1
|
Costantino M, Giudice V, Moccia G, Ragozzino M, Calabrese S, Caiazzo F, Beatrice M, Longanella W, Caruccio S, Iacuzzo C, Giugliano C, Marongiu MB, Genovese G, Serio B, Vozzella EA, Filippelli A, De Caro F. Sex, Age, and Previous Herpes Zoster Infection Role on Adverse Events Following Immunization with Adjuvanted Recombinant Vaccine. Pathogens 2025; 14:195. [PMID: 40005570 PMCID: PMC11858321 DOI: 10.3390/pathogens14020195] [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: 01/06/2025] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Adverse events following immunizations (AEFIs) with recombinant zoster vaccine (RZV) are underexplored in fragile populations. This study aims to assess incidence, duration, and characteristics of AEFIs, focusing on the impact of sex, age, and prior Herpes Zoster (HZ) infection in a frail population, including solid organ transplant recipients. We conducted an observational study on patients receiving RZV, and AEFIs were classified as local or systemic and analyzed for incidence, duration, and patterns across groups. We showed that females had a higher incidence of AEFIs (p = 0.02), both local and systemic symptoms, such as swelling +/- redness at the site of injection and fatigue, after the first and second doses. Younger adults experienced more systemic reactions, while older adults reported more local events (e.g., redness and swelling, p = 0.01). Moreover, patients with previous HZ infection exhibited a higher incidence of AEFIs after the second dose (68% vs. 38%, p = 0.001). In conclusion, sex, age, and clinical history significantly influenced AEFI incidence and manifestations. Therefore, it is important to personalize vaccination strategies in frail populations, by tailored administration and monitoring plans, especially in females and individuals with prior HZ infection, to improve vaccine safety and patient outcomes.
Collapse
Affiliation(s)
- Maria Costantino
- Department of Medicine, Surgery, and Dentistry, University of Salerno, 84081 Baronissi, Italy; (V.G.); (G.M.); (S.C.); (A.F.); (F.D.C.)
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | - Valentina Giudice
- Department of Medicine, Surgery, and Dentistry, University of Salerno, 84081 Baronissi, Italy; (V.G.); (G.M.); (S.C.); (A.F.); (F.D.C.)
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | - Giuseppina Moccia
- Department of Medicine, Surgery, and Dentistry, University of Salerno, 84081 Baronissi, Italy; (V.G.); (G.M.); (S.C.); (A.F.); (F.D.C.)
| | - Monica Ragozzino
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | - Salvatore Calabrese
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | - Francesco Caiazzo
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | - Massimo Beatrice
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | - Walter Longanella
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | - Simona Caruccio
- Department of Medicine, Surgery, and Dentistry, University of Salerno, 84081 Baronissi, Italy; (V.G.); (G.M.); (S.C.); (A.F.); (F.D.C.)
| | - Candida Iacuzzo
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | - Carmen Giugliano
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | | | - Giovanni Genovese
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | - Bianca Serio
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | - Emilia Anna Vozzella
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | - Amelia Filippelli
- Department of Medicine, Surgery, and Dentistry, University of Salerno, 84081 Baronissi, Italy; (V.G.); (G.M.); (S.C.); (A.F.); (F.D.C.)
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| | - Francesco De Caro
- Department of Medicine, Surgery, and Dentistry, University of Salerno, 84081 Baronissi, Italy; (V.G.); (G.M.); (S.C.); (A.F.); (F.D.C.)
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (M.R.); (S.C.); (F.C.); (M.B.); (W.L.); (C.I.); (C.G.); (G.G.); (B.S.)
| |
Collapse
|
2
|
Golichenari B, Heiat M, Rezaei E, Ramshini A, Sahebkar A, Gholipour N. Compromising the immunogenicity of diphtheria toxin-based immunotoxins through epitope engineering: An in silico approach. J Pharmacol Toxicol Methods 2025; 131:107571. [PMID: 39693813 DOI: 10.1016/j.vascn.2024.107571] [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: 09/07/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
Immunotoxins are genetically engineered recombinant proteins consisting of a targeting moiety, such as an antibody, and a cytotoxic toxin moiety of microbial origin. Pseudomonas exotoxin A and diphtheria toxin (DT) have been abundantly used in immunotoxins, with the latter applied as the toxin moiety of the FDA-approved drug Denileukin diftitox (ONTAK®). However, the use of immunotoxins provokes an adverse immune response in the host body against the toxin moiety, limiting their efficacy. In silico approaches have received increasing attention in protein engineering. In this study, the epitopes responsible for immunogenicity were identified through multiple platforms. By subtracting conserved and ligand-binding residues, K33, T111, and E112 were identified as common epitopes across all platforms. Substitution analysis evaluated alternative residues regarding their impact on protein stability, considering 19 different amino acid substitutions. Among the mutants explored, the T111A-E112G mutant exhibited the most destabilizing substitution for DT, thereby reducing immunogenicity. Finally, a 3D model of the mutant was generated and verified. The model was then docked with its native ligand NADH, and the complex's molecular behavior was simulated using molecular dynamics.
Collapse
Affiliation(s)
- Behrouz Golichenari
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Disease (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ehsan Rezaei
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amirreza Ramshini
- Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Nazila Gholipour
- Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Abstract
Substance use disorders (SUD) present a worldwide challenge with few effective therapies except for the relative efficacy of opioid pharmacotherapies, despite limited treatment access. However, the proliferation of illicit fentanyl use initiated a dramatic and cascading epidemic of lethal overdoses. This rise in fentanyl overdoses regenerated an interest in vaccine immunotherapy, which, despite an optimistic start in animal models over the past 50 years, yielded disappointing results in human clinical trials of vaccines against nicotine, stimulants (cocaine and methamphetamine), and opioids. After a brief review of clinical and selected preclinical vaccine studies, the "lessons learned" from the previous vaccine clinical trials are summarized, and then the newest challenge of a vaccine against fentanyl and its analogs is explored. Animal studies have made significant advances in vaccine technology for SUD treatment over the past 50 years, and the resulting anti-fentanyl vaccines show remarkable promise for ending this epidemic of fentanyl deaths.
Collapse
Affiliation(s)
- Thomas R Kosten
- Waggoner Professor of Psychiatry, Pharmacology, Neuroscience, Immunology, Baylor College of Medicine, Houston
| |
Collapse
|
4
|
Purcell RA, Theisen RM, Arnold KB, Chung AW, Selva KJ. Polyfunctional antibodies: a path towards precision vaccines for vulnerable populations. Front Immunol 2023; 14:1183727. [PMID: 37600816 PMCID: PMC10433199 DOI: 10.3389/fimmu.2023.1183727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Collapse
Affiliation(s)
- Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Robert M. Theisen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Kosten TR, Domingo CB, Haile CN, Nielsen DA. A Clinical Trial of Entolimod a TLR-5 Adjuvant for Vaccines Using Diphtheria or Tetanus as Carrier Proteins. Vaccines (Basel) 2022; 10:1592. [PMID: 36298456 PMCID: PMC9611255 DOI: 10.3390/vaccines10101592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 11/30/2022] Open
Abstract
Anti-drug vaccines previously failed clinical trials because they did not provide a sufficient titer or duration of antibodies (AB), but new adjuvants enhance both AB titers and efficacy duration. This clinical trial assessed AB titers after a single booster of commercial tetanus-diphtheria (Td) vaccine in 40 males randomized as 15 to Td alone and 25 to Td combined with the TLR5 adjuvant, Entolimod (Ent). Ent significantly increased ABs against diphtheria (DPT) (0.46 vs. 0.29 IU/mL increase; n = 40, p < 0.05), but against tetanus (TT) only if baseline TT AB was below 3 IU/mL (3.1 vs. 2.1 IU/mL; n = 20; p < 0.05). These 20 participants also showed a two-fold increase in anti-TT AB titer more often when given Ent than non-Ent (33% vs. 82%) (p < 0.03). Anti-Ent AB was low and appeared unlikely to reduce Ent efficacy after repeated Ent administration. Medical safety was excellent, and a TLR5 missense polymorphism reduced anti-DPT AB production, but Ent increased anti-DPT AB titers to levels induced in subjects with genetically “normal” TRL5 functioning. Further clinical testing of TLR5 adjuvants like Ent seems warranted for anti-drug vaccines.
Collapse
Affiliation(s)
- Thomas R. Kosten
- Department of Psychiatry and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Coreen B. Domingo
- Department of Psychiatry and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Colin N. Haile
- Department of Psychology, University of Houston, Houston, TX 77004, USA
| | - David A. Nielsen
- Department of Psychiatry, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
6
|
Smatti MK, Alkhatib HA, Al Thani AA, Yassine HM. Will Host Genetics Affect the Response to SARS-CoV-2 Vaccines? Historical Precedents. Front Med (Lausanne) 2022; 9:802312. [PMID: 35360730 PMCID: PMC8962369 DOI: 10.3389/fmed.2022.802312] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/10/2022] [Indexed: 11/25/2022] Open
Abstract
Recent progress in genomics and bioinformatics technologies have allowed for the emergence of immunogenomics field. This intersection of immunology and genetics has broadened our understanding of how the immune system responds to infection and vaccination. While the immunogenetic basis of the huge clinical variability in response to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is currently being extensively studied, the host genetic determinants of SARS-CoV-2 vaccines remain largely unknown. Previous reports evidenced that vaccines may not protect all populations or individuals equally, due to multiple host- and vaccine-specific factors. Several studies on vaccine response to measles, rubella, hepatitis B, smallpox, and influenza highlighted the contribution of genetic mutations or polymorphisms in modulating the innate and adaptive immunity following vaccination. Specifically, genetic variants in genes encoding virus receptors, antigen presentation, cytokine production, or related to immune cells activation and differentiation could influence how an individual responds to vaccination. Although such knowledge could be utilized to generate personalized vaccine strategies to optimize the vaccine response, studies in this filed are still scarce. Here, we briefly summarize the scientific literature related to the immunogenetic determinants of vaccine-induced immunity, highlighting the possible role of host genetics in response to SARS-CoV-2 vaccines as well.
Collapse
Affiliation(s)
- Maria K. Smatti
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | | | - Hadi M. Yassine
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Biomedical Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
7
|
Colucci M, De Santis E, Totti B, Miroballo M, Tamiro F, Rossi G, Piepoli A, De Vincentis G, Greco A, Mangia A, Cianci R, Di Mauro L, Miscio G, Giambra V. Associations between Allelic Variants of the Human IgH 3' Regulatory Region 1 and the Immune Response to BNT162b2 mRNA Vaccine. Vaccines (Basel) 2021; 9:1207. [PMID: 34696315 PMCID: PMC8540755 DOI: 10.3390/vaccines9101207] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/11/2021] [Accepted: 10/16/2021] [Indexed: 01/28/2023] Open
Abstract
The escalation of Coronavirus disease 2019 (COVID-19) has required the development of safe and effective vaccines against the severe acute respiratory syndrome coronavirus 2-associated (SARS-CoV-2), which is the causative agent of the disease. Here, we determined the levels of antibodies, antigen-specific B cells, against a recombinant GFP-tagged SARS-CoV-2 spike (S) protein and total T and NK cell subsets in subjects up to 20 days after the injection of the BNT162b2 (Pfizer-BioNTech) vaccine using a combined approach of serological and flow cytometry analyses. In former COVID-19 patients and highly responsive individuals, a significant increase of antibody production was detected, simultaneous with an expansion of antigen-specific B cell response and the total number of NK-T cells. Additionally, through a genetic screening of a specific polymorphic region internal to the 3' regulatory region 1 (3'RR1) of human immunoglobulin constant-gene (IgH) locus, we identified different single-nucleotide polymorphic (SNP) variants associated with either highly or lowly responsive subjects. Taken together, these results suggest that favorable genetic backgrounds and immune profiles support the progression of an effective response to BNT162b2 vaccination.
Collapse
Affiliation(s)
- Mattia Colucci
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (M.C.); (E.D.S.); (B.T.); (M.M.); (F.T.)
| | - Elisabetta De Santis
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (M.C.); (E.D.S.); (B.T.); (M.M.); (F.T.)
| | - Beatrice Totti
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (M.C.); (E.D.S.); (B.T.); (M.M.); (F.T.)
| | - Mattia Miroballo
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (M.C.); (E.D.S.); (B.T.); (M.M.); (F.T.)
| | - Francesco Tamiro
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (M.C.); (E.D.S.); (B.T.); (M.M.); (F.T.)
| | - Giovanni Rossi
- Department of Hematology and Stem Cell Transplant Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy;
| | - Ada Piepoli
- Hospital Health Department, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (A.P.); (G.D.V.)
| | - Gabriella De Vincentis
- Hospital Health Department, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (A.P.); (G.D.V.)
| | - Antonio Greco
- Complex Structure of Geriatrics, Department of Medical Sciences, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy;
| | - Alessandra Mangia
- Liver Unit, Department of Medical Sciences, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy;
| | - Rossella Cianci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Rome, Italy;
| | - Lazzaro Di Mauro
- Clinical Laboratory Analysis and Transfusional Medicine, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (L.D.M.); (G.M.)
| | - Giuseppe Miscio
- Clinical Laboratory Analysis and Transfusional Medicine, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (L.D.M.); (G.M.)
| | - Vincenzo Giambra
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (M.C.); (E.D.S.); (B.T.); (M.M.); (F.T.)
| |
Collapse
|
8
|
Knuutila A, Dalby T, Barkoff AM, Jørgensen CS, Fuursted K, Mertsola J, Markey K, He Q. Differences in epitope-specific antibodies to pertussis toxin after infection and acellular vaccinations. Clin Transl Immunology 2020; 9:e1161. [PMID: 32765879 PMCID: PMC7396262 DOI: 10.1002/cti2.1161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/15/2020] [Accepted: 07/02/2020] [Indexed: 11/29/2022] Open
Abstract
Objectives Pertussis toxin (PT) is a component of all acellular pertussis vaccines. PT must be detoxified to be included in acellular vaccines, which results in conformational changes in the functional epitopes of PTs. Therefore, induced epitope‐specific antibodies to PT may vary after vaccinations or natural infections, and this information could reveal biomarkers implicated for protection and successful immunisation. Methods Pertussis toxin epitope‐specific antibodies in sera from 152 vaccinated children and 72 serologically confirmed patients were tested with a blocking ELISA, based on monoclonal antibodies that target protective PT epitopes. Results All study groups induced considerable antibody titres to subunit 1 (S1). Of interest, S3 7E10‐specific antibodies were present in patients, but not after vaccinations (P < 0.001). The impact of glutaraldehyde treatment of PT was visible on epitope 1D7 (S1), whereas epitopes 1B7 (S1) and 10D (S1) were more preserved. Antibodies to these epitopes were higher after three primary vaccine doses than after a single booster dose. Conclusion The high amount of 7E10‐specific antibodies in patients suggests this epitope might be functionally relevant in protection. The overall characteristics of epitope‐specific antibodies are influenced by infection or vaccination background, by the used detoxification method of PT and by the amount of the toxin used in immunisation.
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
| | - Kevin Markey
- National Institute for Biological Standards and Control Potters Bar UK
| | - Qiushui He
- Institute of Biomedicine University of Turku Turku Finland.,Department of Medical Microbiology Capital Medical University Beijing China
| |
Collapse
|
9
|
OMIC Technologies and Vaccine Development: From the Identification of Vulnerable Individuals to the Formulation of Invulnerable Vaccines. J Immunol Res 2019; 2019:8732191. [PMID: 31183393 PMCID: PMC6512027 DOI: 10.1155/2019/8732191] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 12/16/2022] Open
Abstract
Routine vaccination is among the most effective clinical interventions to prevent diseases as it is estimated to save over 3 million lives every year. However, the full potential of global immunization programs is not realised because population coverage is still suboptimal. This is also due to the inadequate immune response and paucity of informative correlates of protection upon immunization of vulnerable individuals such as newborns, preterm infants, pregnant women, and elderly individuals as well as those patients affected by chronic and immune compromising medical conditions. In addition, these groups are undervaccinated for a number of reasons, including lack of awareness of vaccine-preventable diseases and uncertainty or misconceptions about the safety and efficacy of vaccination by parents and healthcare providers. The presence of these nonresponders/undervaccinated individuals represents a major health and economic burden to society, which will become particularly difficult to address in settings with limited public resources. This review describes innovative and experimental approaches that can help identify specific genomic profiles defining nonresponder individuals for whom specific interventions might be needed. We will provide examples that show how such information can be useful to identify novel biomarkers of safety and immunogenicity for future vaccine trials. Finally, we will discuss how system biology “OMICs” data can be used to design bioinformatic tools to predict the vaccination outcome providing genetic and molecular “signatures” of protective immune response. This strategy may soon enable identification of signatures highly predictive of vaccine safety, immunogenicity, and efficacy/protection thereby informing personalized vaccine interventions in vulnerable populations.
Collapse
|
10
|
Pezeshki A, Ovsyannikova IG, McKinney BA, Poland GA, Kennedy RB. The role of systems biology approaches in determining molecular signatures for the development of more effective vaccines. Expert Rev Vaccines 2019; 18:253-267. [PMID: 30700167 DOI: 10.1080/14760584.2019.1575208] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Emerging infectious diseases are a major threat to public health, and while vaccines have proven to be one of the most effective preventive measures for infectious diseases, we still do not have safe and effective vaccines against many human pathogens, and emerging diseases continually pose new threats. The purpose of this review is to discuss how the creation of vaccines for these new threats has been hindered by limitations in the current approach to vaccine development. Recent advances in high-throughput technologies have enabled scientists to apply systems biology approaches to collect and integrate increasingly large datasets that capture comprehensive biological changes induced by vaccines, and then decipher the complex immune response to those vaccines. AREAS COVERED This review covers advances in these technologies and recent publications that describe systems biology approaches to understanding vaccine immune responses and to understanding the rational design of new vaccine candidates. EXPERT OPINION Systems biology approaches to vaccine development provide novel information regarding both the immune response and the underlying mechanisms and can inform vaccine development.
Collapse
Affiliation(s)
| | | | - Brett A McKinney
- b Department of Mathematics , University of Tulsa , Tulsa , OK , USA.,c Tandy School of Computer Science , University of Tulsa , Tulsa , OK , USA
| | - Gregory A Poland
- a Mayo Vaccine Research Group , Mayo Clinic , Rochester , MN , USA
| | | |
Collapse
|
11
|
Poyntz HC, Jones A, Jauregui R, Young W, Gestin A, Mooney A, Lamiable O, Altermann E, Schmidt A, Gasser O, Weyrich L, Jolly CJ, Linterman MA, Gros GL, Hawkins ED, Forbes-Blom E. Genetic regulation of antibody responsiveness to immunization in substrains of BALB/c mice. Immunol Cell Biol 2018; 97:39-53. [PMID: 30152893 PMCID: PMC6378622 DOI: 10.1111/imcb.12199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 08/20/2018] [Accepted: 08/23/2018] [Indexed: 12/17/2022]
Abstract
Antibody‐mediated immunity is highly protective against disease. The majority of current vaccines confer protection through humoral immunity, but there is high variability in responsiveness across populations. Identifying immune mechanisms that mediate low antibody responsiveness may provide potential strategies to boost vaccine efficacy. Here, we report diverse antibody responsiveness to unadjuvanted as well as adjuvanted immunization in substrains of BALB/c mice, resulting in high and low antibody response phenotypes. Furthermore, these antibody phenotypes were not affected by changes in environmental factors such as the gut microbiota composition. Antigen‐specific B cells following immunization had a marked difference in capability to class switch, resulting in perturbed IgG isotype antibody production. In vitro, a B‐cell intrinsic defect in the regulation of class‐switch recombination was identified in mice with low IgG antibody production. Whole genome sequencing identified polymorphisms associated with the magnitude of antibody produced, and we propose candidate genes that may regulate isotype class‐switching capability. This study highlights that mice sourced from different vendors can have significantly altered humoral immune response profiles, and provides a resource to interrogate genetic regulators of antibody responsiveness. Together these results further our understanding of immune heterogeneity and suggest additional research on the genetic influences of adjuvanted vaccine strategies is warranted for enhancing vaccine efficacy.
Collapse
Affiliation(s)
- Hazel C Poyntz
- Malaghan Institute of Medical Research, Victoria University of Wellington, Gate 7, Kelburn Parade, Wellington, 6012, New Zealand.,High-Value Nutrition National Science Challenge, New Zealand
| | - Angela Jones
- Malaghan Institute of Medical Research, Victoria University of Wellington, Gate 7, Kelburn Parade, Wellington, 6012, New Zealand
| | - Ruy Jauregui
- Grasslands Research Centre, AgResearch, Tennent Drive, Palmerston North, New Zealand
| | - Wayne Young
- Grasslands Research Centre, AgResearch, Tennent Drive, Palmerston North, New Zealand.,Riddet Institute, Massey University, Palmerston North, 4474, New Zealand
| | - Aurélie Gestin
- Malaghan Institute of Medical Research, Victoria University of Wellington, Gate 7, Kelburn Parade, Wellington, 6012, New Zealand
| | - Anna Mooney
- Malaghan Institute of Medical Research, Victoria University of Wellington, Gate 7, Kelburn Parade, Wellington, 6012, New Zealand
| | - Olivier Lamiable
- Malaghan Institute of Medical Research, Victoria University of Wellington, Gate 7, Kelburn Parade, Wellington, 6012, New Zealand
| | - Eric Altermann
- Grasslands Research Centre, AgResearch, Tennent Drive, Palmerston North, New Zealand.,Riddet Institute, Massey University, Palmerston North, 4474, New Zealand
| | - Alfonso Schmidt
- Malaghan Institute of Medical Research, Victoria University of Wellington, Gate 7, Kelburn Parade, Wellington, 6012, New Zealand
| | - Olivier Gasser
- Malaghan Institute of Medical Research, Victoria University of Wellington, Gate 7, Kelburn Parade, Wellington, 6012, New Zealand
| | - Laura Weyrich
- Australian Centre for Ancient DNA, University of Adelaide, North Terrace, Adelaide, SA, 5005, Australia
| | - Christopher J Jolly
- Centenary Institute and Sydney Medical School, University of Sydney, Missenden Road, Sydney, NSW, 2050, Australia
| | - Michelle A Linterman
- Lymphocyte Signaling and Development, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Graham Le Gros
- Malaghan Institute of Medical Research, Victoria University of Wellington, Gate 7, Kelburn Parade, Wellington, 6012, New Zealand
| | - Edwin D Hawkins
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Elizabeth Forbes-Blom
- Malaghan Institute of Medical Research, Victoria University of Wellington, Gate 7, Kelburn Parade, Wellington, 6012, New Zealand.,High-Value Nutrition National Science Challenge, New Zealand
| |
Collapse
|
12
|
Yudin NS, Igoshin AV, Lutova SL, Gon Y, Voevoda MI, Belyavskaya VA. ASSOCIATION BETWEEN POLYMORPHISMS IN GENES ENCODING 2′-5′-OLIGOADENYLATE SYNTHETASES AND THE HUMORAL IMMUNE RESPONSE UPON VACCINATION AGAINST TICK-BORNE ENCEPHALITIS. Vavilovskii Zhurnal Genet Selektsii 2018. [DOI: 10.18699/vj18.381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Vaccination forms active immunity and represents an effective way of preventing tick-borne encephalitis (TBE). However, excessive vaccination is unjustified in terms of economics and medical ethics. One of the individualized approaches to vaccines is the selection of vaccine doses depending on the expected levels of immune response. Therefore, there is a need for new methods for assessing potential human immune responses prior to vaccination. The aim of this study was to determine possible association between single nucleotide polymorphisms (SNPs) located within OAS2 and OAS3 genes, which have been previously associated with the development of severe forms of TBE, and the formation of antibodies and cytokines upon vaccination against TBE. The study involved 97 volunteers of both sexes who had not previously been vaccinated against TBE and had no contact with ticks. Venous blood samples were collected one month after vaccination against TBE using the EnceVir vaccine. Levels of specific IgG antibodies against tick-borne encephalitis virus and interleukin 4 (IL-4) were analyzed. Genomic DNA samples were genotyped for the SNPs rs2285932, rs2072136, rs1293762, rs15895 and rs1732778 in genes encoding 2’-5’-oligoadenylate synthetases OAS2 and OAS3. Antibody production in response to vaccine administration was significantly associated with SNP rs1732778 in the regulatory region of the OAS2 gene. This indicator was significantly higher in people with heterozygous genotypes G/A as compared to people with homozygous genotypes G/G and A/A. Carriers of the A allele (G/A or A/A genotypes) of the same SNP had reduced IL-4 levels as compared to the homozygous G/G individuals. Thus, the data obtained indicate that SNP rs1732778 in the regulatory region of the OAS2 gene correlates with the formation of antiviral IgG antibodies and changes in IL-4 levels upon vaccination. Evidently, the genetic polymorphism in OAS2 gene should be considered when performing individualized TBE vaccinations.
Collapse
|
13
|
Živković I, Petrović R, Arsenović-Ranin N, Petrušić V, Minić R, Bufan B, Popović O, Leposavić G. Sex bias in mouse humoral immune response to influenza vaccine depends on the vaccine type. Biologicals 2018; 52:18-24. [PMID: 29426673 DOI: 10.1016/j.biologicals.2018.01.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 10/18/2022] Open
Abstract
The study explored influence of biological sex on development of humoral immune response to seasonal trivalent whole inactivated virus (WIV) and split virus (SV) influenza vaccines in outbred Swiss mouse model. To this end, mice of both sexes were immunized with WIV (WIV mice) and SV vaccines (SV mice) and examined for specific antibody response. Irrespective of sex, total IgG and neutralizing antibody responses to distinct virus strains were weaker in SV than in WIV mice. In WIV mice of both sexes, irrespective of strain specificity, IgG isotype response was dominated by IgG2a antibodies, while in SV mice nearly equal representation of IgG2a and IgG1 antibodies was found. The analyses of sex differences showed higher titers of H1N1-specific and both H1N1- and H3N2-specific total IgG and neutralizing antibodies in female WIV and SV mice, respectively. Additionally, sexual dimorphism in IgG subclass profile depended on vaccine type. Specifically, compared with males, in females WIV shifted IgG2a/IgG1 antibody ratio towards IgG2a isotype on the account of weaker IgG1 response, whereas in SV mice, irrespective of virus strain, IgG2a and IgG1 isotypes were equally represented in both sexes. These findings indicate the vaccine type-dependent sex bias in antibody response to inactivated influenza vaccines.
Collapse
Affiliation(s)
- Irena Živković
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Raisa Petrović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Vladimir Petrušić
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Rajna Minić
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Biljana Bufan
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Olga Popović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Gordana Leposavić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
14
|
Elding Larsson H, Lynch KF, Lönnrot M, Haller MJ, Lernmark Å, Hagopian WA, She JX, Simell O, Toppari J, Ziegler AG, Akolkar B, Krischer JP, Rewers MJ, Hyöty H. Pandemrix® vaccination is not associated with increased risk of islet autoimmunity or type 1 diabetes in the TEDDY study children. Diabetologia 2018; 61:193-202. [PMID: 28990147 PMCID: PMC5774660 DOI: 10.1007/s00125-017-4448-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 08/14/2017] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESIS During the A/H1N1 2009 (A/California/04/2009) pandemic, mass vaccination with a squalene-containing vaccine, Pandemrix®, was performed in Sweden and Finland. The vaccination was found to cause narcolepsy in children and young adults with the HLA-DQ 6.2 haplotype. The aim of this study was to investigate if exposure to Pandemrix® similarly increased the risk of islet autoimmunity or type 1 diabetes. METHODS In The Environmental Determinants of Diabetes in the Young (TEDDY) study, children are followed prospectively for the development of islet autoimmunity and type 1 diabetes. In October 2009, when the mass vaccination began, 3401 children at risk for islet autoimmunity and type 1 diabetes were followed in Sweden and Finland. Vaccinations were recorded and autoantibodies against insulin, GAD65 and insulinoma-associated protein 2 were ascertained quarterly before the age of 4 years and semi-annually thereafter. RESULTS By 5 August 2010, 2413 of the 3401 (71%) children observed as at risk for an islet autoantibody or type 1 diabetes on 1 October 2009 had been vaccinated with Pandemrix®. By 31 July 2016, 232 children had at least one islet autoantibody before 10 years of age, 148 had multiple islet autoantibodies and 96 had developed type 1 diabetes. The risk of islet autoimmunity was not increased among vaccinated children. The HR (95% CI) for the appearance of at least one islet autoantibody was 0.75 (0.55, 1.03), at least two autoantibodies was 0.85 (0.57, 1.26) and type 1 diabetes was 0.67 (0.42, 1.07). In Finland, but not in Sweden, vaccinated children had a lower risk of islet autoimmunity (0.47 [0.29, 0.75]), multiple autoantibodies (0.50 [0.28, 0.90]) and type 1 diabetes (0.38 [0.20, 0.72]) compared with those who did not receive Pandemrix®. The analyses were adjusted for confounding factors. CONCLUSIONS/INTERPRETATION Children with an increased genetic risk for type 1 diabetes who received the Pandemrix® vaccine during the A/H1N1 2009 pandemic had no increased risk of islet autoimmunity, multiple islet autoantibodies or type 1 diabetes. In Finland, the vaccine was associated with a reduced risk of islet autoimmunity and type 1 diabetes.
Collapse
Affiliation(s)
- Helena Elding Larsson
- Department of Clinical Sciences Malmö, Lund University CRC, Skåne University Hospital SUS, Jan Waldenströms gata 35; 60:11, 20502, Malmö, Sweden.
| | - Kristian F Lynch
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Maria Lönnrot
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- Department of Dermatology, Tampere University Hospital, Tampere, Finland
| | - Michael J Haller
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Åke Lernmark
- Department of Clinical Sciences Malmö, Lund University CRC, Skåne University Hospital SUS, Jan Waldenströms gata 35; 60:11, 20502, Malmö, Sweden
| | | | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Olli Simell
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Jorma Toppari
- Department of Pediatrics, Turku University Hospital, Turku, Finland
- Department of Physiology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Anette-G Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, and Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Forschergruppe Diabetes e.V., Neuherberg, Germany
| | - Beena Akolkar
- National Institute of Diabetes & Digestive & Kidney Diseases, Bethesda, MD, USA
| | - Jeffrey P Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Marian J Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Heikki Hyöty
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| |
Collapse
|
15
|
Poland GA, Ovsyannikova IG, Kennedy RB. Personalized vaccinology: A review. Vaccine 2017; 36:5350-5357. [PMID: 28774561 PMCID: PMC5792371 DOI: 10.1016/j.vaccine.2017.07.062] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/19/2017] [Accepted: 07/12/2017] [Indexed: 12/18/2022]
Abstract
At the current time, the field of vaccinology remains empirical in many respects. Vaccine development, vaccine immunogenicity, and vaccine efficacy have, for the most part, historically been driven by an empiric “isolate-inactivate-inject” paradigm. In turn, a population-level public health paradigm of “the same dose for everyone for every disease” model has been the normative thinking in regard to prevention of vaccine-preventable infectious diseases. In addition, up until recently, no vaccines had been designed specifically to overcome the immunosenescence of aging, consistent with a post-WWII mentality of developing vaccines and vaccine programs for children. It is now recognized that the current lack of knowledge concerning how immune responses to vaccines are generated is a critical barrier to understanding poor vaccine responses in the elderly and in immunoimmaturity, discovery of new correlates of vaccine immunogenicity (vaccine response biomarkers), and a directed approach to new vaccine development. The new fields of vaccinomics and adversomics provide models that permit global profiling of the innate, humoral, and cellular immune responses integrated at a systems biology level. This has advanced the science beyond that of reductionist scientific approaches by revealing novel interactions between and within the immune system and other biological systems (beyond transcriptional level), which are critical to developing “downstream” adaptive humoral and cellular responses to infectious pathogens and vaccines. Others have applied systems level approaches to the study of antibody responses (a.k.a. “systems serology”), [1] high-dimensional cell subset immunophenotyping through CyTOF, [2,3] and vaccine induced metabolic changes [4]. In turn, this knowledge is being utilized to better understand the following: identifying who is at risk for which infections; the level of risk that exists regarding poor immunogenicity and/or serious adverse events; and the type or dose of vaccine needed to fully protect an individual. In toto, such approaches allow for a personalized approach to the practice of vaccinology, analogous to the substantial inroads that individualized medicine is playing in other fields of human health and medicine. Herein we briefly review the field of vaccinomics, adversomics, and personalized vaccinology.
Collapse
Affiliation(s)
- G A Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA.
| | - I G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA
| | - R B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
16
|
Sakai A, Noguchi E, Fukushima T, Tagawa M, Iwabuchi A, Kita M, Kakisaka K, Miyasaka A, Takikawa Y, Sumazaki R. Identification of amino acids in antigen-binding site of class II HLA proteins independently associated with hepatitis B vaccine response. Vaccine 2016; 35:703-710. [PMID: 28043736 DOI: 10.1016/j.vaccine.2016.08.068] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/29/2016] [Accepted: 08/23/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Genetic factors in class II human leukocyte antigen (HLA) have been reported to be associated with inter-individual variation in hepatitis B virus (HBV) vaccine response. However, the mechanism underlying the associations remains elusive. In particular, the broad linkage disequilibrium in HLA region complicates the localization of the independent effects of genetic variants. Thus, the present study aimed to identify the most probable causal variations in class II HLA loci involved in the immune response to HBV vaccine. METHODS We performed a case-control study to assess whether HLA-DRB1, -DQB1, and -DPB1 4-digit alleles were associated with the response to primary HBV vaccination in 574 healthy Japanese students. To identify causative variants, we next assessed independently associated amino acid variants in these loci using conditional logistic regression analysis. Furthermore, to clarify the functional effects of these variants on HLA proteins, we performed computational structural studies. RESULTS HLA-DRB1∗01:01, HLA-DRB1∗08:03, HLA-DQB1∗05:01, and HLA-DPB1∗04:02 were significantly associated with sufficient response, whereas HLA-DPB1∗05:01 was associated with poor response. We then identified amino acids independently associated with sufficient response, namely, leucine at position 26 of HLA-DRβ1 and glycine-glycine-proline-methionine at positions 84-87 of HLA-DPβ1. These amino acids were located in antigen-binding pocket 4 of HLA-DR and pocket 1 of HLA-DP, respectively, which are important structures for selective binding of antigenic peptides. In addition, the detected variations in HLA-DP protein were responsible for the differences in the electrostatic potentials of the pocket, which can explain in part the sufficient/poor vaccine responses. CONCLUSION HLA-DRβ1 position 26 and HLA-DPβ1 positions 84-87 are independently associated with anti-HBs production against HBV vaccine. Our results suggest that HBsAg presentation through these HLA pocket structures plays an important role in the inter-individual variability of HBV vaccination.
Collapse
Affiliation(s)
- Aiko Sakai
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.
| | - Emiko Noguchi
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.
| | - Takashi Fukushima
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.
| | - Manabu Tagawa
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Atsushi Iwabuchi
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masaki Kita
- Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan
| | - Keisuke Kakisaka
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| | - Akio Miyasaka
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| | - Yasuhiro Takikawa
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| | - Ryo Sumazaki
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
17
|
Gröndahl-Yli-Hannuksela K, Vahlberg T, Ilonen J, Mertsola J, He Q. Polymorphism of IL-10 gene promoter region: association with T cell proliferative responses after acellular pertussis vaccination in adults. Immunogenetics 2016; 68:733-41. [PMID: 27282930 DOI: 10.1007/s00251-016-0923-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/01/2016] [Indexed: 10/21/2022]
Abstract
Individual variation in immune responses is always encountered after vaccination. This phenomenon is also seen after acellular pertussis vaccination. The aim of this present study was to investigate whether single nucleotide polymorphisms (SNPs) in the IL-10 gene promoter region (rs1800890, rs1800896, rs1800871), IL-12B (rs2546890), IL-12RB1 (rs372889), IL-17A (rs2275913), and IL-23R (rs11209026) affect the immune responses after acellular pertussis vaccination. The T cell proliferative response was evaluated in 38 Finnish young adults who received a second booster dose of a vaccine combination of diphtheria, tetanus, and acellular pertussis, 10 years after the previous booster. The response was evaluated with a proliferation assay in which vaccine antigens pertussis toxin (PT), filamentous hemagglutinin (FHA), and pertactin (PRN) were used for the stimulation, before and 1 month after the second vaccination. Specific proliferation of peripheral blood mononuclear cells against pertussis antigens was affected by IL-10 SNP in the promoter region at position -1082 (A>G, rs1800896). One month after the vaccination, subjects with the AA and AG genotypes had a significantly higher T cell proliferative response against PT and FHA compared to those with the GG genotype. Subjects with the GG genotype had the lowest responses. As a conclusion, our preliminary results indicate that IL-10 SNP -1082 might play an important role in T cell-mediated immune responses after acellular pertussis vaccination.
Collapse
Affiliation(s)
- Kirsi Gröndahl-Yli-Hannuksela
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland.,Department of Infectious Disease Surveillance and Control, National Institute for Health and Welfare, Turku, Finland
| | - Tero Vahlberg
- Department of Biostatistics, University of Turku, Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, University of Turku, Turku, Finland
| | - Jussi Mertsola
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Qiushui He
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland. .,Department of Infectious Disease Surveillance and Control, National Institute for Health and Welfare, Turku, Finland. .,Department of Medical Microbiology, Capital Medical University, Beijing, China.
| |
Collapse
|
18
|
Gianchecchi E, Torelli A, Piccini G, Piccirella S, Montomoli E. N. meningitidis and TLR Polymorphisms: A Fascinating Immunomodulatory Network. Vaccines (Basel) 2016; 4:vaccines4020020. [PMID: 27240411 PMCID: PMC4931637 DOI: 10.3390/vaccines4020020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/29/2016] [Accepted: 05/20/2016] [Indexed: 12/21/2022] Open
Abstract
N. meningitidis infections represent a global health problem that can lead to the development of serious permanent sequelae. Although the use of antibiotics and prevention via vaccination have reduced the incidence of meningococcal disease, our understanding regarding N. meningitidis pathogenesis is still limited, especially of those mechanisms responsible for IMD and fulminant or deadly septic shock. These severe clinical presentations occur in a limited number of subjects, whereas about 10% of healthy individuals are estimated to carry the bacteria as a commensal. Since TLR activation is involved in the defense against N. meningitidis, several studies have highlighted the association between host TLR SNPs and a higher susceptibility and severity of N. meningitidis infections. Moreover, TLR SNPs induced variations in immunological responses and in their persistence upon vaccination against meningococcal disease. In the absence of mass vaccination programs, the early identification of risk factors for meningococcal disease would be recommended in order to start immunization strategies and antibiotic treatment in those subjects carrying the risk variants. In addition, it could allow us to identify individuals with a higher risk for severe disease and sequelae in order to develop a personalized healthcare of high-risk subjects based on their genomic profile. In this review, we have illustrated important preliminary correlations between TLR variants and meningococcal susceptibility/severity and with vaccine-induced immune responses.
Collapse
Affiliation(s)
| | - Alessandro Torelli
- Strada del Petriccio e Belriguardo, 53100 Siena, Italy.
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| | | | | | - Emanuele Montomoli
- Strada del Petriccio e Belriguardo, 53100 Siena, Italy.
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy.
| |
Collapse
|
19
|
Mentzer AJ, O'Connor D, Pollard AJ, Hill AVS. Searching for the human genetic factors standing in the way of universally effective vaccines. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0341. [PMID: 25964463 DOI: 10.1098/rstb.2014.0341] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Vaccines have revolutionized modern public health. The effectiveness of some vaccines is limited by the variation in response observed between individuals and across populations. There is compelling evidence that a significant proportion of this variability can be attributed to human genetic variation, especially for those vaccines administered in early life. Identifying and understanding the determinants of this variation could have a far-reaching influence upon future methods of vaccine design and deployment. In this review, we summarize the genetic studies that have been undertaken attempting to identify the genetic determinants of response heterogeneity for the vaccines against hepatitis B, measles and rubella. We offer a critical appraisal of these studies and make a series of suggestions about how modern genetic techniques, including genome-wide association studies, could be used to characterize the genetic architecture of vaccine response heterogeneity. We conclude by suggesting how the findings from such studies could be translated to improve vaccine effectiveness and target vaccination in a more cost-effective manner.
Collapse
Affiliation(s)
- Alexander J Mentzer
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Daniel O'Connor
- Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK Oxford Biomedical Research Centre, Oxford OX3 7LE, UK
| | - Andrew J Pollard
- Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK Oxford Biomedical Research Centre, Oxford OX3 7LE, UK
| | - Adrian V S Hill
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK Oxford Biomedical Research Centre, Oxford OX3 7LE, UK
| |
Collapse
|
20
|
Georgopoulos AP, James LM, Mahan MY, Joseph J, Georgopoulos A, Engdahl BE. Reduced Human Leukocyte Antigen (HLA) Protection in Gulf War Illness (GWI). EBioMedicine 2015; 3:79-85. [PMID: 26870819 PMCID: PMC4739436 DOI: 10.1016/j.ebiom.2015.11.037] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/16/2015] [Accepted: 11/20/2015] [Indexed: 11/29/2022] Open
Abstract
Background Gulf War Illness (GWI) is a disease of unknown etiology with symptoms suggesting the involvement of an immune process. Here we tested the hypothesis that Human Leukocyte Antigen (HLA) composition might differ between veterans with and without GWI. Methods We identified 144 unique alleles of Class I and II HLA genes in 82 veterans (66 with and 16 without GWI). We tested the hypothesis that a subset of HLA alleles may classify veterans in their respective group using a stepwise linear discriminant analysis. In addition, each participant rated symptom severity in 6 domains according to established GWI criteria, and an overall symptom severity was calculated. Findings We found 6 Class II alleles that classified participants 84.1% correctly (13/16 control and 56/66 GWI). The number of copies of the 6 alleles was significantly higher in the control group, suggesting a protective role. This was supported by a significant negative dependence of overall symptom severity on the number of allele copies, such that symptom severity was lower in participants with larger numbers of allele copies. Interpretation These results indicate a reduced HLA protection (i.e. genetic susceptibility) in veterans with GWI. Funding University of Minnesota and U.S. Department of Veterans Affairs. Differences in Human Leukocyte Antigen distinguished veterans with Gulf War Illness (GWI) from healthy Gulf War era veterans. Veterans with GWI show genetic susceptibility which is associated with increased severity of hallmark symptoms of GWI.
A large number of Gulf War veterans suffer from diffuse and debilitating symptoms that resemble altered immune functioning. This study evaluated whether the Human Leukocyte Antigen (HLA) gene, which is central to immune functioning, differs between veterans with GWI and unaffected Gulf War veterans. The findings highlight robust differences in HLA composition between the two groups and suggest that veterans with GWI are characterized by genetic susceptibility that confers risk for typical GWI symptoms. These findings provide compelling genetic evidence of immune dysfunction in GWI.
Collapse
Affiliation(s)
- Apostolos P Georgopoulos
- Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Center for Cognitive Sciences, University of Minnesota, Minneapolis, MN 55455, USA; Graduate Program in Biomedical Informatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Lisa M James
- Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Center for Cognitive Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Margaret Y Mahan
- Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; Graduate Program in Biomedical Informatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jasmine Joseph
- Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; Graduate Program in Biomedical Informatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Angeliki Georgopoulos
- Metabolic Service, Department of Medicine, Department of Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Brian E Engdahl
- Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN 55417, USA; Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Center for Cognitive Sciences, University of Minnesota, Minneapolis, MN 55455, USA; Department of Psychology, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
21
|
Tsang JS. Utilizing population variation, vaccination, and systems biology to study human immunology. Trends Immunol 2015; 36:479-93. [PMID: 26187853 PMCID: PMC4979540 DOI: 10.1016/j.it.2015.06.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/19/2015] [Accepted: 06/19/2015] [Indexed: 12/27/2022]
Abstract
The move toward precision medicine has highlighted the importance of understanding biological variability within and across individuals in the human population. In particular, given the prevalent involvement of the immune system in diverse pathologies, an important question is how much and what information about the state of the immune system is required to enable accurate prediction of future health and response to medical interventions. Towards addressing this question, recent studies using vaccination as a model perturbation and systems-biology approaches are beginning to provide a glimpse of how natural population variation together with multiplexed, high-throughput measurement and computational analysis can be used to uncover predictors of immune response quality in humans. Here I discuss recent developments in this emerging field, with emphasis on baseline correlates of vaccination responses, sources of immune-state variability, as well as relevant features of study design, data generation, and computational analysis.
Collapse
Affiliation(s)
- John S Tsang
- Systems Genomics and Bioinformatics Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD 20892, USA; Trans-NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
22
|
Pellegrino P, Falvella FS, Cheli S, Perrotta C, Clementi E, Radice S. The role of Toll-like receptor 4 polymorphisms in vaccine immune response. THE PHARMACOGENOMICS JOURNAL 2015; 16:96-101. [PMID: 25823688 DOI: 10.1038/tpj.2015.21] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/22/2014] [Accepted: 01/28/2015] [Indexed: 12/22/2022]
Abstract
Toll-like receptors (TLRs) are a class of pattern recognition receptors that are deputed to recognise a range of molecular structures in pathogens. One of the most studied members of this family is the TLR4, which is essential for the signalling of lipopolysaccharide. The gene encoding for TLR4 is highly polymorphic and this genetic variability may explain in part the interindividual variability observed in several clinical setting, including the response to vaccination. Herein, we review and systematise the available scientific evidence about the effect of TLR4 polymorphisms on vaccine response, including approved prophylactic, new therapeutic cancer vaccines and recently approved vaccine adjuvants. Data reviewed in this analysis indicate that TLR4 polymorphisms significantly affect vaccine response. If these results are confirmed by further analyses, the use of these genetic biomarkers may become a useful tool to tailor vaccination in specific subsets of patients.
Collapse
Affiliation(s)
- P Pellegrino
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - F S Falvella
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - S Cheli
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - C Perrotta
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - E Clementi
- Scientific Institute, IRCCS E. Medea, Lecco, Italy.,Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, Consiglio Nazionale delle Ricerche Institute of Neuroscience, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - S Radice
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| |
Collapse
|
23
|
Newport MJ. The genetic regulation of infant immune responses to vaccination. Front Immunol 2015; 6:18. [PMID: 25699041 PMCID: PMC4313718 DOI: 10.3389/fimmu.2015.00018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 01/09/2015] [Indexed: 12/16/2022] Open
Abstract
A number of factors are recognized to influence immune responses to vaccinations including age, gender, the dose, and quality of the antigen used, the number of doses given, the route of administration, and the nutritional status of the recipient. Additionally, several immunogenetic studies have identified associations between polymorphisms in genes encoding immune response proteins, both innate and adaptive, and variation in responses to vaccines. Variants in the genes encoding Toll-like receptors, HLA molecules, cytokines, and cytokine receptors have associated with heterogeneity of responses to a wide range of vaccines including measles, hepatitis B, influenza A, BCG, Haemophilus influenzae type b, and certain Neisseria meningitidis serotypes, amongst others. However, the vast majority of these studies have been conducted in older children and adults and there are very few data available from studies conducted in infants. This paper reviews the evidence to date that host genes influencing vaccines responses in these older population and identifies a large gap in our understanding of the genetic regulation of responses in early life. Given the high mortality from infection in early life and the challenges of developing vaccines that generate effective immune responses in the context of the developing immune system further research on infant populations is required.
Collapse
Affiliation(s)
- Melanie J. Newport
- Division of Clinical Medicine, Brighton and Sussex Medical School, Brighton, UK
| |
Collapse
|
24
|
Barbosa T, Barral-Netto M. Challenges in the research and development of new human vaccines. Braz J Med Biol Res 2015; 46:103-8. [PMID: 23558931 PMCID: PMC3854358 DOI: 10.1590/1414-431x20131873] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 01/16/2013] [Indexed: 12/22/2022] Open
Abstract
The field of vaccinology was born from the observations by the fathers of vaccination, Edward Jenner and Louis Pasteur, that a permanent, positive change in the way our bodies respond to life-threatening infectious diseases can be obtained by specific challenge with the inactivated infectious agent performed in a controlled manner, avoiding the development of clinical disease upon exposure to the virulent pathogen. Many of the vaccines still in use today were developed on an empirical basis, essentially following the paradigm established by Pasteur, "isolate, inactivate, and inject" the disease-causing microorganism, and are capable of eliciting uniform, long-term immune memory responses that constitute the key to their proven efficacy. However, vaccines for pathogens considered as priority targets of public health concern are still lacking. The literature tends to focus more often on vaccine research problems associated with specific pathogens, but it is increasingly clear that there are common bottlenecks in vaccine research, which need to be solved in order to advance the development of the field as a whole. As part of a group of articles, the objective of the present report is to pinpoint these bottlenecks, exploring the literature for common problems and solutions in vaccine research applied to different situations. Our goal is to stimulate brainstorming among specialists of different fields related to vaccine research and development. Here, we briefly summarize the topics we intend to deal with in this discussion.
Collapse
Affiliation(s)
- T Barbosa
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz, Salvador, BA, Brasil.
| | | |
Collapse
|
25
|
Ovsyannikova IG, Salk HM, Larrabee BR, Pankratz VS, Poland GA. Single-nucleotide polymorphism associations in common with immune responses to measles and rubella vaccines. Immunogenetics 2014; 66:663-9. [PMID: 25139337 DOI: 10.1007/s00251-014-0796-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 08/11/2014] [Indexed: 02/08/2023]
Abstract
Single-nucleotide polymorphisms (SNPs) in candidate immune response genes were evaluated for associations with measles- and rubella-specific neutralizing antibodies, interferon (IFN)-γ, and interleukin (IL)-6 secretion in two separate association analyses in a cohort of healthy immunized subjects. We identified six SNP associations shared between the measles-specific and rubella-specific immune responses, specifically neutralizing antibody titers (DDX58), secreted IL-6 (IL10RB, IL12B), and secreted IFN-γ (IFNAR2, TLR4). An intronic SNP (rs669260) in the antiviral innate immune receptor gene, DDX58, was significantly associated with increased neutralizing antibody titers for both measles and rubella viral antigens post-MMR vaccination (p values 0.02 and 0.0002, respectively). Significant associations were also found between IL10RB (rs2284552; measles study p value 0.006, rubella study p value 0.00008) and IL12B (rs2546893; measles study p value 0.005, rubella study p value 0.03) gene polymorphisms and variations in both measles- and rubella virus-specific IL-6 responses. We also identified associations between individual SNPs in the IFNAR2 and TLR4 genes that were associated with IFN-γ secretion for both measles and rubella vaccine-specific immune responses. These results are the first to indicate that there are SNP associations in common across measles and rubella vaccine immune responses and that SNPs from multiple genes involved in innate and adaptive immune response regulation may contribute to the overall human antiviral response.
Collapse
Affiliation(s)
- Inna G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim 611C, 200 1st Street S.W., Rochester, MN, 55905, USA
| | | | | | | | | |
Collapse
|
26
|
Posteraro B, Pastorino R, Di Giannantonio P, Ianuale C, Amore R, Ricciardi W, Boccia S. The link between genetic variation and variability in vaccine responses: Systematic review and meta-analyses. Vaccine 2014; 32:1661-9. [DOI: 10.1016/j.vaccine.2014.01.057] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 12/23/2013] [Accepted: 01/24/2014] [Indexed: 01/11/2023]
|
27
|
Kashiwagi Y, Miyata A, Kumagai T, Maehara K, Suzuki E, Nagai T, Ozaki T, Nishimura N, Okada K, Kawashima H, Nakayama T. Production of inflammatory cytokines in response to diphtheria-pertussis-tetanus (DPT), haemophilus influenzae type b (Hib), and 7-valent pneumococcal (PCV7) vaccines. Hum Vaccin Immunother 2013; 10:677-85. [PMID: 24589970 PMCID: PMC4130255 DOI: 10.4161/hv.27264] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Haemophilus influenzae type b (Hib) and 7-valent pneumococcal (PCV7) vaccines both became recommended in Japan in 2010. In this study, cytokine production was investigated in peripheral blood mononuclear cells (PBMCs) cultures stimulated with diphtheria and tetanus toxoids combined with acellular pertussis vaccine (DPT), Hib, and PCV7 separately or concurrent different combinations, all as final off-the-shelf vaccines without the individual vaccine components as controls. Higher IL-1β levels were produced when cultures were stimulated with PCV than with DPT or Hib, and the concurrent stimulation including PCV7 enhanced the production of IL-1β. Although Hib induced higher levels of IL-6, no significant difference was observed in IL-6 production with the concurrent stimulation. The concurrent stimulation with Hib/PCV7 and DPT/Hib/PCV7 produced higher levels of TNF-α and human G-CSF. Cytokine profiles were examined in serum samples obtained from 61 vaccine recipients with febrile reactions and 18 recipients without febrile illness within 24 h of vaccination. No significant difference was observed in cytokine levels of IL-1β, IL-4, IL-6, IL-10, IL-12, IFN-γ, MIP-1, TNF-α, and prostaglandin E2 (PGE2) in sera between the two groups. However, significantly higher levels of human G-CSF were observed in recipients with febrile illness than in those without febrile reactions. Further investigations of the significance of elevated serum G-CSF levels are required in vaccine recipients with febrile illness.
Collapse
Affiliation(s)
- Yasuyo Kashiwagi
- Laboratory of Viral Infection I; Kitasato Institute for Life Sciences; Tokyo, Japan; Department of Pediatrics; Tokyo Medical University; Tokyo, Japan
| | - Akiko Miyata
- Miyata Pediatric Clinic; Tachikawa; Tokyo, Japan
| | | | | | - Eitarou Suzuki
- Suzuki Pediatric Clinic; Ube, Yamaguchi Prefecture, Japan
| | - Takao Nagai
- Nagai Pediatric Clinic; Takamatsu, Kagawa Prefecture, Japan
| | - Takao Ozaki
- Department of Pediatrics; Konan Kosei Hospital; Konan; Aichi Prefecture, Japan
| | - Naoko Nishimura
- Department of Pediatrics; Konan Kosei Hospital; Konan; Aichi Prefecture, Japan
| | - Kenji Okada
- Department of Pediatrics; National Fukuoka Hospital; Fukuoka, Japan
| | | | - Tetsuo Nakayama
- Laboratory of Viral Infection I; Kitasato Institute for Life Sciences; Tokyo, Japan
| |
Collapse
|
28
|
Smolen KK, Ruck CE, Fortuno ES, Ho K, Dimitriu P, Mohn WW, Speert DP, Cooper PJ, Esser M, Goetghebuer T, Marchant A, Kollmann TR. Pattern recognition receptor-mediated cytokine response in infants across 4 continents. J Allergy Clin Immunol 2013; 133:818-26.e4. [PMID: 24290283 PMCID: PMC3969582 DOI: 10.1016/j.jaci.2013.09.038] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 09/16/2013] [Accepted: 09/26/2013] [Indexed: 12/14/2022]
Abstract
Background Susceptibility to infection as well as response to vaccination varies among populations. To date, the underlying mechanisms responsible for these clinical observations have not been fully delineated. Because innate immunity instructs adaptive immunity, we hypothesized that differences between populations in innate immune responses may represent a mechanistic link to variation in susceptibility to infection or response to vaccination. Objective Determine whether differences in innate immune responses exist among infants from different continents of the world. Methods We determined the innate cytokine response following pattern recognition receptor (PRR) stimulation of whole blood from 2-year-old infants across 4 continents (Africa, North America, South America, and Europe). Results We found that despite the many possible genetic and environmental exposure differences in infants across 4 continents, innate cytokine responses were similar for infants from North America, South America, and Europe. However, cells from South African infants secreted significantly lower levels of cytokines than did cells from infants from the 3 other sites, and did so following stimulation of extracellular and endosomal but not cytosolic PRRs. Conclusions Substantial differences in innate cytokine responses to PRR stimulation exist among different populations of infants that could not have been predicted. Delineating the underlying mechanism(s) for these differences will not only aid in improving vaccine-mediated protection but possibly also provide clues for the susceptibility to infection in different regions of the world.
Collapse
Affiliation(s)
- Kinga K Smolen
- Department of Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Candice E Ruck
- Department of Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Edgardo S Fortuno
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevin Ho
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pedro Dimitriu
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - William W Mohn
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - David P Speert
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Philip J Cooper
- Centro de Investigaciones FEPIS, Esmeraldas, Quininde, Ecuador; Molecular and Biochemical Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom; Centro de Investgación en Enfermedades Infecciosas, Escuela de Biología, Pontificia Universidad Católica del Ecuador, Casilla, Quito, Ecuador
| | - Monika Esser
- Immunology Unit, Division of Medical Microbiology, Department of Pathology, NHLS and Stellenbosch University, Matieland, South Africa
| | - Tessa Goetghebuer
- Department of Paediatrics, Centre Hospitalier Universitaire Saint-Pierre, Brussels, Belgium
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Tobias R Kollmann
- Department of Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
29
|
Majumder PP, Sarkar-Roy N, Staats H, Ramamurthy T, Maiti S, Chowdhury G, Whisnant CC, Narayanasamy K, Wagener DK. Genomic correlates of variability in immune response to an oral cholera vaccine. Eur J Hum Genet 2013; 21:1000-6. [PMID: 23249958 PMCID: PMC3746254 DOI: 10.1038/ejhg.2012.278] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 10/23/2012] [Accepted: 11/20/2012] [Indexed: 11/09/2022] Open
Abstract
Cholera is endemic to many countries. Recent major outbreaks of cholera have prompted World Health Organization to recommend oral cholera vaccination as a public-health strategy. Variation in percentage of seroconversion upon cholera vaccination has been recorded across populations. Vaccine-induced responses are influenced by host genetic differences. We have investigated association between single-nucleotide polymorphic (SNP) loci in and around 296 immunologically relevant genes and total anti-lipopolysaccharide (LPS) antibody response to a killed whole-cell vaccine, comprising LPS from multiple strains of Vibrio cholerae. Titers derived from standard vibriocidal assays were also analyzed to gain further insights on validated SNP associations. Vaccination was administered to 1000 individuals drawn from India. Data on two independent random subsets, each comprising ∼500 vaccinees, were used for discovery of genomic associations and validation, respectively. Significant associations of four SNPs and haplotypes in three genes (MARCO, TNFAIP3 and CXCL12) with AR were discovered and validated, of which two in TNFAIP3 and CXCL12 were also significantly associated with immunity (fourfold increase in vibriocidal titers). CXCL12 is a neutrophil and lymphocyte chemoattractant that is upregulated in response to V. cholerae infection. LPS in the vaccine possibly provides signals that mimic those of the live bacterium. TNFAIP3 promotes intestinal epithelial barrier integrity and provides tight junction protein regulation; possible requirements for adequate response to the vaccine. LPS is a potent activator of innate immune responses and a ligand of MARCO. Variants in this gene have been found to be associated with LPS response, but not with high vibriocidal titer level.
Collapse
|
30
|
Kollmann TR. Variation between Populations in the Innate Immune Response to Vaccine Adjuvants. Front Immunol 2013; 4:81. [PMID: 23565115 PMCID: PMC3613898 DOI: 10.3389/fimmu.2013.00081] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 03/18/2013] [Indexed: 12/15/2022] Open
Abstract
The success of the World Health Organization recommended “Expanded Program of Immunization” (EPI) and similar regional or national programs has been astounding. However, infectious threats currently not covered by these programs continue to infect millions of infants around the world. Furthermore, many infants do not receive existing vaccines either on time or for the required number of doses to provide optimal protection. Nor do all infants around the world develop the same protective immune response to the same vaccine. As a result approximately three million infants die every year from vaccine preventable infections. To tackle these issues, new vaccines need to be developed as well as existing ones made easier to administer. This requires identification of age-optimized vaccine schedules and formulations. In order to be most effective this approach will need to take population-based differences in response to vaccines and adjuvants into account. This review summarizes what is currently known about differences between populations around the world in the innate immune response to existing as well as new and promising vaccine adjuvants.
Collapse
Affiliation(s)
- Tobias R Kollmann
- Division of Infectious and Immunological Diseases, Department of Paediatrics, University of British Columbia Vancouver, BC, Canada
| |
Collapse
|
31
|
Gowane GR, Sharma AK, Sankar M, Narayanan K, Das B, Subramaniam S, Pattnaik B. Association of BoLA DRB3 alleles with variability in immune response among the crossbred cattle vaccinated for foot-and-mouth disease (FMD). Res Vet Sci 2013; 95:156-63. [PMID: 23541924 DOI: 10.1016/j.rvsc.2013.03.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 02/23/2013] [Accepted: 03/03/2013] [Indexed: 12/20/2022]
Abstract
Polymorphism of bovine leukocyte antigen (BoLA) DRB3 gene is being intensively investigated for potential association with economically important diseases of cattle. Accordingly, we investigated the association of DRB3 Exon 2 polymorphism as evidenced by the variation in the binding pockets with variability in immune response to inactivated trivalent (O, A and Asia1) foot and mouth disease virus (FMDV) vaccine in a closed population of crossbred cattle. Antibody titer of ≥ 1.8 was set as the cut off value to distinguish the protected (≥ 1.8) and unprotected (<1.8) animals. Eleven different alleles of over 3% frequency were detected in the population. We found that DRB3 alleles 0201, 0801 and 1501 always ranked high for protective immune response whereas alleles 0701, 1103 and 1101 consistently ranked low for unprotected immune response for all the three serotypes. Rank correlation of DRB3 alleles among the three serotypes was positive, high in magnitude and statistically significant (P<0.05). Logistic regression analysis revealed that odds of protection from the vaccine were highest for all the three serotypes if allele (∗)1501 was present and strengthened the results of allele ranking. Predicted amino acid substitution in the peptide binding pockets revealed that all the important sites had high Wu-Kabat index. Similarly, specific residues in pockets were crucial for immune response to FMD vaccine. There were specific substitutions in un-protected alleles such as absence of acidic amino acids substituted by basic amino acid at β71, presence of non-polar cysteine or basic histidine at β30 and presence of polar tyrosine at β37. From the observations, we hypothesize that the substitutions lead to unique conformational changes in the protein products of the studied alleles that would associate with the protective or unprotective antibody response to FMDV vaccine. The knowledge has potential implications in future selection programs if integrated with the complete BoLA haplotype details and production traits of the herd.
Collapse
Affiliation(s)
- G R Gowane
- Central Sheep & Wool Research Institute, Avikanagar via Jaipur, Rajasthan 304 501, India.
| | | | | | | | | | | | | |
Collapse
|
32
|
Ovsyannikova IG, Haralambieva IH, Vierkant RA, O'Byrne MM, Poland GA. Associations between polymorphisms in the antiviral TRIM genes and measles vaccine immunity. Hum Immunol 2013; 74:768-74. [PMID: 23416095 DOI: 10.1016/j.humimm.2013.01.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 01/10/2013] [Accepted: 01/31/2013] [Indexed: 10/27/2022]
Abstract
The role of polymorphisms within the antiviral tripartite motif (TRIM) genes in measles vaccine adaptive immune responses was examined. A limited association was found between TRIM5 (rs7122620) and TRIM25 (rs205499) gene polymorphisms and measles-specific antibody levels. However, many associations were found between TRIM gene SNPs and variations in cellular responses (IFN-γ Elispot and secreted cytokines IL-2, IL-6, IL-10, IFN-γ, and TNF-α). TRIM22 rs2291841 was significantly associated with an increased IFN-γ Elispot response (35 vs. 102 SFC per 2×10(5)PBMC, p=0.009, q=0.71) in Caucasians. A non-synonymous TRIM25 rs205498 (in LD with other SNPs, r(2)≥0.56), as well as the TRIM25 AAAGGAAAGGAGT haplotype, was associated with a decreased IFN-γ Elispot response (t-statistic -2.32, p=0.02) in African-Americans. We also identified polymorphisms in the TRIM5, TRIM22, and TRIM25 genes that were associated with significant differences in cytokine responses. Additional studies are necessary to replicate our findings and to examine the functional consequences of these associations.
Collapse
Affiliation(s)
- Inna G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, United States
| | | | | | | | | |
Collapse
|
33
|
Glass EJ, Baxter R, Leach RJ, Jann OC. Genes controlling vaccine responses and disease resistance to respiratory viral pathogens in cattle. Vet Immunol Immunopathol 2012; 148:90-9. [PMID: 21621277 PMCID: PMC3413884 DOI: 10.1016/j.vetimm.2011.05.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 04/21/2011] [Accepted: 05/02/2011] [Indexed: 12/27/2022]
Abstract
Farm animals remain at risk of endemic, exotic and newly emerging viruses. Vaccination is often promoted as the best possible solution, and yet for many pathogens, either there are no appropriate vaccines or those that are available are far from ideal. A complementary approach to disease control may be to identify genes and chromosomal regions that underlie genetic variation in disease resistance and response to vaccination. However, identification of the causal polymorphisms is not straightforward as it generally requires large numbers of animals with linked phenotypes and genotypes. Investigation of genes underlying complex traits such as resistance or response to viral pathogens requires several genetic approaches including candidate genes deduced from knowledge about the cellular pathways leading to protection or pathology, or unbiased whole genome scans using markers spread across the genome. Evidence for host genetic variation exists for a number of viral diseases in cattle including bovine respiratory disease and anecdotally, foot and mouth disease virus (FMDV). We immunised and vaccinated a cattle cross herd with a 40-mer peptide derived from FMDV and a vaccine against bovine respiratory syncytial virus (BRSV). Genetic variation has been quantified. A candidate gene approach has grouped high and low antibody and T cell responders by common motifs in the peptide binding pockets of the bovine major histocompatibility complex (BoLA) DRB3 gene. This suggests that vaccines with a minimal number of epitopes that are recognised by most cattle could be designed. Whole genome scans using microsatellite and single nucleotide polymorphism (SNP) markers has revealed many novel quantitative trait loci (QTL) and SNP markers controlling both humoral and cell-mediated immunity, some of which are in genes of known immunological relevance including the toll-like receptors (TLRs). The sequencing, assembly and annotation of livestock genomes and is continuing apace. In addition, provision of high-density SNP chips should make it possible to link phenotypes with genotypes in field populations without the need for structured populations or pedigree information. This will hopefully enable fine mapping of QTL and ultimate identification of the causal gene(s). The research could lead to selection of animals that are more resistant to disease and new ways to improve vaccine efficacy.
Collapse
Affiliation(s)
- Elizabeth J Glass
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK.
| | | | | | | |
Collapse
|
34
|
Heijstek MW, van Gageldonk PGM, Berbers GAM, Wulffraat NM. Differences in persistence of measles, mumps, rubella, diphtheria and tetanus antibodies between children with rheumatic disease and healthy controls: a retrospective cross-sectional study. Ann Rheum Dis 2012; 71:948-54. [PMID: 22172491 DOI: 10.1136/annrheumdis-2011-200637] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES To compare the persistence of measles, mumps, rubella, diphtheria and tetanus antibodies between patients with juvenile idiopathic arthritis (JIA) and healthy controls. METHODS Measles, mumps, rubella (MMR) and diphtheria-tetanus toxoid (DT)-specific immunoglobulin G antibody concentrations were compared between 400 patients with JIA and 2176 healthy controls aged 1-19 years. Stored patient samples from the period 1997-2006 were obtained from one Dutch centre for paediatric rheumatology. Healthy control samples had been evaluated previously in a nationwide cohort. Participants had been vaccinated according to the Dutch immunisation programme. Antibody concentrations were measured by ELISA (MMR) or multiplex immunoassay (DT). RESULTS Corrected for age and the number of vaccinations, lower vaccine-specific geometric mean antibody concentrations (GMC) were found in patients with JIA against mumps, rubella, diphtheria and tetanus (p≤0.001). Measles-specific GMC were higher (p<0.001) compared with healthy controls. The prevalence of protective antibody concentrations was significantly lower in patients for mumps (OR 0.4; 95% CI 0.3 to 0.6), rubella (OR 0.4; 0.3 to 0.7), diphtheria (OR 0.1; 0.06 to 0.2) and tetanus (OR 0.1; 0.05 to 0.3). Seroprotection rates against measles did not differ between patients and healthy controls (OR 1.4; 0.8 to 2.5). Methotrexate and glucocorticosteroid use did not affect pathogen-specific GMC or seroprotection rates. CONCLUSIONS Patients with JIA had lower antibody concentrations and seroprotection rates than healthy controls against mumps, rubella, diphtheria and tetanus, but not measles. In these patients, regular assessment of antibody concentrations and further research on responses to other (booster) vaccines are warranted.
Collapse
Affiliation(s)
- Marloes W Heijstek
- Department of Paediatric Immunology, Wilhelmina Children’s Hospital, University Medical Centre, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
35
|
Pajewski NM, Shrestha S, Quinn CP, Parker SD, Wiener H, Aissani B, McKinney BA, Poland GA, Edberg JC, Kimberly RP, Tang J, Kaslow RA. A genome-wide association study of host genetic determinants of the antibody response to Anthrax Vaccine Adsorbed. Vaccine 2012; 30:4778-84. [PMID: 22658931 DOI: 10.1016/j.vaccine.2012.05.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 04/20/2012] [Accepted: 05/14/2012] [Indexed: 11/16/2022]
Abstract
Several lines of evidence have supported a host genetic contribution to vaccine response, but genome-wide assessments for specific determinants have been sparse. Here we describe a genome-wide association study (GWAS) of protective antigen-specific antibody (AbPA) responses among 726 European-Americans who received Anthrax Vaccine Adsorbed (AVA) as part of a clinical trial. After quality control, 736,996 SNPs were tested for association with the AbPA response to 3 or 4 AVA vaccinations given over a 6-month period. No SNP achieved the threshold of genome-wide significance (p=5 × 10(-8)), but suggestive associations (p<1 × 10(-5)) were observed for SNPs in or near the class II region of the major histocompatibility complex (MHC), in the promoter region of SPSB1, and adjacent to MEX3C. Multivariable regression modeling suggested that much of the association signal within the MHC corresponded to previously identified HLA DR-DQ haplotypes involving component HLA-DRB1 alleles of *15:01, *01:01, or *01:02. We estimated the proportion of additive genetic variance explained by common SNP variation for the AbPA response after the 6 month vaccination. This analysis indicated a significant, albeit imprecisely estimated, contribution of variation tagged by common polymorphisms (p=0.032). Future studies will be required to replicate these findings in European Americans and to further elucidate the host genetic factors underlying variable immune response to AVA.
Collapse
Affiliation(s)
- Nicholas M Pajewski
- Department of Biostatistical Sciences, Wake Forest University Health Sciences, Winston Salem, NC 27157-1063, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Tomljenovic L, Shaw CA. “One-size fits all”? Vaccine 2012; 30:2040. [DOI: 10.1016/j.vaccine.2011.11.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Revised: 10/26/2011] [Accepted: 11/03/2011] [Indexed: 11/29/2022]
|
37
|
Gröndahl-Yli-Hannuksela K, Vuononvirta J, Barkoff AM, Viander M, Van Der Meeren O, Mertsola J, He Q. Gene polymorphism in toll-like receptor 4: effect on antibody production and persistence after acellular pertussis vaccination during adolescence. J Infect Dis 2012; 205:1214-9. [PMID: 22383676 DOI: 10.1093/infdis/jis182] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Toll-like receptors play an important role in the regulation of adaptive immunity. This study aimed to investigate whether Toll-like receptor 4 (TLR4) polymorphisms influence the production and persistence of antibodies after acellular pertussis booster vaccination during adolescence. METHODS Seventy-five subjects received a single dose of diphtheria and tetanus toxoids and acellular pertussis vaccine 10 years ago, during adolescence. The same cohort was followed up at 3, 5, and 10 years after this booster vaccination. Pyrosequencing was used for detecting polymorphism in TLR4. Concentrations of anti-pertussis vaccine antibodies were measured by standardized enzyme-linked immunosorbant assay and published elsewhere. RESULTS The fold increase in antibodies to pertussis toxin after original vaccination 10 years ago was significantly lower in subjects with TLR4 polymorphism than in those without (55% vs 86%; P = .028). At the 3-year follow-up evaluation, geometric mean concentrations of anti-pertussis vaccine antibodies were significantly lower in subjects with TLR4 polymorphism, compared with those without the polymorphism (for pertussis toxin, P = .028; for filamentous hemagglutinin, P = .047; and for pertactin, P = .046). CONCLUSIONS This study suggests that TLR4 Asp299Gly polymorphism might influence production and persistence of antibodies after pertussis booster vaccination in adolescents. However, the results should be interpreted with caution as the number of subjects included in this study was limited.
Collapse
Affiliation(s)
- Kirsi Gröndahl-Yli-Hannuksela
- Department of Infectious Disease Surveillance and Control, National Institute for Health and Welfare (THL), Turku, Finland
| | | | | | | | | | | | | |
Collapse
|
38
|
Consistency of HLA associations between two independent measles vaccine cohorts: a replication study. Vaccine 2012; 30:2146-52. [PMID: 22285888 DOI: 10.1016/j.vaccine.2012.01.038] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 01/04/2012] [Accepted: 01/12/2012] [Indexed: 02/05/2023]
Abstract
Associations between HLA genotypes and measles vaccine humoral and cellular immune responses were examined to better understand immunogenetic drivers of vaccine response. Two independent study cohorts of healthy schoolchildren were examined: cohort one, 346 children between 12 and 18 years of age; and cohort two, 388 children between 11 and 19 years of age. All received two age-appropriate doses of measles-containing vaccine. The purpose of this study was to identify and replicate associations between HLA genes and immune responses following measles vaccination found in our first cohort. Associations of comparable magnitudes and with similar p-values were observed between B*3503 (1st cohort p=0.01; 2nd cohort p=0.07), DQA1*0201 (1st cohort p=0.03; 2nd cohort p=0.03), DQB1*0303 (1st cohort p=0.10; 2 cohort p=0.02), DQB1*0602 (1st cohort p=0.07; 2nd cohort p=0.10), and DRB1*0701 (1st cohort p=0.03; 2nd cohort p=0.07) alleles and measles-specific antibody levels. Suggestive, yet consistent, associations were observed between the B7 (1st cohort p=0.01; 2nd cohort p=0.08) supertype and higher measles antibody levels in both cohorts. Also, in both cohorts, the B*0801 and DRB1*0301 alleles, C*0802 and DPA1*0202 alleles, and DRB1*1303 alleles displayed consistent associations with variations in IFN-γ, IL-2 and IL-10 secretion, respectively. This study emphasizes the importance of replicating HLA associations with measles vaccine-induced humoral and cellular immune responses and increases confidence in the results. These data will inform strategies for functional studies and novel vaccine development, including epitope-based measles vaccines. This is the first HLA association replication study with measles vaccine-specific immune responses to date.
Collapse
|
39
|
Kennedy RB, Ovsyannikova IG, Haralambieva IH, O'Byrne MM, Jacobson RM, Pankratz VS, Poland GA. Multigenic control of measles vaccine immunity mediated by polymorphisms in measles receptor, innate pathway, and cytokine genes. Vaccine 2012; 30:2159-67. [PMID: 22265947 DOI: 10.1016/j.vaccine.2012.01.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 11/30/2011] [Accepted: 01/08/2012] [Indexed: 11/28/2022]
Abstract
Measles infection and vaccine response are complex biological processes that involve both viral and host genetic factors. We have previously investigated the influence of genetic polymorphisms on vaccine immune response, including measles vaccines, and have shown that polymorphisms in HLA, cytokine, cytokine receptor, and innate immune response genes are associated with variation in vaccine response but do not account for all of the inter-individual variance seen in vaccinated populations. In the current study we report the findings of a multigenic analysis of measles vaccine immunity, indicating a role for the measles virus receptor CD46, innate pattern-recognition receptors (DDX58, TLR2, 4, 5, 7 and 8) and intracellular signaling intermediates (MAP3K7, NFKBIA), and key antiviral molecules (VISA, OAS2, MX1, PKR) as well as cytokines (IFNA1, IL4, IL6, IL8, IL12B) and cytokine receptor genes (IL2RB, IL6R, IL8RA) in the genetic control of both humoral and cellular immune responses. This multivariate approach provided additional insights into the genetic control of measles vaccine responses over and above the information gained by our previous univariate SNP association analyses.
Collapse
|
40
|
Poland GA, Kennedy RB, Ovsyannikova IG. Vaccinomics and personalized vaccinology: is science leading us toward a new path of directed vaccine development and discovery? PLoS Pathog 2011; 7:e1002344. [PMID: 22241978 PMCID: PMC3248557 DOI: 10.1371/journal.ppat.1002344] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
As is apparent in many fields of science and medicine, the new biology, and particularly new high-throughput genetic sequencing and transcriptomic and epigenetic technologies, are radically altering our understanding and views of science. In this article, we make the case that while mostly ignored thus far in the vaccine field, these changes will revolutionize vaccinology from development to manufacture to administration. Such advances will address a current major barrier in vaccinology-that of empiric vaccine discovery and development, and the subsequent low yield of viable vaccine candidates, particularly for hyper-variable viruses. While our laboratory's data and thinking (and hence also for this paper) has been directed toward viruses and viral vaccines, generalization to other pathogens and disease entities (i.e., anti-cancer vaccines) may be appropriate.
Collapse
Affiliation(s)
- Gregory A Poland
- Mayo Vaccine Research Group, Department of Medicine, Mayo Clinic College of Medicine, Mayo Foundation, Rochester, Minnesota, United States of America.
| | | | | |
Collapse
|
41
|
Lemaire D, Barbosa T, Rihet P. Coping with genetic diversity: the contribution of pathogen and human genomics to modern vaccinology. Braz J Med Biol Res 2011; 45:376-85. [PMID: 22030866 PMCID: PMC3854287 DOI: 10.1590/s0100-879x2011007500142] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 10/04/2011] [Indexed: 11/21/2022] Open
Abstract
Vaccine development faces major difficulties partly because of genetic variation in both infectious organisms and humans. This causes antigenic variation in infectious agents and a high interindividual variability in the human response to the vaccine. The exponential growth of genome sequence information has induced a shift from conventional culture-based to genome-based vaccinology, and allows the tackling of challenges in vaccine development due to pathogen genetic variability. Additionally, recent advances in immunogenetics and genomics should help in the understanding of the influence of genetic factors on the interindividual and interpopulation variations in immune responses to vaccines, and could be useful for developing new vaccine strategies. Accumulating results provide evidence for the existence of a number of genes involved in protective immune responses that are induced either by natural infections or vaccines. Variation in immune responses could be viewed as the result of a perturbation of gene networks; this should help in understanding how a particular polymorphism or a combination thereof could affect protective immune responses. Here we will present: i) the first genome-based vaccines that served as proof of concept, and that provided new critical insights into vaccine development strategies; ii) an overview of genetic predisposition in infectious diseases and genetic control in responses to vaccines; iii) population genetic differences that are a rationale behind group-targeted vaccines; iv) an outlook for genetic control in infectious diseases, with special emphasis on the concept of molecular networks that will provide a structure to the huge amount of genomic data.
Collapse
Affiliation(s)
- D Lemaire
- Universidade Federal da Bahia, Salvador, BA, Brasil
| | | | | |
Collapse
|
42
|
Ovsyannikova IG, Haralambieva IH, Vierkant RA, Pankratz VS, Jacobson RM, Poland GA. The role of polymorphisms in Toll-like receptors and their associated intracellular signaling genes in measles vaccine immunity. Hum Genet 2011; 130:547-61. [PMID: 21424379 PMCID: PMC3924423 DOI: 10.1007/s00439-011-0977-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 03/08/2011] [Indexed: 12/15/2022]
Abstract
Toll-like receptors (TLRs) and their intracellular signaling molecules play an important role in innate immunity. In this study, we examined associations between polymorphisms in TLR family genes and measles vaccine-specific immune responses. We genotyped 764 subjects (11-22 years old) after two doses of measles vaccine for TLR signaling SNP markers (n = 454). The major alleles of coding SNPs in the TLR2 (rs3804100) and TLR4 (rs5030710) genes were associated with a dose-related increase (660 vs. 892 mIU/ml, p = 0.002) and a dose-related decrease (2,209 vs. 830 mIU/ml, p = 0.001) in measles-specific antibodies, respectively. A significant association was found between lower measles antibody levels and the haplotype ACGGCGAGAAAAGAGAAGAGAGAGAA (p = 0.01) in the MAP3K7 gene. Furthermore, the minor allele of a SNP (rs702966) of the KIAA1542 (IRF7) gene was associated with a dose-related decrease in IFN-γ Elispot responses (38 vs. 26 spot-forming cells per 2 × 10(5) PBMCs, p = 0.00002). We observed an additional 12 associations (p < 0.01) between coding (nonsynonymous and synonymous) polymorphisms within the TLRs (TLR2, 7, and 8), IKBKE, TICAM1, NFKBIA, IRAK2, and KIAA1542 genes and variations in measles-specific IL-2, IL-6, IFN-α, IFN-γ, IFNλ-1, and TNF-α secretion levels. Our data demonstrate that polymorphisms in TLR and other related immune response signaling molecules have significant effects on measles vaccine-associated immune responses. These data help to establish the genetic foundation for immune response variation in response to measles immunization and provide important insights for the rational development of new measles vaccines.
Collapse
Affiliation(s)
- Inna G. Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905 USA
- Program in Translational Immunovirology and Biodefense, Mayo Clinic, Rochester, MN 55905 USA
| | | | - Robert A. Vierkant
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905 USA
| | - V. Shane Pankratz
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905 USA
| | - Robert M. Jacobson
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905 USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905 USA
- Program in Translational Immunovirology and Biodefense, Mayo Clinic, Rochester, MN 55905 USA
| | - Gregory A. Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905 USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905 USA
- Program in Translational Immunovirology and Biodefense, Mayo Clinic, Rochester, MN 55905 USA
| |
Collapse
|
43
|
Ovsyannikova IG, Poland GA. Vaccinomics: current findings, challenges and novel approaches for vaccine development. AAPS J 2011; 13:438-44. [PMID: 21671143 PMCID: PMC3160164 DOI: 10.1208/s12248-011-9281-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 05/05/2011] [Indexed: 02/06/2023] Open
Abstract
Recent years have witnessed a growing interest in a field of vaccinology that we have named vaccinomics. The overall idea behind vaccinomics is to identify genetic and other mechanisms and pathways that determine immune responses, and thereby provide new candidate vaccine approaches. Considerable data show that host genetic polymorphisms act as important determinants of innate and adaptive immunity to vaccines. This review highlights examples of the role of immunogenetics and immunogenomics in understanding immune responses to vaccination, which are highly variable across the population. The influence of HLA genes, non-HLA, and innate genes in inter-individual variations in immune responses to viral vaccines are examined using population-based gene/SNP association studies. The ability to understand relationships between immune response gene variants and vaccine-specific immunity may assist in designing new vaccines. At the same time, application of state-of-the-art next-generation sequencing technology (and bioinformatics) is desired to provide new genetic information and its relationship to the immune response.
Collapse
Affiliation(s)
- Inna G. Ovsyannikova
- />Vaccine Research Group, Mayo Clinic, Rochester, Minnesota USA
- />Program in Translational Immunovirology and Biodefense, Rochester, Minnesota USA
- />Department of Medicine, Mayo Clinic, Rochester, Minnesota USA
| | - Gregory A. Poland
- />Vaccine Research Group, Mayo Clinic, Rochester, Minnesota USA
- />Program in Translational Immunovirology and Biodefense, Rochester, Minnesota USA
- />Department of Medicine, Mayo Clinic, Rochester, Minnesota USA
- />Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota USA
- />Mayo Clinic, 611C Guggenheim Building, 200 First Street, SW, Rochester, Minnesota 55905 USA
| |
Collapse
|
44
|
Human leukocyte antigen associations with humoral and cellular immunity following a second dose of measles-containing vaccine: persistence, dampening, and extinction of associations found after a first dose. Vaccine 2011; 29:7982-91. [PMID: 21872631 DOI: 10.1016/j.vaccine.2011.08.060] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 08/11/2011] [Accepted: 08/13/2011] [Indexed: 12/16/2022]
Abstract
Previously we found human leukocyte antigen (HLA) associations with humoral immunity following a single dose of measles-containing vaccine. In this study, we sought to determine if HLA associations exist with humoral and cellular immunity following a second dose of measles-containing vaccine and if the associations we found with humoral immunity after the first dose persist following a second dose. We recruited a population-based sample of 346 schoolchildren, all who previously received two doses of a measles-containing vaccine. Molecular HLA classes I and II typing as well as humoral and cellular immune assays (measles-specific IgG antibody levels and lymphoproliferative response) were performed in these subjects. We found significant associations with class I HLA-B (p=0.05) as well as class II HLA-DPB1 (p=0.01) and -DPA1 (p=0.03) genes for measles vaccine-induced antibody levels after the second dose. Similarly, we found significant associations with class II HLA-DQB1 (p=0.05) and -DRB1 (p=0.01) genes for measles-specific lymphoproliferation after the second dose. While we found HLA associations after the second dose that we previously found after the first dose of measles containing vaccine, fewer alleles had statistically significant associations, suggesting that the second dose had a dampening or extinguishing effect on the HLA associations. It appears that the second dose overcomes HLA restriction through an as yet unknown mechanism. Future studies of HLA associations should consider both the effect of dose and the role that subsequent doses might play on genetic associations found with the response to a first dose.
Collapse
|
45
|
Pajewski NM, Parker SD, Poland GA, Ovsyannikova IG, Song W, Zhang K, McKinney BA, Pankratz VS, Edberg JC, Kimberly RP, Jacobson RM, Tang J, Kaslow RA. The role of HLA-DR-DQ haplotypes in variable antibody responses to anthrax vaccine adsorbed. Genes Immun 2011; 12:457-65. [PMID: 21368772 DOI: 10.1038/gene.2011.15] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Host genetic variation, particularly within the human leukocyte antigen (HLA) loci, reportedly mediates heterogeneity in immune response to certain vaccines; however, no large study of genetic determinants of anthrax vaccine response has been described. We searched for associations between the immunoglobulin G antibody to protective antigen (AbPA) response to Anthrax Vaccine Adsorbed (AVA) in humans, and polymorphisms at HLA class I (HLA-A, -B, and -C) and class II (HLA-DRB1, -DQA1, -DQB1, -DPB1) loci. The study included 794 European-Americans and 200 African-Americans participating in a 43-month, double-blind and placebo-controlled clinical trial of AVA (clinicaltrials.gov identifier NCT00119067). Among European-Americans, genes from tightly linked HLA-DRB1, -DQA1, -DQB1 haplotypes displayed significant overall associations with longitudinal variation in AbPA levels at 4, 8, 26 and 30 weeks from baseline in response to vaccination with three or four doses of AVA (global P=6.53 × 10(-4)). In particular, carriage of the DRB1-DQA1-DQB1 haplotypes (*)1501-(*)0102-(*)0602 (P=1.17 × 10(-5)), (*)0101-(*)0101-(*)0501 (P=0.009) and (*)0102-(*)0101-(*)0501 (P=0.006) was associated with significantly lower AbPA levels. In carriers of two copies of these haplotypes, lower AbPA levels persisted following subsequent vaccinations. No significant associations were observed amongst African-Americans or for any HLA class I allele/haplotype. Further studies will be required to replicate these findings and to explore the role of host genetic variation outside of the HLA region.
Collapse
Affiliation(s)
- N M Pajewski
- Section on Statistical Genetics, University of Alabama at Birmingham, Birmingham, AL 35294-0022, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Davis NA, Crowe JE, Pajewski NM, McKinney BA. Surfing a genetic association interaction network to identify modulators of antibody response to smallpox vaccine. Genes Immun 2010; 11:630-6. [PMID: 20613780 PMCID: PMC3001955 DOI: 10.1038/gene.2010.37] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The variation in antibody response to vaccination likely involves small contributions of numerous genetic variants, such as single-nucleotide polymorphisms (SNPs), which interact in gene networks and pathways. To accumulate the bits of genetic information relevant to the phenotype that are distributed throughout the interaction network, we develop a network eigenvector centrality algorithm (SNPrank) that is sensitive to the weak main effects, gene–gene interactions and small higher-order interactions through hub effects. Analogous to Google PageRank, we interpret the algorithm as the simulation of a random SNP surfer (RSS) that accumulates bits of information in the network through a dynamic probabilistic Markov chain. The transition matrix for the RSS is based on a data-driven genetic association interaction network (GAIN), the nodes of which are SNPs weighted by the main-effect strength and edges weighted by the gene–gene interaction strength. We apply SNPrank to a GAIN analysis of a candidate-gene association study on human immune response to smallpox vaccine. SNPrank implicates a SNP in the retinoid X receptor α (RXRA) gene through a network interaction effect on antibody response. This vitamin A- and D-signaling mediator has been previously implicated in human immune responses, although it would be neglected in a standard analysis because its significance is unremarkable outside the context of its network centrality. This work suggests SNPrank to be a powerful method for identifying network effects in genetic association data and reveals a potential vitamin regulation network association with antibody response.
Collapse
Affiliation(s)
- N A Davis
- Department of Mathematical and Computer Sciences, University of Tulsa, Tulsa, OK 74104, USA
| | | | | | | |
Collapse
|
47
|
Majumder PP, Staats HF, Sarkar-Roy N, Varma B, Ghosh T, Maiti S, Narayanasamy K, Whisnant CC, Stephenson JL, Wagener DK. Genetic determinants of immune-response to a polysaccharide vaccine for typhoid. THE HUGO JOURNAL 2010; 3:17-30. [PMID: 21119757 DOI: 10.1007/s11568-010-9134-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Revised: 12/24/2009] [Accepted: 02/08/2010] [Indexed: 01/14/2023]
Abstract
UNLABELLED Differences in immunological response among vaccine recipients are determined both by their genetic differences and environmental factors. Knowledge of genetic determinants of immunological response to a vaccine can be used to design a vaccine that circumvents immunogenetic restrictions. The currently available vaccine for typhoid is a pure polysaccharide vaccine, immune response to which is T-cell independent. Little is known about whether genetic variation among vaccinees associates with variation in their antibody response to a polysaccharide vaccine. We conducted a study on 1,000 individuals resident in an area at high-risk for typhoid; vaccinated them with the typhoid vaccine, measured their antibody response to the vaccine, assayed >2,000 curated SNPs chosen from 283 genes that are known to participate in immune-response; and analyzed these data using a strategy to (a) minimize the statistical problems associated with testing of multiple hypotheses, and (b) internally cross-validate inferences, using a half-sample design, with little loss of statistical power. The first stage analysis, using the first half-sample, identified 54 SNPs in 43 genes to be significantly associated with immune response. In the second-stage, these inferences were cross-validated using the second half-sample. First-stage results of only 8 SNPs (out of 54) in 7 genes (out of 43) were cross-validated. We tested additional SNPs in these 7 genes, and found 8 more SNPs to be significantly associated. Haplotypes constructed with these SNPs in these 7 genes also showed significant association. These 7 genes are DEFB1, TLR1, IL1RL1, CTLA4, MAPK8, CD86 and IL17D. The overall picture that has emerged from this study is that (a) immune response to polysaccharide antigens is qualitatively different from that to protein antigens, and (b) polymorphisms in genes involved in polysaccharide recognition, signal transduction, inhibition of T-cell proliferation, pro-inflammatory signaling and eventual production of antimicrobial peptides are associated with antibody response to the polysaccharide vaccine for typhoid. ELECTRONIC SUPPLEMENTARY MATERIAL The online version of this article (doi:10.1007/s11568-010-9134-1) contains supplementary material, which is available to authorized users.
Collapse
|
48
|
Abstract
Vaccines are one of the most cost effective methods to control infectious diseases and at the same time one of the most complex products of the pharmaceutical industry. In contrast to other drugs, vaccines are used mainly in healthy individuals, often in children. For this reason, very high standards are set for their production. Subunit vaccines, especially peptide vaccines, can provide a safe and cost-effective alternative to vaccines produced from attenuated or inactivated pathogen preparations. Biochemical and structural studies of class II MHC-peptide complexes are beginning to provide a conceptual foundation for the rational design of subunit and peptide vaccines. In this review, we show how analysis of peptide-class II MHC complexes together with developing understanding of antigen processing pathways has opened the door to understanding the major rules that govern selection of T cell epitopes. We review progress towards computational prediction of such epitopes, and efforts to evaluate algorithms that incorporate various structural and/or biochemical aspects of the MHC-peptide interaction. Finally, using malaria as a model, we describe the development of a minimal subunit vaccine for the human malaria parasite Plasmodium falciparum.
Collapse
Affiliation(s)
- Lawrence J Stern
- Department of Pathology, Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | |
Collapse
|
49
|
BoLA-DR peptide binding pockets are fundamental for foot-and-mouth disease virus vaccine design in cattle. Vaccine 2009; 28:28-37. [DOI: 10.1016/j.vaccine.2009.09.131] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 08/20/2009] [Accepted: 09/30/2009] [Indexed: 11/24/2022]
|
50
|
Thomas C, Moridani M. Interindividual variations in the efficacy and toxicity of vaccines. Toxicology 2009; 278:204-10. [PMID: 19837123 DOI: 10.1016/j.tox.2009.10.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 10/08/2009] [Indexed: 01/29/2023]
Abstract
A number of currently available vaccines have shown significant differences in the magnitude of immune responses and toxicity in individuals undergoing vaccination. A number of factors may be involved in the variations in immune responses, which include age, gender, race, amount and quality of the antigen, the dose administered and to some extent the route of administration, and genetics of immune system. Hence, it becomes imperative that researchers have tools such as genomics and proteomics at their disposal to predict which set of population is more likely to be non-responsive or develop toxicity to vaccines. In this article, we briefly review the influence of pharmacogenomics biomarkers on the efficacy and toxicity of some of the most frequently reported vaccines that showed a high rate of variability in response and toxicity towards hepatitis B, measles, mumps, rubella, influenza, and AIDS/HIV.
Collapse
Affiliation(s)
- Chandan Thomas
- Department of Pharmaceutical Sciences, School of Pharmacy, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | | |
Collapse
|