
Collapse
|
29
|
Chen Z, Gao X, Yu D. Longevity of vaccine protection: Immunological mechanism, assessment methods, and improving strategy. VIEW 2022. [DOI: 10.1002/viw.20200103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Zhian Chen
- The University of Queensland Diamantina Institute, Faculty of Medicine The University of Queensland Brisbane Queensland Australia
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research Australian National University Canberra Australia
| | - Xin Gao
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research Australian National University Canberra Australia
| | - Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine The University of Queensland Brisbane Queensland Australia
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research Australian National University Canberra Australia
| |
Collapse
|
30
|
Lim WW, Cowling BJ. Mechanistic Correlates of Protection for SARS-CoV-2 Vaccines. Epidemiology 2022; 33:e1. [PMID: 34799478 PMCID: PMC8614193 DOI: 10.1097/ede.0000000000001419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
- Wey Wen Lim
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Benjamin J Cowling
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China, Laboratory of Data Discovery for Health, Hong Kong Science and Technology Park, Hong Kong, China,
| |
Collapse
|
31
|
McLean HQ, Levine MZ, King JP, Flannery B, Belongia EA. Serologic response to sequential vaccination with enhanced influenza vaccines: Open label randomized trial among adults aged 65-74 years. Vaccine 2021; 39:7146-7152. [PMID: 34774360 DOI: 10.1016/j.vaccine.2021.10.072] [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: 04/13/2021] [Revised: 10/15/2021] [Accepted: 10/27/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND The effects of sequential vaccination with enhanced influenza vaccines are poorly understood. We conducted an exploratory open-label study to assess serologic response to sequential vaccination in older adults. METHODS 160 adults aged 65 through 74 years were randomized (1:1:1) to receive trivalent inactivated standard dose (SD), high-dose (HD), or MF59-adjuvanted (AD) vaccine in 2016/17. In 2017/18, HD and AD recipients received the same vaccine; SD recipients were re-randomized to HD or AD. Hemagglutination inhibition assays were performed using turkey erythrocytes against A/California/7/2009(H1N1)-like and B/Brisbane/60/2008(B/Victoria)-like in both seasons, and A/Michigan/45/2015(H1N1)-like in season 2. Microneutralization assays were performed against cell-propagated A/Hong Kong/4801/2014(H3N2)-like using MDCK-SIAT1 cells. Postvaccination geometric mean titer (GMT), percent with titer ≥ 40, and mean fold rise (MFR, ratio of postvaccination versus prevaccination titer) in season 2 were compared across groups, and ratio of MFR in season 2 versus season 1 was assessed for each strain. RESULTS Analysis included 152 participants (55 HD → HD, 58 AD → AD, 19 SD → HD, and 20 SD → AD). Season 2 postvaccination GMTs and percent with titer ≥ 40 did not differ between HD → HD and AD → AD recipients for vaccine strains examined. However, a higher percent of HD → HD and AD → AD recipients had postvaccination titer ≥ 40 than SD → AD recipients for A/H1N1 (86%-89% versus 60%) and SD → AD and SD → HD recipients for A/H3N2 (83%-87% versus 40%-53%). GMTs were higher in AD → AD versus SD → AD recipients for A/H1N1 (p = .01) and A/H3N2 (p = .002). MFRs in season 2 were low in all groups for A/H3N2 (1.5-2.2) and B/Victoria (1.7-2.3). MFR was lower in season 2 versus 1 for HD → HD and AD → AD recipients for all vaccine strains (1.6-3.7 versus 2.6-6.2). CONCLUSIONS Sequential vaccination with enhanced vaccines did not reduce immunogenicity in adults aged 65 through 74 years. Serologic response to cell-propagated A/H3N2 was suboptimal for all vaccines. ClinicalTrials.gov identifier. NCT02872311.
Collapse
Affiliation(s)
- Huong Q McLean
- Center for Clinical Epidemiology & Population Health, Marshfield Clinic Research Institute, 1000 N Oak Ave (ML2), Marshfield, WI 54449, United States.
| | - Min Z Levine
- Influenza Division, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, United States.
| | - Jennifer P King
- Center for Clinical Epidemiology & Population Health, Marshfield Clinic Research Institute, 1000 N Oak Ave (ML2), Marshfield, WI 54449, United States.
| | - Brendan Flannery
- Influenza Division, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, United States.
| | - Edward A Belongia
- Center for Clinical Epidemiology & Population Health, Marshfield Clinic Research Institute, 1000 N Oak Ave (ML2), Marshfield, WI 54449, United States.
| |
Collapse
|
32
|
A method to estimate probability of disease and vaccine efficacy from clinical trial immunogenicity data. NPJ Vaccines 2021; 6:133. [PMID: 34737322 PMCID: PMC8568947 DOI: 10.1038/s41541-021-00377-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 08/11/2021] [Indexed: 12/02/2022] Open
Abstract
Vaccine efficacy is often assessed by counting disease cases in a clinical trial. A new quantitative framework proposed here ("PoDBAY," Probability of Disease Bayesian Analysis), estimates vaccine efficacy (and confidence interval) using immune response biomarker data collected shortly after vaccination. Given a biomarker associated with protection, PoDBAY describes the relationship between biomarker and probability of disease as a sigmoid probability of disease ("PoD") curve. The PoDBAY framework is illustrated using clinical trial simulations and with data for influenza, zoster, and dengue virus vaccines. The simulations demonstrate that PoDBAY efficacy estimation (which integrates the PoD and biomarker data), can be accurate and more precise than the standard (case-count) estimation, contributing to more sensitive and specific decisions than threshold-based correlate of protection or case-count-based methods. For all three vaccine examples, the PoD fit indicates a substantial association between the biomarkers and protection, and efficacy estimated by PoDBAY from relatively little immunogenicity data is predictive of the standard estimate of efficacy, demonstrating how PoDBAY can provide early assessments of vaccine efficacy. Methods like PoDBAY can help accelerate and economize vaccine development using an immunological predictor of protection. For example, in the current effort against the COVID-19 pandemic it might provide information to help prioritize (rank) candidates both earlier in a trial and earlier in development.
Collapse
|
33
|
Echaniz-Aviles G, García-Cisneros S, Sánchez-Alemán MA, Olamendi-Portugal M, Romero-Martinez M, Deantonio R, Cervantes-Apolinar MY, Ortega-Barria E, Cortes-Alcala R, Alpuche-Aranda CM. Estimating Bordetella pertussis seroprevalence in adolescents and young adults in Mexico using the 2012 National Health and Nutrition Survey (ENSANUT). Vaccine 2021; 39:5839-5844. [PMID: 34465476 DOI: 10.1016/j.vaccine.2021.08.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/28/2021] [Accepted: 08/16/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Low vaccination rates and under-detection of pertussis infections in adolescents and young adults have an impact on the transmission of pertussis to infants. In this study, the proportion of adolescents and young adults with IgG antibodies against B. pertussis antigens, representing recent infection or vaccination, was estimated in a population-based probabilistic survey in Mexico. METHODS Sera and data from 1,581 subjects, including 1,102 adolescents and 479 young adults (10-19 and 20-25 years old, respectively) randomly selected from Mexico's 2012 National Health and Nutrition Survey, were analyzed. IgG antibodies against pertussis toxin (PT) were measured with the CDC/FDA ELISA. A subset of 234 samples was additionally tested with Bp-IgG PT ELISA kit (EUROIMMUN AG, Lubeck, Germany). Threshold values from corresponding test kits were used to identify recent infection or vaccination. RESULTS Overall anti-PT IgG seroprevalence was 3.9% (95% CI: 2.3-6.3); 3.1% (95% CI: 1.9-5.0) in adolescents, and 4.9% (95% CI: 2.2-11) in young adults. Seroprevalence did not significantly vary by sex, socioeconomic status, region or rural/urban location. Compared to the CDC/FDA ELISA, the EUROIMMUN test showed a 76% sensitivity and 88% specificity. The weighted estimates represent a considerable burden of recent infection in adolescents and young adults; however, most adolescents and adults were seronegative and, therefore, susceptible to pertussis infection. CONCLUSION Since booster vaccination to B. pertussis after toddlerhood is not recommended in the Mexican national policy, anti-PT IgG seropositivity may be reasonably attributed to recent infection. Assessing pertussis seroprevalence requires careful consideration of the diagnostic test threshold interpretation and epidemiological model used.
Collapse
Affiliation(s)
- G Echaniz-Aviles
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - S García-Cisneros
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - M A Sánchez-Alemán
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - M Olamendi-Portugal
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - M Romero-Martinez
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | | | | | | | | | - C M Alpuche-Aranda
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico.
| |
Collapse
|
34
|
A New Electrochemiluminescence-Based Multiplex Assay for the Assessment of Human Antibody Responses to Bordetella pertussis Vaccines. Infect Dis Ther 2021; 10:2539-2561. [PMID: 34476771 PMCID: PMC8412870 DOI: 10.1007/s40121-021-00530-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/13/2021] [Indexed: 01/19/2023] Open
Abstract
Introduction Commercially available enzyme-linked immunosorbent assay (ELISA) kits designed for pertussis diagnostic purposes are frequently used to assess antibody responses to pertussis vaccines in clinical trials, but have limited accuracy and are not calibrated against international standards. We developed a new electrochemiluminescence (ECL)-based multiplexed assay and compared its performance to two commercial Bordetella pertussis ELISA kits and to historical in-house ELISAs. Methods The ECL assay quantifies serum concentrations of antibodies against four B. pertussis antigens: pertussis toxin (PT), filamentous hemagglutinin (FHA), pertactin (PRN), and fimbrial agglutinogen (FIM). The assay was validated for precision, accuracy, dilutability, lower limit of quantification, and specificity. Sera from a clinical trial (CTRI/2016/11/007434) were used to compare the ECL assay to two commercial ELISA kits available from GenWay BioTech and Demeditec Diagnostics for accuracy, linearity, specificity, and concordance to both internal (WWO-2-043) and international (NIBSC 06/140) references. Sera from four clinical trials (NCT02587520, NCT00255047, NCT00347958, NCT01346293) were used to compare the concordance to clinical ELISAs. Informed consent was ensured prior to using any sera. Results Precision, accuracy, dilutability, lower limit of quantification, and specificity were demonstrated for the ECL assay. Concordance between the ECL assay and established clinical ELISAs was met for antibody responses to PT, FIM, and PRN, but not for FHA. The ECL assay demonstrated higher accuracy and linearity than the ELISA kits. While concordance between the ECL and commercial kits was low, the ECL assay better distinguished between pre- and post-vaccination clinical samples. Conclusion The new ECL assay was validated for the quantitative evaluation of anti-PT, anti-FHA, anti-FIM, and anti-PRN IgG antibodies in samples from clinical trials, and demonstrated equivalent or better performance than two commercially available ELISA kits.
Collapse
|
35
|
Seaton KE, Spreng RL, Abraha M, Reichartz M, Rojas M, Feely F, Huntwork RHC, Dutta S, Mudrak SV, Alam SM, Gregory S, Jongert E, Coccia M, Ulloa-Montoya F, Wille-Reece U, Tomaras GD, Dennison SM. Subclass and avidity of circumsporozoite protein specific antibodies associate with protection status against malaria infection. NPJ Vaccines 2021; 6:110. [PMID: 34462438 PMCID: PMC8405700 DOI: 10.1038/s41541-021-00372-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 07/30/2021] [Indexed: 12/19/2022] Open
Abstract
RTS,S/AS01 is an advanced pre-erythrocytic malaria vaccine candidate with demonstrated vaccine efficacy up to 86.7% in controlled human malaria infection (CHMI) studies; however, reproducible immune correlates of protection (CoP) are elusive. To identify candidates of humoral correlates of vaccine mediated protection, we measured antibody magnitude, subclass, and avidity for Plasmodium falciparum (Pf) circumsporozoite protein (CSP) by multiplex assays in two CHMI studies with varying RTS,S/AS01B vaccine dose and timing regimens. Central repeat (NANP6) IgG1 magnitude correlated best with protection status in univariate analyses and was the most predictive for protection in a multivariate model. NANP6 IgG3 magnitude, CSP IgG1 magnitude, and total serum antibody dissociation phase area-under-the-curve for NANP6, CSP, NPNA3, and N-interface binding were also associated with protection status in the regimen adjusted univariate analysis. Identification of multiple immune response features that associate with protection status, such as antibody subclasses, fine specificity and avidity reported here may accelerate development of highly efficacious vaccines against P. falciparum.
Collapse
Grants
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- United States Department of Defense | United States Army | Army Medical Command | Walter Reed Army Institute of Research (WRAIR)
- PATH Malaria Vaccine Initiative
- GlaxoSmithKline (GlaxoSmithKline plc.)
Collapse
Affiliation(s)
- Kelly E Seaton
- Duke Human Vaccine Institute, Durham, NC, USA.
- Duke Center for Human Systems Immunology, Durham, NC, USA.
- Duke University Department of Surgery, Durham, NC, USA.
| | - Rachel L Spreng
- Duke Human Vaccine Institute, Durham, NC, USA.
- Duke Center for Human Systems Immunology, Durham, NC, USA.
| | - Milite Abraha
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | - Matthew Reichartz
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | | | - Frederick Feely
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | - Richard H C Huntwork
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | - Sheetij Dutta
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Sarah V Mudrak
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | - S Munir Alam
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke University Department of Pathology, Durham, NC, USA
| | - Scott Gregory
- PATH's Malaria Vaccine Initiative, Washington, DC, USA
| | | | | | | | - Ulrike Wille-Reece
- PATH's Malaria Vaccine Initiative, Washington, DC, USA
- GSK, Rockville, MD, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Durham, NC, USA.
- Duke Center for Human Systems Immunology, Durham, NC, USA.
- Duke University Department of Surgery, Durham, NC, USA.
- Duke University Department of Immunology, Durham, NC, USA.
- Duke University Department of Molecular Genetics and Microbiology, Durham, NC, USA.
| | - S Moses Dennison
- Duke Human Vaccine Institute, Durham, NC, USA.
- Duke Center for Human Systems Immunology, Durham, NC, USA.
- Duke University Department of Surgery, Durham, NC, USA.
| |
Collapse
|
36
|
Barkoff AM, Knuutila A, Mertsola J, He Q. Evaluation of Anti-PT Antibody Response after Pertussis Vaccination and Infection: The Importance of Both Quantity and Quality. Toxins (Basel) 2021; 13:toxins13080508. [PMID: 34437379 PMCID: PMC8402585 DOI: 10.3390/toxins13080508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/21/2022] Open
Abstract
Pertussis toxin (PT) is considered the main virulence factor causing whooping cough or pertussis. The protein is widely studied and its composition was revealed and sequenced already during the 1980s. The human immune system creates a good response against PT when measured in quantity. However, the serum anti-PT antibodies wane rapidly, and only a small amount of these antibodies are found a few years after vaccination/infection. Therefore, multiple approaches to study the functionality (quality) of these antibodies, e.g., avidity, neutralizing capacity, and epitope specificity, have been investigated. In addition, the long-term B cell memory (Bmem) to PT is crucial for good protection throughout life. In this review, we summarize the findings from functional PT antibody and Bmem studies. These results are discussed in line with the quantity of serum anti-PT antibodies. PT neutralizing antibodies and anti-PT antibodies with proper avidity are crucial for good protection against the disease, and certain epitopes have been identified to have multiple functions in the protection. Although PT-specific Bmem responses are detectable at least five years after vaccination, long-term surveillance is lacking. Variation of the natural boosting of circulating Bordetella pertussis in communities is an important confounding factor in these memory studies.
Collapse
Affiliation(s)
- Alex-Mikael Barkoff
- Research Center for Infection and Immunity, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.-M.B.); (A.K.); (J.M.)
| | - Aapo Knuutila
- Research Center for Infection and Immunity, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.-M.B.); (A.K.); (J.M.)
| | - Jussi Mertsola
- Research Center for Infection and Immunity, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.-M.B.); (A.K.); (J.M.)
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, 20520 Turku, Finland
| | - Qiushui He
- Research Center for Infection and Immunity, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.-M.B.); (A.K.); (J.M.)
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
- Correspondence: ; Tel.: +358-40-472-2255
| |
Collapse
|
37
|
Diks AM, Khatri I, Oosten LE, de Mooij B, Groenland RJ, Teodosio C, Perez-Andres M, Orfao A, Berbers GAM, Zwaginga JJ, van Dongen JJM, Berkowska MA. Highly Sensitive Flow Cytometry Allows Monitoring of Changes in Circulating Immune Cells in Blood After Tdap Booster Vaccination. Front Immunol 2021; 12:666953. [PMID: 34177905 PMCID: PMC8223751 DOI: 10.3389/fimmu.2021.666953] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022] Open
Abstract
Antigen-specific serum immunoglobulin (Ag-specific Ig) levels are broadly used as correlates of protection. However, in several disease and vaccination models these fail to predict immunity. In these models, in-depth knowledge of cellular processes associated with protective versus poor responses may bring added value. We applied high-throughput multicolor flow cytometry to track over-time changes in circulating immune cells in 10 individuals following pertussis booster vaccination (Tdap, Boostrix®, GlaxoSmithKline). Next, we applied correlation network analysis to extensively investigate how changes in individual cell populations correlate with each other and with Ag-specific Ig levels. We further determined the most informative cell subsets and analysis time points for future studies. Expansion and maturation of total IgG1 plasma cells, which peaked at day 7 post-vaccination, was the most prominent cellular change. Although these cells preceded the increase in Ag-specific serum Ig levels, they did not correlate with the increase of Ig levels. In contrast, strong correlation was observed between Ag-specific IgGs and maximum expansion of total IgG1 and IgA1 memory B cells at days 7 to 28. Changes in circulating T cells were limited, implying the need for a more sensitive approach. Early changes in innate immune cells, i.e. expansion of neutrophils, and expansion and maturation of monocytes up to day 5, most likely reflected their responses to local damage and adjuvant. Here we show that simultaneous monitoring of multiple circulating immune subsets in blood by flow cytometry is feasible. B cells seem to be the best candidates for vaccine monitoring.
Collapse
Affiliation(s)
- Annieck M. Diks
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Indu Khatri
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, Netherlands
| | | | - Bas de Mooij
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Rick J. Groenland
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Cristina Teodosio
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Martin Perez-Andres
- Cancer Research Centre (IBMCC, USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Alberto Orfao
- Cancer Research Centre (IBMCC, USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Guy A. M. Berbers
- Center for Infectious Disease Control, National Institute of Public Health and the Environment, Bilthoven, Netherlands
| | - Jaap Jan Zwaginga
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | | |
Collapse
|
38
|
Walti CS, Krantz EM, Maalouf J, Boonyaratanakornkit J, Keane-Candib J, Joncas-Schronce L, Stevens-Ayers T, Dasgupta S, Taylor JJ, Hirayama AV, Bar M, Gardner RA, Cowan AJ, Green DJ, Boeckh MJ, Maloney DG, Turtle CJ, Hill JA. Antibodies against vaccine-preventable infections after CAR-T cell therapy for B cell malignancies. JCI Insight 2021; 6:146743. [PMID: 33914708 PMCID: PMC8262349 DOI: 10.1172/jci.insight.146743] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/28/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Little is known about pathogen-specific humoral immunity after chimeric antigen receptor–modified T (CAR-T) cell therapy for B cell malignancies. METHODS We conducted a prospective cross-sectional study of CD19-targeted or B cell maturation antigen–targeted (BCMA-targeted) CAR-T cell therapy recipients at least 6 months posttreatment and in remission. We measured pathogen-specific IgG against 12 vaccine-preventable infections and the number of viral and bacterial epitopes to which IgG was detected (“epitope hits”) using a serological profiling assay. The primary outcome was the proportion of participants with IgG levels above a threshold correlated with seroprotection for vaccine-preventable infections. RESULTS We enrolled 65 children and adults a median of 20 months after CD19- (n = 54) or BCMA- (n = 11) CAR-T cell therapy. Among 30 adults without IgG replacement therapy (IGRT) in the prior 16 weeks, 27 (90%) had hypogammaglobulinemia. These individuals had seroprotection to a median of 67% (IQR, 59%–73%) of tested infections. Proportions of participants with seroprotection per pathogen were comparable to population-based studies, but most individuals lacked seroprotection to specific pathogens. Compared with CD19-CAR-T cell recipients, BCMA-CAR-T cell recipients were half as likely to have seroprotection (prevalence ratio, 0.47; 95% CI, 0.18–1.25) and had fewer pathogen-specific epitope hits (mean difference, –90 epitope hits; 95% CI, –157 to –22). CONCLUSION Seroprotection for vaccine-preventable infections in adult CD19-CAR-T cell recipients was comparable to the general population. BCMA-CAR-T cell recipients had fewer pathogen-specific antibodies. Deficits in both groups support the need for vaccine and immunoglobulin replacement therapy studies. FUNDING Swiss National Science Foundation (Early Postdoc Mobility grant P2BSP3_188162), NIH/National Cancer Institute (NIH/NCI) (U01CA247548 and P01CA018029), NIH/NCI Cancer Center Support Grants (P30CA0087-48 and P30CA015704-44), American Society for Transplantation and Cellular Therapy, and Juno Therapeutics/BMS. In this prospective study, we investigated antibodies against vaccine-preventable infections and other pathogen-specific antibodies in individuals with remission after CAR-T cell therapy for B lineage malignancies.
Collapse
Affiliation(s)
- Carla S Walti
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Elizabeth M Krantz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Joyce Maalouf
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jim Boonyaratanakornkit
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Jacob Keane-Candib
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Laurel Joncas-Schronce
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Terry Stevens-Ayers
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Sayan Dasgupta
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Justin J Taylor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Alexandre V Hirayama
- Clinical Research Division, and.,Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Merav Bar
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, and.,Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Rebecca A Gardner
- Clinical Research Division, and.,Seattle Children's Hospital, Seattle, Washington, USA
| | - Andrew J Cowan
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, and.,Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Damian J Green
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, and.,Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Michael J Boeckh
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, and.,Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - David G Maloney
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, and.,Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Cameron J Turtle
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, and.,Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, and.,Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
39
|
Koch T, Fathi A, Addo MM. The COVID-19 Vaccine Landscape. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1318:549-573. [PMID: 33973199 DOI: 10.1007/978-3-030-63761-3_31] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The history of vaccine development spans centuries. At first, whole pathogens were used as vaccine agents, either inactivated or attenuated, to reduce virulence in humans. Safety and tolerability were increased by including only specific proteins as antigens and using cell culture methods, while novel vaccine strategies, like nucleic acid- or vector-based vaccines, hold high promise for the future. Vaccines have generally not been employed as the primary tools in outbreak response, but this might change since advances in medical technology in the last decades have made the concept of developing vaccines against novel pathogens a realistic strategy. Wandering the uncharted territory of a novel pathogen, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we can learn from other human Betacoronaviridae that emerged in the last decades, SARS-CoV-1 and MERS-CoV. We can identify the most likely target structures of immunity, establish animal models that emulate human disease and immunity as closely as possible, and learn about complex mechanisms of immune interaction such as cross-reactivity or antibody-dependent enhancement (ADE). However, significant knowledge gaps remain. What are the correlates of protection? How do we best induce immunity in vulnerable populations like the elderly? Will the immunity induced by vaccination (or by natural infection) wane over time? To date, at least 149 vaccine candidates against SARS-CoV-2 are under development. At the time of writing, at least 17 candidates have already progressed past preclinical studies (in vitro models and in vivo animal experiments) into clinical development. This chapter will provide an overview of this rapidly developing field.
Collapse
Affiliation(s)
- Till Koch
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany. .,German Center for Infection Research, Hamburg-Lubeck-Borstel-Riems, Hamburg, Germany.
| | - Anahita Fathi
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research, Hamburg-Lubeck-Borstel-Riems, Hamburg, Germany
| | - Marylyn M Addo
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research, Hamburg-Lubeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
40
|
Herzog C, Van Herck K, Van Damme P. Hepatitis A vaccination and its immunological and epidemiological long-term effects - a review of the evidence. Hum Vaccin Immunother 2021; 17:1496-1519. [PMID: 33325760 PMCID: PMC8078665 DOI: 10.1080/21645515.2020.1819742] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/16/2020] [Accepted: 09/01/2020] [Indexed: 01/11/2023] Open
Abstract
Hepatitis A virus (HAV) infections continue to represent a significant disease burden causing approximately 200 million infections, 30 million symptomatic illnesses and 30,000 deaths each year. Effective and safe hepatitis A vaccines have been available since the early 1990s. Initially developed for individual prophylaxis, HAV vaccines are now increasingly used to control hepatitis A in endemic areas. The human enteral HAV is eradicable in principle, however, HAV eradication is currently not being pursued. Inactivated HAV vaccines are safe and, after two doses, elicit seroprotection in healthy children, adolescents, and young adults for an estimated 30-40 years, if not lifelong, with no need for a later second booster. The long-term effects of the single-dose live-attenuated HAV vaccines are less well documented but available data suggest they are safe and provide long-lasting immunity and protection. A universal mass vaccination strategy (UMV) based on two doses of inactivated vaccine is commonly implemented in endemic countries and eliminates clinical hepatitis A disease in toddlers within a few years. Consequently, older age groups also benefit due to the herd protection effects. Single-dose UMV programs have shown promising outcomes but need to be monitored for many more years in order to document an effective immune memory persistence. In non-endemic countries, prevention efforts need to focus on 'new' risk groups, such as men having sex with men, prisoners, the homeless, and families visiting friends and relatives in endemic countries. This narrative review presents the current evidence regarding the immunological and epidemiological long-term effects of the hepatitis A vaccination and finally discusses emerging issues and areas for research.
Collapse
Affiliation(s)
- Christian Herzog
- Department of Medicine, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Koen Van Herck
- Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Department of Public Health, Ghent University, Ghent, Belgium
| | - Pierre Van Damme
- Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
41
|
Adam L, Rosenbaum P, Bonduelle O, Combadière B. Strategies for Immunomonitoring after Vaccination and during Infection. Vaccines (Basel) 2021; 9:365. [PMID: 33918841 PMCID: PMC8070333 DOI: 10.3390/vaccines9040365] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 01/08/2023] Open
Abstract
Immunomonitoring is the study of an individual's immune responses over the course of vaccination or infection. In the infectious context, exploring the innate and adaptive immune responses will help to investigate their contribution to viral control or toxicity. After vaccination, immunomonitoring of the correlate(s) and surrogate(s) of protection is a major asset for measuring vaccine immune efficacy. Conventional immunomonitoring methods include antibody-based technologies that are easy to use. However, promising sensitive high-throughput technologies allowed the emergence of holistic approaches. This raises the question of data integration methods and tools. These approaches allow us to increase our knowledge on immune mechanisms as well as the identification of key effectors of the immune response. However, the depiction of relevant findings requires a well-rounded consideration beforehand about the hypotheses, conception, organization and objectives of the immunomonitoring. Therefore, well-standardized and comprehensive studies fuel insight to design more efficient, rationale-based vaccines and therapeutics to fight against infectious diseases. Hence, we will illustrate this review with examples of the immunomonitoring approaches used during vaccination and the COVID-19 pandemic.
Collapse
Affiliation(s)
| | | | | | - Behazine Combadière
- Inserm, Centre d’Immunologie et des Maladies Infectieuses, Sorbonne Université, 75013 Paris, France; (L.A.); (P.R.); (O.B.)
| |
Collapse
|
42
|
Vaz-de-Lima LR, Sato APS, Pawloski LC, Fernandes EG, Rajam G, Sato HK, Patel D, Li H, de Castilho EA, Tondella ML, Schiffer J. Effect of maternal Tdap on infant antibody response to a primary vaccination series with whole cell pertussis vaccine in São Paulo, Brazil. Vaccine X 2021; 7:100087. [PMID: 33817624 PMCID: PMC8010450 DOI: 10.1016/j.jvacx.2021.100087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Maternal Tetanus, diphtheria, and acellular pertussis (Tdap) vaccination provides antibody transfer to newborn infants and may affect their antibody response to the primary vaccination series. This study aimed to assess the effect of Tdap vaccination during pregnancy on infant antibody response to the whole cell pertussis (DTwP) primary series. METHODS Plasma from 318 pregnant women (243 Tdap-vaccinated and 75 unvaccinated) and their infants (cord blood) was collected at delivery; infant blood was again collected at 2 and 7 months, before and after their primary DTwP series. Anti-pertussis toxin (PT), pertactin (PRN), filamentous hemagglutinin (FHA), fimbriae 2/3 (FIM) and adenylate cyclase toxin (ACT) IgG antibodies were quantified by a microsphere-based multiplex antibody capture assay and anti-PT neutralizing antibodies by the Real Time Cell analysis system. RESULTS Infant geometric mean concentrations (GMCs) of IgG anti-Tdap antigens were significantly higher (p < 0.001) among the Tdap-vaccinated (PT: 57.22 IU/mL; PRN: 464.86 IU/mL; FHA: 424.0 IU/mL), versus the unvaccinated group (4 IU/mL, 15.43 IU/mL, 31.99 IU/mL, respectively) at delivery. Anti-FIM and ACT GMCs were similar between the two groups. At 2 months of age, anti-PT, PRN, and FHA GMCs remained higher (p < 0.001) in the Tdap-vaccinated group (12.64 IU/mL; 108.76 IU/mL; 87.41 IU/mL, respectively) than the unvaccinated group (1.02 IU/mL; 4.46 IU/mL; 6.89 IU/mL). However, at 7 months, after receiving the third DTwP dose, the anti-PT GMC was higher (p = 0.016) in the unvaccinated group (7.91 IU/mL) compared to the vaccinated group (2.27 IU/mL), but without differences for anti-PRN, FHA, FIM and ACT GMCs. CONCLUSION Elevated antibody levels suggest that maternal Tdap vaccination might protect infants until 2 months of age. Reduced anti-PT levels at 7 months indicate potential blunting of immune response in infants. Surveillance would help determine if blunting alters vaccine immunity and impacts pertussis prevention in infants.
Collapse
Affiliation(s)
- Lourdes R.A. Vaz-de-Lima
- Centro de Imunologia, Instituto Adolfo Lutz, Coordenadoria de Controle de Doenças da Secretaria de Estado da Saúde, São Paulo, Brazil
| | - Ana Paula S. Sato
- Departmento de Epidemiologia, Faculdade de Saúde Pública, Universidade de São Paulo – USP, Brazil
| | - Lucia C. Pawloski
- Division of Bacterial Diseases, NCIRD, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Eder G. Fernandes
- Divisão de Imunização, Centro de Vigilância Epidemiológica Prof. Alexandre Vranjac, Coordenadoria de Controle de Doenças da Secretaria de Estado da Saúde SP, Brazil
| | - Gowrisankar Rajam
- Division of Bacterial Diseases, NCIRD, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Helena K. Sato
- Divisão de Imunização, Centro de Vigilância Epidemiológica Prof. Alexandre Vranjac, Coordenadoria de Controle de Doenças da Secretaria de Estado da Saúde SP, Brazil
| | - Divya Patel
- Division of Bacterial Diseases, NCIRD, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Han Li
- Division of Bacterial Diseases, NCIRD, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Maria Lucia Tondella
- Division of Bacterial Diseases, NCIRD, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jarad Schiffer
- Division of Bacterial Diseases, NCIRD, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | |
Collapse
|
43
|
Cody Meissner H. Approaching the End of the Era of Uncontrolled Respiratory Syncytial Virus Disease. J Infect Dis 2021; 223:737-739. [PMID: 33350442 DOI: 10.1093/infdis/jiaa755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/17/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- H Cody Meissner
- Tufts Children's Hospital, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Tissues: the unexplored frontier of antibody mediated immunity. Curr Opin Virol 2021; 47:52-67. [PMID: 33581646 DOI: 10.1016/j.coviro.2021.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/01/2021] [Accepted: 01/05/2021] [Indexed: 12/14/2022]
Abstract
Pathogen-specific immunity evolves in the context of the infected tissue. However, current immune correlates analyses and vaccine efficacy metrics are based on immune functions from peripheral cells. Less is known about tissue-resident mechanisms of immunity. While antibodies represent the primary correlate of immunity following most clinically approved vaccines, how antibodies interact with localized, compartment-specific immune functions to fight infections, remains unclear. Emerging data demonstrate a unique community of immune cells that reside within different tissues. These tissue-specific immunological communities enable antibodies to direct both expected and unexpected local attack strategies to control, disrupt, and eliminate infection in a tissue-specific manner. Defining the full breadth of antibody effector functions, how they selectively contribute to control at the site of infection may provide clues for the design of next-generation vaccines able to direct the control, elimination, and prevention of compartment specific diseases of both infectious and non-infectious etiologies.
Collapse
|
45
|
Abstract
The natural history of cytomegalovirus (CMV) infection is complex. Individuals may experience primary infection, reactivation of latent infection, or reinfection with a new strain despite natural immunity. The ability of this virus to continue to replicate despite substantial immune responses is attributable to the many immune evasion genes encoded within its genome. Given this complex natural history and immunology, the design of clinical trials of CMV vaccines may require components not usually found in trials of vaccines designed to protect against viruses that cause only acute infections. In this article, we focus on specific aspects of clinical trial design that could be adopted to address the complexities of CMV infections. We consider women of childbearing age, toddlers, recipients of solid organ transplantation, and stem cell transplant patients, emphasizing the parallels between women and solid organ transplantation that could allow vaccines to be developed in parallel in both these patient groups. We emphasize the potential for studies of passive immunity to inform the selection of immunogens as candidates for active immunization and vice versa. We also illustrate how application of whole-genomic sequencing could document whether vaccines protect against reactivation or reinfection of CMV or both.
Collapse
Affiliation(s)
- Paul Griffiths
- Institute for Immunity and Transplantation, UCL, London, United Kingdom
| | - Brenna Hughes
- Department of Obstetrics and Gynecology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
46
|
Schenk J, Abrams S, Theeten H, Van Damme P, Beutels P, Hens N. Immunogenicity and persistence of trivalent measles, mumps, and rubella vaccines: a systematic review and meta-analysis. THE LANCET. INFECTIOUS DISEASES 2021; 21:286-295. [PMID: 32888410 PMCID: PMC9665966 DOI: 10.1016/s1473-3099(20)30442-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/23/2020] [Accepted: 04/29/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Despite the universal use of the two-dose trivalent measles-mumps-rubella (MMR) vaccine in the past two decades, outbreaks of these diseases still occur in countries with high vaccine uptake, giving rise to concerns about primary and secondary failure of MMR vaccine components. We aimed to provide seroconversion and waning rate estimates for the measles, mumps, and rubella components of MMR vaccines. METHODS In this systematic review and meta-analysis we searched PubMed (including MEDLINE), Web of Science, and Embase for randomised controlled trials, cohort studies, or longitudinal studies reporting the immunogenicity and persistence of MMR vaccines, published in English from database inception to Dec 31, 2019. Studies were included if they investigated vaccine-induced immunity in healthy individuals who received a trivalent MMR vaccine, including different dosages and timepoints of vaccine administration. Studies featuring coadministration of MMR with other vaccines, maternal immunity to the MMR vaccine, or non-trivalent formulations of the vaccine were excluded. Pooled seroconversion and waning rates were estimated by random-effects meta-analyses. This study is registered with PROSPERO, CRD42019116705. FINDINGS We identified 3615 unique studies, 62 (1·7%) of which were eligible for analysis. Estimated overall seroconversion rates were 96·0% (95% CI 94·5-97·4; I2=91·1%) for measles, 93·3% (91·1-95·2; I2=94·9%) for mumps when excluding the Rubini strain, 91·1% (87·4-94·1; I2=96·6%) for mumps when including the Rubini strain, and 98·3% (97·3-99·2; I2=93·0%) for rubella. Estimated overall annual waning rates were 0·009 (95% CI 0·005-0·016; I2=85·2%) for measles, 0·024 (0·016-0·039; I2=94·7%) for mumps, and 0·012 (0·010-0·014; I2=93·3%) for rubella. INTERPRETATION Our meta-analysis provides estimates of primary and secondary vaccine failure, which are essential to improve the accuracy of mathematical and statistical modelling to understand and predict the occurrence of future measles, mumps, and rubella outbreaks in countries with high vaccine uptake. FUNDING European Research Council.
Collapse
Affiliation(s)
- Julie Schenk
- Data Science Institute, I-BioStat, UHasselt, Diepenbeek, Belgium.
| | - Steven Abrams
- Data Science Institute, I-BioStat, UHasselt, Diepenbeek, Belgium; Global Health Institute, Department of Epidemiology and Social Medicine, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Heidi Theeten
- Centre for the Evaluation of Vaccination, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Pierre Van Damme
- Centre for the Evaluation of Vaccination, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Philippe Beutels
- Centre for Health Economic Research and Modelling Infectious Diseases, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Niel Hens
- Data Science Institute, I-BioStat, UHasselt, Diepenbeek, Belgium; Centre for Health Economic Research and Modelling Infectious Diseases, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
47
|
Adhikari J, Zhao H, Fernandez E, Huang Y, Diamond MS, Fremont DH, Gross ML. Hydrogen-deuterium exchange mass spectrometry identifies spatially distinct antibody epitopes on domain III of the Zika virus envelope protein. JOURNAL OF MASS SPECTROMETRY : JMS 2021; 56:e4685. [PMID: 36101787 PMCID: PMC9467453 DOI: 10.1002/jms.4685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/10/2020] [Indexed: 06/15/2023]
Abstract
Zika Virus (ZIKV) has become a global public health concern because it causes fetal microcephaly and other neurological complications in humans. Currently, there are no approved treatments or vaccines for ZIKV infection. We describe here the detailed epitopes for six monoclonal antibodies (mAbs) that bind to domain III of the envelope protein of ZIKV, some of which have therapeutic potential. We show that by using hydrogen-deuterium exchange mass spectrometry (HDX-MS), we can identify three spatially distinct epitopes for the six mAbs investigated. The HDX-MS approach identified epitopes for three mAbs that agreed well with recently reported X-ray crystallography data. The HDX-MS determined epitopes for the other three anti-ZIKV mAbs for which there were no crystal structures, and the epitopes were confirmed by structure-guided mutagenesis and biolayer interferometry (BLI) competition binding assay. Our results have implications for the design of vaccine and antibody therapeutics against ZIKV and demonstrate the use of HDX-MS as a rapid and valid approach for epitope mapping.
Collapse
Affiliation(s)
- Jagat Adhikari
- Department of Chemistry, Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Haiyan Zhao
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Estefania Fernandez
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Yining Huang
- Department of Chemistry, Washington University in St. Louis, Saint Louis, Missouri, USA
- Present address: Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Michael L Gross
- Department of Chemistry, Washington University in St. Louis, Saint Louis, Missouri, USA
| |
Collapse
|
48
|
Griffiths P, Reeves M. Pathogenesis of human cytomegalovirus in the immunocompromised host. Nat Rev Microbiol 2021; 19:759-773. [PMID: 34168328 PMCID: PMC8223196 DOI: 10.1038/s41579-021-00582-z] [Citation(s) in RCA: 258] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2021] [Indexed: 12/16/2022]
Abstract
Human cytomegalovirus (HCMV) is a herpesvirus that infects ~60% of adults in developed countries and more than 90% in developing countries. Usually, it is controlled by a vigorous immune response so that infections are asymptomatic or symptoms are mild. However, if the immune system is compromised, HCMV can replicate to high levels and cause serious end organ disease. Substantial progress is being made in understanding the natural history and pathogenesis of HCMV infection and disease in the immunocompromised host. Serial measures of viral load defined the dynamics of HCMV replication and are now used routinely to allow intervention with antiviral drugs in individual patients. They are also used as pharmacodynamic read-outs to evaluate prototype vaccines that may protect against HCMV replication and to define immune correlates of this protection. This novel information is informing the design of randomized controlled trials of new antiviral drugs and vaccines currently under evaluation. In this Review, we discuss immune responses to HCMV and countermeasures deployed by the virus, the establishment of latency and reactivation from it, exogenous reinfection with additional strains, pathogenesis, development of end organ disease, indirect effects of infection, immune correlates of control of replication, current treatment strategies and the evaluation of novel vaccine candidates.
Collapse
Affiliation(s)
- Paul Griffiths
- Institute for Immunity and Transplantation, University College London, London, UK.
| | - Matthew Reeves
- grid.83440.3b0000000121901201Institute for Immunity and Transplantation, University College London, London, UK
| |
Collapse
|
49
|
Viral Vector Vaccines against Bluetongue Virus. Microorganisms 2020; 9:microorganisms9010042. [PMID: 33375723 PMCID: PMC7823852 DOI: 10.3390/microorganisms9010042] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/20/2022] Open
Abstract
Bluetongue virus (BTV), the prototype member of the genus Orbivirus (family Reoviridae), is the causative agent of an important livestock disease, bluetongue (BT), which is transmitted via biting midges of the genus Culicoides. To date, up to 29 serotypes of BTV have been described, which are classified as classical (BTV 1–24) or atypical (serotypes 25–27), and its distribution has been expanding since 1998, with important outbreaks in the Mediterranean Basin and devastating incursions in Northern and Western Europe. Classical vaccine approaches, such as live-attenuated and inactivated vaccines, have been used as prophylactic measures to control BT through the years. However, these vaccine approaches fail to address important matters like vaccine safety profile, effectiveness, induction of a cross-protective immune response among serotypes, and implementation of a DIVA (differentiation of infected from vaccinated animals) strategy. In this context, a wide range of recombinant vaccine prototypes against BTV, ranging from subunit vaccines to recombinant viral vector vaccines, have been investigated. This article offers a comprehensive outline of the live viral vectors used against BTV.
Collapse
|
50
|
Beijnen EMS, van Haren SD. Vaccine-Induced CD8 + T Cell Responses in Children: A Review of Age-Specific Molecular Determinants Contributing to Antigen Cross-Presentation. Front Immunol 2020; 11:607977. [PMID: 33424857 PMCID: PMC7786054 DOI: 10.3389/fimmu.2020.607977] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Infections are most common and most severe at the extremes of age, the young and the elderly. Vaccination can be a key approach to enhance immunogenicity and protection against pathogens in these vulnerable populations, who have a functionally distinct immune system compared to other age groups. More than 50% of the vaccine market is for pediatric use, yet to date vaccine development is often empiric and not tailored to molecular distinctions in innate and adaptive immune activation in early life. With modern vaccine development shifting from whole-cell based vaccines to subunit vaccines also comes the need for formulations that can elicit a CD8+ T cell response when needed, for example, by promoting antigen cross-presentation. While our group and others have identified many cellular and molecular determinants of successful activation of antigen-presenting cells, B cells and CD4+ T cells in early life, much less is known about the ontogeny of CD8+ T cell induction. In this review, we summarize the literature pertaining to the frequency and phenotype of newborn and infant CD8+ T cells, and any evidence of induction of CD8+ T cells by currently licensed pediatric vaccine formulations. In addition, we review the molecular determinants of antigen cross-presentation on MHC I and successful CD8+ T cell induction and discuss potential distinctions that can be made in children. Finally, we discuss recent advances in development of novel adjuvants and provide future directions for basic and translational research in this area.
Collapse
Affiliation(s)
- Elisabeth M. S. Beijnen
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
- 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
| | - Simon D. van Haren
- 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
|