1
|
Santi T, Jo J, Harahap AR, Werdhani RA, Hadinegoro SRS, SahBandar IN, Prayitno A, Munasir Z, Vandenplas Y, Hegar B. The Improvement of Adaptive Immune Responses towards COVID-19 Following Diphtheria-Tetanus-Pertussis and SARS-CoV-2 Vaccinations in Indonesian Children: Exploring the Roles of Heterologous Immunity. Vaccines (Basel) 2024; 12:1032. [PMID: 39340062 PMCID: PMC11435621 DOI: 10.3390/vaccines12091032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/20/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Routine childhood vaccination, e.g., for diphtheria, tetanus, and pertussis (DTP), might provide additional protection against SARS-CoV-2 infection. This concept of heterologous immunity was explored in healthy children receiving both DTP and inactivated SARS-CoV-2 vaccines. METHODS A cross-sectional study was performed on 154 healthy children aged 6-8 years old in Jakarta, Indonesia. Their vaccination status for the DTP (including a diphtheria-tetanus booster vaccine at 5 years old) and CoronaVac (from 6 years old) vaccines were recorded. Peripheral blood samples were collected from all participants, in which anti-diphtheria toxoid IgG and anti-SARS-CoV-2 S-RBD antibodies and T cell-derived IFN-γ were measured. RESULTS The study participants with complete DTP vaccination had significantly higher titers of anti-diphtheria toxoid IgG than the ones without (median = 0.9349 versus 0.2113 IU/mL; p < 0.0001). Upon stratification based on DTP and CoronaVac vaccination statuses, the participants with complete DTP and CoronaVac vaccinations had the highest titer of anti-SARS-CoV-2 S-RBD antibodies (median = 1196 U/mL) and the highest concentration of SARS-CoV-2-specific T cell-derived IFN-γ (median = 560.9 mIU/mL) among all the groups. CONCLUSIONS Healthy children aged 6-8 years old with complete DTP and CoronaVac vaccinations exhibited stronger SARS-CoV-2-specific T cell immune responses. This might suggest an additional benefit of routine childhood vaccination in generating protection against novel pathogens, presumably via heterologous immunity.
Collapse
Affiliation(s)
- Theresia Santi
- Doctoral Program in Medical Science, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Juandy Jo
- Department of Biology, Faculty of Health Sciences, Universitas Pelita Harapan, Tangerang 15811, Indonesia
- Mochtar Riady Institute for Nanotechnology, Tangerang 15811, Indonesia
| | - Alida Roswita Harahap
- Doctoral Program in Medical Science, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Retno Asti Werdhani
- Department of Community Medicine, Cipto Mangunkkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Sri Rezeki S Hadinegoro
- Department of Child Health, Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Ivo Novita SahBandar
- Department of Microbiology, School of Medicine, Iwate Medical University, Morioka 028-3694, Japan
| | - Ari Prayitno
- Department of Child Health, Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Zakiudin Munasir
- Department of Child Health, Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Yvan Vandenplas
- Department of Pediatric, Universitair Ziekenhuis Brussel, 1090 Jette, Belgium
| | - Badriul Hegar
- Department of Child Health, Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Indonesian Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| |
Collapse
|
2
|
Peralta Alvarez MP, Jones H, Redondo Azema H, Davis C, White AD, Sarfas C, Dennis M, Li S, Wright D, Puentes E, Kimuda S, Belij-Rammerstorfer S, Aguilo N, Martin C, Sharpe S, McShane H, Tanner R. Low-dose M.tb infection but not BCG or MTBVAC vaccination enhances heterologous antibody titres in non-human primates. Front Immunol 2024; 15:1387454. [PMID: 38799468 PMCID: PMC11116990 DOI: 10.3389/fimmu.2024.1387454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Mycobacteria are known to exert a range of heterologous effects on the immune system. The mycobacteria-based Freund's Complete Adjuvant is a potent non-specific stimulator of the immune response used in immunization protocols promoting antibody production, and Mycobacterium bovis Bacille Calmette Guérin (BCG) vaccination has been linked with decreased morbidity and mortality beyond the specific protection it provides against tuberculosis (TB) in some populations and age groups. The role of heterologous antibodies in this phenomenon, if any, remains unclear and under-studied. Methods We set out to evaluate antibody responses to a range of unrelated pathogens following infection with Mycobacterium tuberculosis (M.tb) and vaccination with BCG or a candidate TB vaccine, MTBVAC, in non-human primates. Results We demonstrate a significant increase in the titer of antibodies against SARS-CoV-2, cytomegalovirus, Epstein-Barr virus, tetanus toxoid, and respiratory syncytial virus antigens following low-dose aerosol infection with M.tb. The magnitude of some of these responses correlated with TB disease severity. However, vaccination with BCG administered by the intradermal, intravenous or aerosol routes, or intradermal delivery of MTBVAC, did not increase antibody responses against unrelated pathogens. Discussion Our findings suggest that it is unlikely that heterologous antibodies contribute to the non-specific effects of these vaccines. The apparent dysregulation of B cell responses associated with TB disease warrants further investigation, with potential implications for risk of B cell cancers and novel therapeutic strategies.
Collapse
Affiliation(s)
| | - Holly Jones
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Hugo Redondo Azema
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Chloe Davis
- Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Andrew D. White
- United Kingdom (UK) Health Security Agency, Salisbury, United Kingdom
| | - Charlotte Sarfas
- United Kingdom (UK) Health Security Agency, Salisbury, United Kingdom
| | - Mike Dennis
- United Kingdom (UK) Health Security Agency, Salisbury, United Kingdom
| | - Shuailin Li
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Daniel Wright
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Eugenia Puentes
- Clinical Research Department y Research and Development Department, Biofabri, Grupo Zendal, Pontevedra, Spain
| | - Simon Kimuda
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, London, United Kingdom
| | | | - Nacho Aguilo
- University of Zaragoza, Spanish Network for Research on Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Martin
- University of Zaragoza, Spanish Network for Research on Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Sally Sharpe
- United Kingdom (UK) Health Security Agency, Salisbury, United Kingdom
| | - Helen McShane
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Rachel Tanner
- Department of Biology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
3
|
Zimmermann P, Curtis N. Why Does the Severity of COVID-19 Differ With Age?: Understanding the Mechanisms Underlying the Age Gradient in Outcome Following SARS-CoV-2 Infection. Pediatr Infect Dis J 2022; 41:e36-e45. [PMID: 34966142 PMCID: PMC8740029 DOI: 10.1097/inf.0000000000003413] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/03/2021] [Indexed: 11/26/2022]
Abstract
Although there are many hypotheses for the age-related difference in the severity of COVID-19, differences in innate, adaptive and heterologous immunity, together with differences in endothelial and clotting function, are the most likely mechanisms underlying the marked age gradient. Children have a faster and stronger innate immune response to SARS-CoV-2, especially in the nasal mucosa, which rapidly controls the virus. In contrast, adults can have an overactive, dysregulated and less effective innate response that leads to uncontrolled pro-inflammatory cytokine production and tissue injury. More recent exposure to other viruses and routine vaccines in children might be associated with protective cross-reactive antibodies and T cells against SARS-CoV-2. There is less evidence to support other mechanisms that have been proposed to explain the age-related difference in outcome following SARS-CoV-2 infection, including pre-existing immunity from exposure to common circulating coronaviruses, differences in the distribution and expression of the entry receptors ACE2 and TMPRSS2, and difference in viral load.
Collapse
Affiliation(s)
- Petra Zimmermann
- From the Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Department of Paediatrics, Fribourg Hospital HFR, Fribourg, Switzerland
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, Australia
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, Australia
- Infectious Diseases Unit, The Royal Children’s Hospital Melbourne, Parkville, Australia
| |
Collapse
|
4
|
Tarabini RF, Rigo MM, Faustino Fonseca A, Rubin F, Bellé R, Kavraki LE, Ferreto TC, Amaral Antunes D, de Souza APD. Large-Scale Structure-Based Screening of Potential T Cell Cross-Reactivities Involving Peptide-Targets From BCG Vaccine and SARS-CoV-2. Front Immunol 2022; 12:812176. [PMID: 35095907 PMCID: PMC8793865 DOI: 10.3389/fimmu.2021.812176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/21/2021] [Indexed: 12/22/2022] Open
Abstract
Although not being the first viral pandemic to affect humankind, we are now for the first time faced with a pandemic caused by a coronavirus. The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has been responsible for the COVID-19 pandemic, which caused more than 4.5 million deaths worldwide. Despite unprecedented efforts, with vaccines being developed in a record time, SARS-CoV-2 continues to spread worldwide with new variants arising in different countries. Such persistent spread is in part enabled by public resistance to vaccination in some countries, and limited access to vaccines in other countries. The limited vaccination coverage, the continued risk for resistant variants, and the existence of natural reservoirs for coronaviruses, highlight the importance of developing additional therapeutic strategies against SARS-CoV-2 and other coronaviruses. At the beginning of the pandemic it was suggested that countries with Bacillus Calmette-Guérin (BCG) vaccination programs could be associated with a reduced number and/or severity of COVID-19 cases. Preliminary studies have provided evidence for this relationship and further investigation is being conducted in ongoing clinical trials. The protection against SARS-CoV-2 induced by BCG vaccination may be mediated by cross-reactive T cell lymphocytes, which recognize peptides displayed by class I Human Leukocyte Antigens (HLA-I) on the surface of infected cells. In order to identify potential targets of T cell cross-reactivity, we implemented an in silico strategy combining sequence-based and structure-based methods to screen over 13,5 million possible cross-reactive peptide pairs from BCG and SARS-CoV-2. Our study produced (i) a list of immunogenic BCG-derived peptides that may prime T cell cross-reactivity against SARS-CoV-2, (ii) a large dataset of modeled peptide-HLA structures for the screened targets, and (iii) new computational methods for structure-based screenings that can be used by others in future studies. Our study expands the list of BCG peptides potentially involved in T cell cross-reactivity with SARS-CoV-2-derived peptides, and identifies multiple high-density "neighborhoods" of cross-reactive peptides which could be driving heterologous immunity induced by BCG vaccination, therefore providing insights for future vaccine development efforts.
Collapse
Affiliation(s)
- Renata Fioravanti Tarabini
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Health Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | | | - André Faustino Fonseca
- Antunes Lab, Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Felipe Rubin
- School of Technology - Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Rafael Bellé
- Laboratório de alto desempenho – Centro de Apoio ao desenvolvimento cientifico e tecnológico da (IDEIA), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Lydia E Kavraki
- Kavraki Lab, Department of Computer Science, Rice University, Houston, TX, United States
| | - Tiago Coelho Ferreto
- School of Technology - Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil,Laboratório de alto desempenho – Centro de Apoio ao desenvolvimento cientifico e tecnológico da (IDEIA), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Dinler Amaral Antunes
- Antunes Lab, Department of Biology and Biochemistry, University of Houston, Houston, TX, United States,*Correspondence: Ana Paula Duarte de Souza, ; Dinler Amaral Antunes,
| | - Ana Paula Duarte de Souza
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Health Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil,*Correspondence: Ana Paula Duarte de Souza, ; Dinler Amaral Antunes,
| |
Collapse
|
5
|
Fiege JK, Block KE, Pierson MJ, Nanda H, Shepherd FK, Mickelson CK, Stolley JM, Matchett WE, Wijeyesinghe S, Meyerholz DK, Vezys V, Shen SS, Hamilton SE, Masopust D, Langlois RA. Mice with diverse microbial exposure histories as a model for preclinical vaccine testing. Cell Host Microbe 2021; 29:1815-1827.e6. [PMID: 34731647 DOI: 10.1016/j.chom.2021.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 08/30/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022]
Abstract
Laboratory mice comprise an expeditious model for preclinical vaccine testing; however, vaccine immunogenicity in these models often inadequately translates to humans. Reconstituting physiologic microbial experience to specific pathogen-free (SPF) mice induces durable immunological changes that better recapitulate human immunity. We examined whether mice with diverse microbial experience better model human responses post vaccination. We co-housed laboratory mice with pet-store mice, which have varied microbial exposures, and then assessed immune responses to influenza vaccines. Human transcriptional responses to influenza vaccination are better recapitulated in co-housed mice. Although SPF and co-housed mice were comparably susceptible to acute influenza infection, vaccine-induced humoral responses were dampened in co-housed mice, resulting in poor control upon challenge. Additionally, protective heterosubtypic T cell immunity was compromised in co-housed mice. Because SPF mice exaggerated humoral and T cell protection upon influenza vaccination, reconstituting microbial experience in laboratory mice through co-housing may better inform preclinical vaccine testing.
Collapse
Affiliation(s)
- Jessica K Fiege
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katharine E Block
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mark J Pierson
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hezkiel Nanda
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Frances K Shepherd
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Clayton K Mickelson
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - J Michael Stolley
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - William E Matchett
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sathi Wijeyesinghe
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - David K Meyerholz
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Vaiva Vezys
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Steven S Shen
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sara E Hamilton
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Ryan A Langlois
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
6
|
Mysore V, Cullere X, Settles ML, Ji X, Kattan MW, Desjardins M, Durbin-Johnson B, Gilboa T, Baden LR, Walt DR, Lichtman AH, Jehi L, Mayadas TN. Protective heterologous T cell immunity in COVID-19 induced by the trivalent MMR and Tdap vaccine antigens. MED 2021; 2:1050-1071.e7. [PMID: 34414383 PMCID: PMC8363466 DOI: 10.1016/j.medj.2021.08.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/25/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND T cells control viral infection, promote vaccine durability, and in coronavirus disease 2019 (COVID-19) associate with mild disease. We investigated whether prior measles-mumps-rubella (MMR) or tetanus-diphtheria-pertussis (Tdap) vaccination elicits cross-reactive T cells that mitigate COVID-19. METHODS Antigen-presenting cells (APC) loaded ex vivo with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), MMR, or Tdap antigens and autologous T cells from COVID-19-convalescent participants, uninfected individuals, and COVID-19 mRNA-vaccinated donors were co-cultured. T cell activation and phenotype were detected by interferon-γ (IFN-γ) enzyme-linked immunospot (ELISpot) assays and flow cytometry. ELISAs (enzyme-linked immunosorbant assays) and validation studies identified the APC-derived cytokine(s) driving T cell activation. TCR clonotyping and single-cell RNA sequencing (scRNA-seq) identified cross-reactive T cells and their transcriptional profile. A propensity-weighted analysis of COVID-19 patients estimated the effects of MMR and Tdap vaccination on COVID-19 outcomes. FINDINGS High correlation was observed between T cell responses to SARS-CoV-2 (spike-S1 and nucleocapsid) and MMR and Tdap proteins in COVID-19-convalescent and -vaccinated individuals. The overlapping T cell population contained an effector memory T cell subset (effector memory re-expressing CD45RA on T cells [TEMRA]) implicated in protective, anti-viral immunity, and their detection required APC-derived IL-15, known to sensitize T cells to activation. Cross-reactive TCR repertoires detected in antigen-experienced T cells recognizing SARS-CoV-2, MMR, and Tdap epitopes had TEMRA features. Indices of disease severity were reduced in MMR- or Tdap-vaccinated individuals by 32%-38% and 20%-23%, respectively, among COVID-19 patients. CONCLUSIONS Tdap and MMR memory T cells reactivated by SARS-CoV-2 may provide protection against severe COVID-19. FUNDING This study was supported by a National Institutes of Health (R01HL065095, R01AI152522, R01NS097719) donation from Barbara and Amos Hostetter and the Chleck Foundation.
Collapse
Affiliation(s)
- Vijayashree Mysore
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Xavier Cullere
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Matthew L Settles
- Bioinformatics Core Facility in the Genome Center, University of California, Davis, Davis, CA 95616, USA
| | - Xinge Ji
- Quantitative Health Science Department, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michael W Kattan
- Quantitative Health Science Department, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michaël Desjardins
- Department of Medicine, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | | | - Tal Gilboa
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Lindsey R Baden
- Department of Medicine, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - David R Walt
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Lara Jehi
- Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Tanya N Mayadas
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
7
|
Laupèze B, Del Giudice G, Doherty MT, Van der Most R. Vaccination as a preventative measure contributing to immune fitness. NPJ Vaccines 2021; 6:93. [PMID: 34315886 PMCID: PMC8316335 DOI: 10.1038/s41541-021-00354-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
The primary goal of vaccination is the prevention of pathogen-specific infection. The indirect consequences may include maintenance of homeostasis through prevention of infection-induced complications; trained immunity that re-programs innate cells to respond more efficiently to later, unrelated threats; slowing or reversing immune senescence by altering the epigenetic clock, and leveraging the pool of memory B and T cells to improve responses to new infections. Vaccines may exploit the plasticity of the immune system to drive longer-term immune responses that promote health at a broader level than just the prevention of single, specific infections. In this perspective, we discuss the concept of “immune fitness” and how to potentially build a resilient immune system that could contribute to better health. We argue that vaccines may contribute positively to immune fitness in ways that are only beginning to be understood, and that life-course vaccination is a fundamental tool for achieving healthy aging.
Collapse
|
8
|
Mysore V, Cullere X, Settles ML, Ji X, Kattan MW, Desjardins M, Durbin-Johnson B, Gilboa T, Baden LR, Walt DR, Lichtman A, Jehi L, Mayadas TN. Protective heterologous T cell immunity in COVID-19 induced by MMR and Tdap vaccine antigens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.05.03.441323. [PMID: 33972940 PMCID: PMC8109200 DOI: 10.1101/2021.05.03.441323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
T cells are critical for control of viral infection and effective vaccination. We investigated whether prior Measles-Mumps-Rubella (MMR) or Tetanus-Diphtheria-pertussis (Tdap) vaccination elicit cross-reactive T cells that mitigate COVID-19. Using co-cultures of antigen presenting cells (APC) loaded with antigens and autologous T cells, we found a high correlation between responses to SARS-CoV-2 (Spike-S1 and Nucleocapsid) and MMR and Tdap vaccine proteins in both SARS-CoV-2 infected individuals and individuals immunized with mRNA-based SARS-CoV-2 vaccines. The overlapping T cell population contained effector memory T cells (TEMRA) previously implicated in anti-viral immunity and their activation required APC-derived IL-15. TCR- and scRNA-sequencing detected cross-reactive clones with TEMRA features among the cells recognizing SARS-CoV-2, MMR and Tdap epitopes. A propensity-weighted analysis of 73,582 COVID-19 patients revealed that severe disease outcomes (hospitalization and transfer to intensive care unit or death) were reduced in MMR or Tdap vaccinated individuals by 38-32% and 23-20% respectively. In summary, SARS-CoV-2 re-activates memory T cells generated by Tdap and MMR vaccines, which may reduce disease severity.
Collapse
|
9
|
Stojanovic M, Lukic I, Marinkovic E, Kovacevic A, Miljkovic R, Tobias J, Schabussova I, Zlatović M, Barisani-Asenbauer T, Wiedermann U, Inic-Kanada A. Cross-Reactive Effects of Vaccines: Heterologous Immunity between Tetanus and Chlamydia. Vaccines (Basel) 2020; 8:vaccines8040719. [PMID: 33271962 PMCID: PMC7712554 DOI: 10.3390/vaccines8040719] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Vaccines can have heterologous effects on the immune system, i.e., effects other than triggering an immune response against the disease targeted by the vaccine. We investigated whether monoclonal antibodies (mAbs) specific for tetanus could cross-react with Chlamydia and confer heterologous protection against chlamydial infection. The capability of two tetanus-specific mAbs, namely mAb26 and mAb51, to prevent chlamydial infection has been assessed: (i) in vitro, by performing a neutralization assay using human conjunctival epithelial (HCjE) cells infected with Chlamydia trachomatis serovar B, and (ii) in vivo, by using a guinea pig model of Chlamydiacaviae-induced inclusion conjunctivitis. The mAb26 has been superior in comparison with mAb51 in the prevention of chlamydial infection in HCjE cells. The mAb26 has conferred ≈40% inhibition of the infection, compared to less than 5% inhibition in the presence of the mAb51. In vivo, mAb26 significantly diminished ocular pathology intensity in guinea pigs infected with C. caviae compared to either the mAb51-treated or sham-treated guinea pigs. Our data provide insights that tetanus immunization generates antibodies which induce heterologous chlamydial immunity and promote protection beyond the intended target pathogen.
Collapse
Affiliation(s)
- Marijana Stojanovic
- Institute of Virology, Vaccines, and Sera–TORLAK, 11152 Belgrade, Serbia; (M.S.); (I.L.); (E.M.); (A.K.); (R.M.)
| | - Ivana Lukic
- Institute of Virology, Vaccines, and Sera–TORLAK, 11152 Belgrade, Serbia; (M.S.); (I.L.); (E.M.); (A.K.); (R.M.)
| | - Emilija Marinkovic
- Institute of Virology, Vaccines, and Sera–TORLAK, 11152 Belgrade, Serbia; (M.S.); (I.L.); (E.M.); (A.K.); (R.M.)
| | - Ana Kovacevic
- Institute of Virology, Vaccines, and Sera–TORLAK, 11152 Belgrade, Serbia; (M.S.); (I.L.); (E.M.); (A.K.); (R.M.)
| | - Radmila Miljkovic
- Institute of Virology, Vaccines, and Sera–TORLAK, 11152 Belgrade, Serbia; (M.S.); (I.L.); (E.M.); (A.K.); (R.M.)
| | - Joshua Tobias
- Center for Pathophysiology Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.T.); (I.S.); (T.B.-A.); (U.W.)
| | - Irma Schabussova
- Center for Pathophysiology Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.T.); (I.S.); (T.B.-A.); (U.W.)
| | - Mario Zlatović
- Faculty of Chemistry, University of Belgrade, 11000 Belgrade, Serbia;
| | - Talin Barisani-Asenbauer
- Center for Pathophysiology Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.T.); (I.S.); (T.B.-A.); (U.W.)
| | - Ursula Wiedermann
- Center for Pathophysiology Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.T.); (I.S.); (T.B.-A.); (U.W.)
| | - Aleksandra Inic-Kanada
- Center for Pathophysiology Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.T.); (I.S.); (T.B.-A.); (U.W.)
- Correspondence: ; Tel.: +43-1-40160-33-154
| |
Collapse
|
10
|
Ben Fredj M, Dhouib W, Kacem M, Bennasrallah C, Mehrez O, Abroug H, Zemni I, Gardabou A, Jamel K, Chouchene S, Rouatbi N, Belguith Sriha A. Trends and health burden of hospitalized acute respiratory infections and impact of Haemophilus influenza immunization in a Tunisian university hospital: a twelve-year study. Libyan J Med 2020; 15:1783048. [PMID: 32552441 PMCID: PMC7482779 DOI: 10.1080/19932820.2020.1783048] [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: 12/17/2019] [Accepted: 06/09/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND We aimed to describe the episodes and trends of admissions for community-acquired Respiratory Infections (RI) over a 12-year period and to assess the impact of Haemophilus influenza type b (Hib) vaccine on RI admissions in children aged up to 3 years. METHODS We conducted a twelve-year retrospective observational study on all community-acquired RI admitted to Fattouma Bourguiba Hospital in Monastir Governorate (Tunisia) from 1 January 2002 to 31 December 2013. RI cases were selected from the Regional Registry of Hospital Morbidity. Data were coded according to ICD-10. To assess the impact of the Hib vaccine, three cohorts were defined based on vaccine status (unvaccinated cohort, first vaccinated cohort (VC) by monovalent form and second VC by pentavalent combination). RESULTS Admissions for RI represented 17.6% (CI95%: 17.3-18.1) of all communicable diseases hospitalizations (n = 6 061/34 289). The crude incidence rates (CIR) per 100,000 inh were 24.2 for upper RI (URI) and 77.5 and for Lower RI (LRI) (p < 0.0001). Pneumonias represented 53.9% of LRI. Sex-ratio (male/female) was 1.12 for URI and 1.64 for LRI (p < 0.0001). At admission, the median age was 22 years (IQR: 3-52). Admission for Pneumonia increased significantly during study period (slope 'b' = 5.16; p < 0.0001) especially in children up to 5 years old (slope 'b' = 5.53) and in elderly (slope 'b' = 2.13). Among children up to 3 years old, the CIRs per 100,000 for Hib pneumonia admission were 11.6 in Non-Vaccinated Cohort (NVC), 10.6 in Vaccinated Cohort (VC) by protocol 1 (Hib Vaccine monovalent) and 0.80 in VC by protocol 2 (pentavalent vaccine combination).The relative risk reduction was 99% for protocol 2 (p < 0.001). CONCLUSION Admissions for RI in a tertiary level hospital were common with an increasing trend. The Hib immunization program, in particular the pentavalent combination, has had a positive impact on the reduction of related acute diseases.
Collapse
Affiliation(s)
- Manel Ben Fredj
- Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
- Department of Preventive and Community Medicine, University Hospital Fattouma Bourguiba, Monastir, Tunisia
- Research Laboratory “Technology and Medical Imaging” 12ES06, Tunisia
| | - Wafa Dhouib
- Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
- Department of Preventive and Community Medicine, University Hospital Fattouma Bourguiba, Monastir, Tunisia
- Research Laboratory “Technology and Medical Imaging” 12ES06, Tunisia
| | - Meriem Kacem
- Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
- Department of Preventive and Community Medicine, University Hospital Fattouma Bourguiba, Monastir, Tunisia
- Research Laboratory “Technology and Medical Imaging” 12ES06, Tunisia
| | - Cyrine Bennasrallah
- Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
- Department of Preventive and Community Medicine, University Hospital Fattouma Bourguiba, Monastir, Tunisia
- Research Laboratory “Technology and Medical Imaging” 12ES06, Tunisia
| | - Ons Mehrez
- Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
| | - Hela Abroug
- Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
- Department of Preventive and Community Medicine, University Hospital Fattouma Bourguiba, Monastir, Tunisia
- Research Laboratory “Technology and Medical Imaging” 12ES06, Tunisia
| | - Imen Zemni
- Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
- Department of Preventive and Community Medicine, University Hospital Fattouma Bourguiba, Monastir, Tunisia
- Research Laboratory “Technology and Medical Imaging” 12ES06, Tunisia
| | - Aicha Gardabou
- Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
| | - Koubaa Jamel
- Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
- Otorhinolaryngology Department, University Hospital Fattouma Bourguiba, Monastir, Tunisia
| | - Slaheddine Chouchene
- Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
- Pediatric Department, University Hospital Fattouma Bourguiba, Monastir, Tunisia
| | - Naceur Rouatbi
- Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
- Pneumology Department, University Hospital Fattouma Bourguiba, Monastir, Tunisia
| | - Asma Belguith Sriha
- Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
- Department of Preventive and Community Medicine, University Hospital Fattouma Bourguiba, Monastir, Tunisia
- Research Laboratory “Technology and Medical Imaging” 12ES06, Tunisia
| |
Collapse
|
11
|
Bulut O, Kilic G, Domínguez-Andrés J, Netea MG. Overcoming immune dysfunction in the elderly: trained immunity as a novel approach. Int Immunol 2020; 32:741-753. [PMID: 32766848 PMCID: PMC7680842 DOI: 10.1093/intimm/dxaa052] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/29/2020] [Indexed: 12/20/2022] Open
Abstract
People with advanced age have a higher susceptibility to infections and exhibit increased mortality and morbidity as the ability of the immune system to combat infections decreases with age. While innate immune cells display functional defects such as decreased phagocytosis, chemotaxis and cytokine production, adaptive immune cells exhibit reduced receptor diversity, defective antibody production and a sharp decline in naive cell populations. Successful responses to vaccination in the elderly are critical to prevent common infections such as influenza and pneumonia, but vaccine efficacy decreases in older individuals compared with young adults. Trained immunity is a newly emerging concept that showed that innate immune cells possess non-specific immunological memory established through epigenetic and metabolic reprogramming upon encountering certain pathogenic stimuli. Clinical studies suggest that trained immunity can be utilized to enhance immune responses against infections and improve the efficiency of vaccinations in adults; however, how trained immunity responses are shaped with advanced age is still an open question. In this review, we provide an overview of the age-related changes in the immune system with a focus on innate immunity, discuss current vaccination strategies for the elderly, present the concept of trained immunity and propose it as a novel approach to enhance responses against infections and vaccinations in the elderly population.
Collapse
Affiliation(s)
- Ozlem Bulut
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, GA Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, GA Nijmegen, The Netherlands
| | - Gizem Kilic
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, GA Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, GA Nijmegen, The Netherlands
| | - Jorge Domínguez-Andrés
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, GA Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, GA Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, GA Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, GA Nijmegen, The Netherlands
- Department of Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
12
|
Fischinger S, Dolatshahi S, Jennewein MF, Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Michael N, Vasan S, Ackerman ME, Streeck H, Alter G. IgG3 collaborates with IgG1 and IgA to recruit effector function in RV144 vaccinees. JCI Insight 2020; 5:140925. [PMID: 33031099 PMCID: PMC7710302 DOI: 10.1172/jci.insight.140925] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/30/2020] [Indexed: 01/08/2023] Open
Abstract
While the RV144 HIV vaccine trial led to moderately reduced risk of HIV acquisition, emerging data from the HVTN702 trial point to the critical need to reexamine RV144-based correlates of reduced risk of protection. While in RV144, the induction of V2-binding, non-IgA, IgG3 antibody responses with nonneutralizing functions were linked to reduced risk of infection, the interactions between these signatures remain unclear. Thus, here we comprehensively profile the humoral immune response in 300 RV144 vaccinees to decipher the relationships between humoral biomarkers of protection. We found that vaccine-specific IgG1, IgG3, and IgA were highly correlated. However, ratios of IgG1:IgG3:IgA provided insights into subclass/isotype polyclonal functional regulation. For instance, in the absence of high IgG1 levels, IgG3 antibodies exhibited limited functional activity, pointing to IgG3 as a critical contributor, but not sole driver, of effective antiviral humoral immunity. Higher IgA levels were linked to enhanced antibody effector function, including neutrophil phagocytosis (ADNP), complement deposition (ADCD), and antibody-dependent NK degranulation (CD107a), some of which were increased in infected vaccinees in a case/control data set, suggesting that IgA-driven functions compromised immunity. These data highlight the interplay between IgG1, IgG3, and IgA, pointing to the need to profile the relationships between subclass/isotype selection. The induction of V2-binding, non-IgA, IgG3 antibody responses with non-neutralizing functions were linked to reduced risk of infection in RV144 vaccinees.
Collapse
Affiliation(s)
- Stephanie Fischinger
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA.,PhD Program of Virology and Immunology, University of Duisburg-Essen, Essen, Germany
| | | | | | | | | | | | | | - Sandhya Vasan
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Hendrik Streeck
- Institute of Virology, Universitätsklinikum Bonn, Bonn, Germany
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
13
|
Lee CH, Salio M, Napolitani G, Ogg G, Simmons A, Koohy H. Predicting Cross-Reactivity and Antigen Specificity of T Cell Receptors. Front Immunol 2020; 11:565096. [PMID: 33193332 PMCID: PMC7642207 DOI: 10.3389/fimmu.2020.565096] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022] Open
Abstract
Adaptive immune recognition is mediated by specific interactions between heterodimeric T cell receptors (TCRs) and their cognate peptide-MHC (pMHC) ligands, and the methods to accurately predict TCR:pMHC interaction would have profound clinical, therapeutic and pharmaceutical applications. Herein, we review recent developments in predicting cross-reactivity and antigen specificity of TCR recognition. We discuss current experimental and computational approaches to investigate cross-reactivity and antigen-specificity of TCRs and highlight how integrating kinetic, biophysical and structural features may offer valuable insights in modeling immunogenicity. We further underscore the close inter-relationship of these two interconnected notions and the need to investigate each in the light of the other for a better understanding of T cell responsiveness for the effective clinical applications.
Collapse
Affiliation(s)
- Chloe H. Lee
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC WIMM Centre for Computational Biology, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Mariolina Salio
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Giorgio Napolitani
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Graham Ogg
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Alison Simmons
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford, United Kingdom
| | - Hashem Koohy
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC WIMM Centre for Computational Biology, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Larenas-Linnemann D, Rodríguez-Pérez N, Arias-Cruz A, Blandón-Vijil MV, Del Río-Navarro BE, Estrada-Cardona A, Gereda JE, Luna-Pech JA, Navarrete-Rodríguez EM, Onuma-Takane E, Pozo-Beltrán CF, Rojo-Gutiérrez MI. Enhancing innate immunity against virus in times of COVID-19: Trying to untangle facts from fictions. World Allergy Organ J 2020; 13:100476. [PMID: 33072240 PMCID: PMC7546230 DOI: 10.1016/j.waojou.2020.100476] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/24/2020] [Accepted: 09/30/2020] [Indexed: 12/14/2022] Open
Abstract
Introduction In light of the current COVID-19 pandemic, during which the world is confronted with a new, highly contagious virus that suppresses innate immunity as one of its initial virulence mechanisms, thus escaping from first-line human defense mechanisms, enhancing innate immunity seems a good preventive strategy. Methods Without the intention to write an official systematic review, but more to give an overview of possible strategies, in this review article we discuss several interventions that might stimulate innate immunity and thus our defense against (viral) respiratory tract infections. Some of these interventions can also stimulate the adaptive T- and B-cell responses, but our main focus is on the innate part of immunity. We divide the reviewed interventions into: 1) lifestyle related (exercise, >7 h sleep, forest walking, meditation/mindfulness, vitamin supplementation); 2) Non-specific immune stimulants (letting fever advance, bacterial vaccines, probiotics, dialyzable leukocyte extract, pidotimod), and 3) specific vaccines with heterologous effect (BCG vaccine, mumps-measles-rubeola vaccine, etc). Results For each of these interventions we briefly comment on their definition, possible mechanisms and evidence of clinical efficacy or lack of it, especially focusing on respiratory tract infections, viral infections, and eventually a reduced mortality in severe respiratory infections in the intensive care unit. At the end, a summary table demonstrates the best trials supporting (or not) clinical evidence. Conclusion Several interventions have some degree of evidence for enhancing the innate immune response and thus conveying possible benefit, but specific trials in COVID-19 should be conducted to support solid recommendations.
Collapse
Key Words
- ACE2, Angiotensin converting enzime-2
- APC, Antigen-presenting cell
- BCG, Bacillus Calmette-Guérin
- BV, Bacterial vaccine
- Bacillus calmette-guérin
- Bacterial vaccine
- CCL-5, Chemokine (C–C motif) ligand 5
- CI, Confidence interval
- CNS, Central nervous system
- COVID-19
- COVID-19, Coronavirus disease-2019
- CXCR3A, CXC chemokine receptor 3A
- DAMPs, Damage-associated molecular patterns
- DC, Dendritic cell
- DLE, Dialyzable leukocyte extract
- Exercise
- Gαs: G protein coupled receptor alfa-subunits, HSP
- Heat shock proteins, HLA-DR
- Immune response
- Immunoglobulin, IGFBP6
- Innate
- Insulin-like growth-factor-binding-protein 6, IL
- Intercellular adhesion molecule type 1, IFN
- Interferon, IG
- Interleukin, MBSR
- MCP-1, Monocyte chemoattractant protein-1
- MMR
- MODS, Multi-organ dysfunction syndrome
- Major histocompatibility complex class II cell surface receptor, ICAM-1
- Mindfulness
- Mindfulness-based stress reduction, mCa++: Intramitochondrial calcium
- MyD88, Myeloid differentiation primary response 88
- NF-κB, Nuclear factor kappaB
- NK, Natural killer
- NK-Cell
- NOD2, Nucleotide-binding oligomerization domain-containing protein 2
- OR, Odds ratio
- OxPhos: Oxidative phosphorylation, PAMPs
- PKC, Protein kinase C
- PPD, Purified protein derivative (tuberculin)
- PUFA, Polyunsaturated fatty acid
- Pathogen-associated molecular patterns, PBMC
- Peripheral blood mononuclear cell, PI3K/Akt: Phosphatidylinositol 3-kinase pathway
- R0: Basic reproduction number, REM
- Rapid eye movement, RIPK2
- Reactive nitrogen species, ROS
- Reactive oxygen species, SARS-CoV-2
- Receptor iteracting serine/threonine kinase 2, RNA
- Ribonucleic acid, RNS
- Severe acute respiratory syndrome coronavirus 2, SIRS
- Sleep
- Systemic inflammatory response syndrome, TCR:T-cell receptor
- TLR, Toll-like receptor
- TNF-α, Tumor necrosis factor alpha
- TRPV, Thermolabile calcium channels
- Th, T helper-cell
- Trained immunity
- URTI, Upper-respiratory tract infection
Collapse
Affiliation(s)
- Désirée Larenas-Linnemann
- Médica Sur, Clinical Foundation and Hospital, Mexico City, Mexico
- Corresponding author. Médica Sur, Fundación clínica y hospital, Puente de piedra 150, T2Toriello Guerra, Tlalpan, Ciudad de México, México, 14050, Mexico. E-mails:
| | | | - Alfredo Arias-Cruz
- State University of Nuevo León, School of Medicine and University Hospital Dr. José Eleuterio González, Monterrey, Nuevo Leon, Mexico
| | | | | | | | | | - Jorge A. Luna-Pech
- Departamento de Disciplinas Filosóficas, Metodológicas e Instrumentales (CUCS), Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | | | - Ernesto Onuma-Takane
- Fundación Clínica y Hospital Médica Sur, Ciudad de México, México, Mexico City, Mexico
| | | | | |
Collapse
|
15
|
Mendoza JL, Fischer S, Gee MH, Lam LH, Brackenridge S, Powrie FM, Birnbaum M, McMichael AJ, Garcia KC, Gillespie GM. Interrogating the recognition landscape of a conserved HIV-specific TCR reveals distinct bacterial peptide cross-reactivity. eLife 2020; 9:58128. [PMID: 32716298 PMCID: PMC7384859 DOI: 10.7554/elife.58128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/01/2020] [Indexed: 11/20/2022] Open
Abstract
T cell cross-reactivity ensures that diverse pathogen-derived epitopes encountered during a lifetime are recognized by the available TCR repertoire. A feature of cross-reactivity where previous exposure to one microbe can alter immunity to subsequent, non-related pathogens has been mainly explored for viruses. Yet cross-reactivity to additional microbes is important to consider, especially in HIV infection where gut-intestinal barrier dysfunction could facilitate T cell exposure to commensal/pathogenic microbes. Here we evaluated the cross-reactivity of a ‘public’, HIV-specific, CD8 T cell-derived TCR (AGA1 TCR) using MHC class I yeast display technology. Via screening of MHC-restricted libraries comprising ~2×108 sequence-diverse peptides, AGA1 TCR specificity was mapped to a central peptide di-motif. Using the top TCR-enriched library peptides to probe the non-redundant protein database, bacterial peptides that elicited functional responses by AGA1-expressing T cells were identified. The possibility that in context-specific settings, MHC class I proteins presenting microbial peptides influence virus-specific T cell populations in vivo is discussed.
Collapse
Affiliation(s)
- Juan L Mendoza
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States
| | - Suzanne Fischer
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States
| | - Marvin H Gee
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States
| | - Lilian H Lam
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | - Simon Brackenridge
- Nuffield Department of Medicine, University of Oxford, NDM Research Building, Old Road Campus, Headington, Oxford, United Kingdom
| | - Fiona M Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | - Michael Birnbaum
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States.,Koch Institute at MIT, Cambridge, United States
| | - Andrew J McMichael
- Nuffield Department of Medicine, University of Oxford, NDM Research Building, Old Road Campus, Headington, Oxford, United Kingdom
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, United States
| | - Geraldine M Gillespie
- Nuffield Department of Medicine, University of Oxford, NDM Research Building, Old Road Campus, Headington, Oxford, United Kingdom
| |
Collapse
|
16
|
Kharchenko EP. The Coronavirus SARS-Cov-2: the Complexity of Infection Pathogenesis, the Search of Vaccines and Possible Future Pandemics. ACTA ACUST UNITED AC 2020. [DOI: 10.31631/2073-3046-2020-19-3-4-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
17
|
Leng Q, Tarbe M, Long Q, Wang F. Pre-existing heterologous T-cell immunity and neoantigen immunogenicity. Clin Transl Immunology 2020; 9:e01111. [PMID: 32211191 PMCID: PMC7085466 DOI: 10.1002/cti2.1111] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/13/2020] [Accepted: 01/30/2020] [Indexed: 12/22/2022] Open
Abstract
Neoantigens are tumor‐specific mutated proteins that are exempt from central tolerance and are therefore capable of efficiently eliciting effective T‐cell responses. The identification of immunogenic neoantigens in tumor‐specific mutated proteins has promising clinical implications for cancer immunotherapy. However, the factors that may contribute to neoantigen immunogenicity are not yet fully understood. Through molecular mimicry of antigens arising during cancer progression, the gut microbiota and previously encountered pathogens potentially have profound impacts on T‐cell responses to previously unencountered tumor neoantigens. Here, we review the characteristics of immunogenic neoantigens and how host exposure to microbes may affect T‐cell responses to neoantigens. We address the hypothesis that pre‐existing heterologous memory T‐cell immunity is a major factor that influences neoantigen immunogenicity in individual cancer patients. Accumulating data suggest that differences in individual histories of microbial exposure should be taken into account during the optimisation of algorithms that predict neoantigen immunogenicity.
Collapse
Affiliation(s)
- Qibin Leng
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University State Key Laboratory of Respiratory Diseases, Guangzhou Medical University Guangzhou China.,The Joint Center for Infection and Immunity Guangzhou Women and Children's Medical Center Guangzhou Institute of Pediatrics Guangzhou Medical University Guangzhou China.,Institute Pasteur of Shanghai Chinese Academy of Science Shanghai China
| | - Marion Tarbe
- The Joint Center for Infection and Immunity Guangzhou Women and Children's Medical Center Guangzhou Institute of Pediatrics Guangzhou Medical University Guangzhou China.,Institute Pasteur of Shanghai Chinese Academy of Science Shanghai China
| | - Qi Long
- Department of Biostatistics, Epidemiology and Informatics Perelman School of Medicine University of Pennsylvania Philadelphia PA USA
| | - Feng Wang
- Department of Immunology and Microbiology Center for Microbiota and Immunological Diseases Shanghai General Hospital Shanghai Institute of Immunology Shanghai Jiao Tong University School of Medicine Shanghai China.,Research Center of Translational Medicine Shanghai Children's Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
18
|
Chang YC, Lin CJ, Hsiao YH, Chang YH, Liu SJ, Hsu HY. Therapeutic Effects of BCG Vaccination on Type 1 Diabetes Mellitus: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J Diabetes Res 2020; 2020:8954125. [PMID: 32309449 PMCID: PMC7139880 DOI: 10.1155/2020/8954125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/29/2020] [Accepted: 03/11/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) causes the irreversible destruction of pancreatic beta cells. The Bacillus Calmette-Guerin (BCG) vaccine can modulate the immune response and decelerate disease progression. The aim of this study is to investigate the efficacy of the BCG vaccine for the treatment of T1DM. OBJECTIVE Six databases were systematically searched from inception to the end of August 2019. The randomized controlled trials (RCTs) that evaluated glycemic control in response to the BCG vaccine for T1DM were enrolled. The primary outcome was glycated hemoglobin (HbA1c) level, and secondary outcomes included fasting and stimulated C-peptide level, daily insulin dosage, and clinical remission. The revised Cochrane risk of bias tool was used for quality assessment, and meta-analyses were conducted to evaluate the efficacy of the BCG vaccine. RESULTS Four studies with a total of 198 subjects were included. The results of HbA1c and fasting C-peptide levels were extracted for further quantitative assessment. The pooled meta-analysis demonstrated no significant difference in HbA1c levels (mean difference [MD], -0.12; 95% confidence interval [CI], -0.53 to 0.30; I 2 = 56%) or fasting C-peptide levels (MD, -0.15; 95% CI, -0.35 to 0.06; I 2 = 0%) in the BCG intervention group as compared with that in the placebo group. CONCLUSIONS There is no robust evidence to support the use of the BCG vaccine for the treatment of T1DM although the HbA1c levels tended to improve. Additional RCTs to assess the long-term effects of the BCG vaccine on glycemic control are warranted.
Collapse
Affiliation(s)
- Yu-Chen Chang
- Department of Family Medicine, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Chien-Ju Lin
- Department of Family Medicine, Hsinchu MacKay Memorial Hospital, No. 690, Section 2, Guangfu Road, East District, Hsinchu City 30071, Taiwan
| | - Yu-Hsuan Hsiao
- Department of Family Medicine, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Yu-Han Chang
- Department of Family Medicine, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, Taipei City 10449, Taiwan
| | - Shu-Jung Liu
- Department of Medical Library, MacKay Memorial Hospital, Tamsui Branch, No. 45, Minsheng Road, Tamsui District, New Taipei City 25160, Taiwan
| | - Hsin-Yin Hsu
- Department of Family Medicine, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, Taipei City 10449, Taiwan
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, No. 17, Xu-Zhou Rd., Taipei City 10055, Taiwan
| |
Collapse
|
19
|
Sumbria D, Berber E, Rouse BT. Factors Affecting the Tissue Damaging Consequences of Viral Infections. Front Microbiol 2019; 10:2314. [PMID: 31636623 PMCID: PMC6787772 DOI: 10.3389/fmicb.2019.02314] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 09/23/2019] [Indexed: 12/15/2022] Open
Abstract
Humans and animals are infected by multiple endogenous and exogenous viruses but few agents cause overt tissue damage. We review the circumstances which favor overt disease expression. These can include intrinsic virulence of the agent, new agents acquired from heterologous species, the circumstances of infection such as dose and route, current infection with other agents which includes the composition of the microbiome at mucosal and other sites, past history of exposure to other infections as well as the immune status of the host. We also briefly discuss promising therapeutic strategies that can expand immune response patterns that minimize tissue damaging responses to viral infections.
Collapse
Affiliation(s)
| | | | - Barry T. Rouse
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, Knoxville, TN, United States
| |
Collapse
|
20
|
Nelson SA, Sant AJ. Imprinting and Editing of the Human CD4 T Cell Response to Influenza Virus. Front Immunol 2019; 10:932. [PMID: 31134060 PMCID: PMC6514101 DOI: 10.3389/fimmu.2019.00932] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022] Open
Abstract
Immunity to influenza is unique among pathogens, in that immune memory is established both via intermittent lung localized infections with highly variable influenza virus strains and by intramuscular vaccinations with inactivated protein-based vaccines. Studies in the past decades have suggested that the B cell responses to influenza infection and vaccination are highly biased by an individual's early history of influenza infection. This reactivity likely reflects both the competitive advantage that memory B cells have in an immune response and the relatively limited diversity of epitopes in influenza hemagglutinin that are recognized by B cells. In contrast, CD4 T cells recognize a wide array of epitopes, with specificities that are heavily influenced by the diversity of influenza antigens available, and a multiplicity of functions that are determined by both priming events and subsequent confrontations with antigens. Here, we consider the events that prime and remodel the influenza-specific CD4 T cell response in humans that have highly diverse immune histories and how the CD4 repertoire may be edited in terms of functional potential and viral epitope specificity. We discuss the consequences that imprinting and remodeling may have on the potential of different human hosts to rapidly respond with protective cellular immunity to infection. Finally, these issues are discussed in the context of future avenues of investigation and vaccine strategies.
Collapse
Affiliation(s)
| | - Andrea J. Sant
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
21
|
Caputo M, Raupach-Rosin H, Karch A, Borte M, Lehmann I, Liebert UG, Standl M, Heinrich J, Mikolajczyk RT. Vaccinations and Infections Are Associated With Unrelated Antibody Titers: An Analysis From the German Birth Cohort Study LISA. Front Pediatr 2019; 7:254. [PMID: 31294004 PMCID: PMC6603196 DOI: 10.3389/fped.2019.00254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 06/04/2019] [Indexed: 11/13/2022] Open
Abstract
The evidence for non-specific effects (NSE) of vaccinations on all-cause morbidity and mortality among children is growing. However, our understanding of the underlying mechanisms is still limited. One hypothesis is that NSE are mediated by antibody titers. We used data of 2,123 children from the population-based birth cohort study LISA conducted in Germany to explore whether routine childhood vaccinations and the individual infection history in the first 2 years of life are associated with unrelated antibody titers. We selected 19 exposures (infections and vaccinations) and investigated their association with levels of 12 IgG antibody titers at the age of 2 years. Based on univariable analyses (ANOVA), we identified 21 crude associations between exposures and titers (p < 0.05), while 11 (95%-CI: 6, 17) spurious associations were expected due to multiple testing. In exploratory multivariable analyses, we observed associations between seven investigated IgG titers and 10 exposures; either administered vaccines [e.g., higher anti-hRSV IgG titer in BCG-vaccinated children (regression-coefficient in standard-deviation-units: 0.38; 95%-CI: 0.12, 0.65)] or infections [e.g., higher anti-measles IgG titer in children with reported chickenpox (0.44; 95%-CI: 0.08, 0.80)]. Our results indicate the existence of associations between immunogenic exposures and unrelated antibody titers. Further studies investigating the underlying immunological mechanisms are required.
Collapse
Affiliation(s)
- Mahrrouz Caputo
- Department of Epidemiology, Helmholtz Centre for Infection Research, Brunswick, Germany.,PhD Programme "Epidemiology," Brunswick, Germany
| | - Heike Raupach-Rosin
- Department of Epidemiology, Helmholtz Centre for Infection Research, Brunswick, Germany
| | - André Karch
- Department of Epidemiology, Helmholtz Centre for Infection Research, Brunswick, Germany.,German Centre for Infection Research (DZIF), Site Brunschwick-Hannover, Brunswick, Germany.,Institute for Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Michael Borte
- Children's Hospital, Municipal Hospital St. Georg Leipzig, Academic Teaching Hospital of the University of Leipzig, Leipzig, Germany
| | - Irina Lehmann
- Department of Environmental Immunology, Core Facility Studies, Helmholtz Centre for Environmental Research- UFZ, Leipzig, Germany.,Charitè - Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | | | - Marie Standl
- Helmholtz Zentrum München- German Research Center for Environmental Health, Institute of Epidemiology, Munich, Germany
| | - Joachim Heinrich
- Helmholtz Zentrum München- German Research Center for Environmental Health, Institute of Epidemiology, Munich, Germany.,Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, University Hospital of Munich, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Rafael T Mikolajczyk
- German Centre for Infection Research (DZIF), Site Brunschwick-Hannover, Brunswick, Germany.,Institute for Medical Epidemiology, Biometry, and Informatics (IMEBI), Medical Faculty of the Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
22
|
Stienstra R, Netea MG. Firing Up Glycolysis: BCG Vaccination Effects on Type 1 Diabetes Mellitus. Trends Endocrinol Metab 2018; 29:813-814. [PMID: 30327169 DOI: 10.1016/j.tem.2018.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/02/2018] [Indexed: 11/29/2022]
Abstract
In addition to the impact of Bacillus Calmette-Guerin (BCG) vaccination on antimicrobial host defence, a novel study reveals beneficial effects on glycaemic control in patients with long-standing type 1 diabetes mellitus (T1DM). These effects are ascribed to an accelerated glucose consumption in immune cells due to increased glycolysis and reduced oxidative phosphorylation.
Collapse
Affiliation(s)
- Rinke Stienstra
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany.
| |
Collapse
|
23
|
Abstract
Coinfections involving viruses are being recognized to influence the disease pattern that occurs relative to that with single infection. Classically, we usually think of a clinical syndrome as the consequence of infection by a single virus that is isolated from clinical specimens. However, this biased laboratory approach omits detection of additional agents that could be contributing to the clinical outcome, including novel agents not usually considered pathogens. The presence of an additional agent may also interfere with the targeted isolation of a known virus. Viral interference, a phenomenon where one virus competitively suppresses replication of other coinfecting viruses, is the most common outcome of viral coinfections. In addition, coinfections can modulate virus virulence and cell death, thereby altering disease severity and epidemiology. Immunity to primary virus infection can also modulate immune responses to subsequent secondary infections. In this review, various virological mechanisms that determine viral persistence/exclusion during coinfections are discussed, and insights into the isolation/detection of multiple viruses are provided. We also discuss features of heterologous infections that impact the pattern of immune responsiveness that develops.
Collapse
|
24
|
The Regulation of Inflammation by Innate and Adaptive Lymphocytes. J Immunol Res 2018; 2018:1467538. [PMID: 29992170 PMCID: PMC6016164 DOI: 10.1155/2018/1467538] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/16/2018] [Indexed: 02/08/2023] Open
Abstract
Inflammation plays an essential role in the control of pathogens and in shaping the ensuing adaptive immune responses. Traditionally, innate immunity has been described as a rapid response triggered through generic and nonspecific means that by definition lacks the ability to remember. Recently, it has become clear that some innate immune cells are epigenetically reprogrammed or “imprinted” by past experiences. These “trained” innate immune cells display altered inflammatory responses upon subsequent pathogen encounter. Remembrance of past pathogen encounters has classically been attributed to cohorts of antigen-specific memory T and B cells following the resolution of infection. During recall responses, memory T and B cells quickly respond by proliferating, producing effector cytokines, and performing various effector functions. An often-overlooked effector function of memory CD4 and CD8 T cells is the promotion of an inflammatory milieu at the initial site of infection that mirrors the primary encounter. This memory-conditioned inflammatory response, in conjunction with other secondary effector T cell functions, results in better control and more rapid resolution of both infection and the associated tissue pathology. Recent advancements in our understanding of inflammatory triggers, imprinting of the innate immune responses, and the role of T cell memory in regulating inflammation are discussed.
Collapse
|
25
|
Sørup S, Jensen AKG, Aaby P, Benn CS. Revaccination With Measles-Mumps-Rubella Vaccine and Infectious Disease Morbidity: A Danish Register-based Cohort Study. Clin Infect Dis 2018; 68:282-290. [DOI: 10.1093/cid/ciy433] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 05/24/2018] [Indexed: 12/28/2022] Open
Affiliation(s)
- Signe Sørup
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen
- Department of Clinical Epidemiology, Aarhus University
| | - Aksel K G Jensen
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen
- Section of Biostatistics, University of Copenhagen, Denmark
| | - Peter Aaby
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen
- Bandim Health Project, Indepth Network, Bissau, Guinea-Bissau
| | - Christine S Benn
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen
- Odense Patient Data Explorative Network, Odense University Hospital/Department of Clinical Research, University of Southern Denmark
| |
Collapse
|
26
|
Glanz JM, Newcomer SR, Daley MF, DeStefano F, Groom HC, Jackson ML, Lewin BJ, McCarthy NL, McClure DL, Narwaney KJ, Nordin JD, Zerbo O. Association Between Estimated Cumulative Vaccine Antigen Exposure Through the First 23 Months of Life and Non-Vaccine-Targeted Infections From 24 Through 47 Months of Age. JAMA 2018; 319:906-913. [PMID: 29509866 PMCID: PMC5885913 DOI: 10.1001/jama.2018.0708] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
IMPORTANCE Some parents are concerned that multiple vaccines in early childhood could weaken their child's immune system. Biological data suggest that increased vaccine antigen exposure could increase the risk for infections not targeted by vaccines. OBJECTIVE To examine estimated cumulative vaccine antigen exposure through the first 23 months of life in children with and without non-vaccine-targeted infections from 24 through 47 months of age. DESIGN, SETTING, AND PARTICIPANTS A nested case-control study was conducted in 6 US health care organizations participating in the Vaccine Safety Datalink. Cases were identified by International Classification of Diseases codes for infectious diseases in the emergency department and inpatient medical settings and then validated by medical record review. Cases of non-vaccine-targeted infection were matched to controls by age, sex, health care organization site, and chronic disease status. Participants were children ages 24 through 47 months, born between January 1, 2003, and September 31, 2013, followed up until December 31, 2015. EXPOSURES Cumulative vaccine antigen exposure, estimated by summing the number of antigens in each vaccine dose received from birth through age 23 months. MAIN OUTCOMES AND MEASURES Non-vaccine-targeted infections, including upper and lower respiratory infections and gastrointestinal infections, from 24 through 47 months of age, and the association between these infections and estimated cumulative vaccine exposure from birth through 23 months. Conditional logistic regression was used to estimate matched odds ratios representing the odds of non-vaccine-targeted infections for every 30-unit increase in estimated cumulative number of antigens received. RESULTS Among the 944 patients (193 cases and 751 controls), the mean (SD) age was 32.5 (6.3) months, 422 (45%) were female, and 61 (7%) had a complex chronic condition. Through the first 23 months, the estimated mean (SD) cumulative vaccine antigen exposure was 240.6 (48.3) for cases and 242.9 (51.1) for controls. The between-group difference for estimated cumulative antigen exposure was -2.3 (95% CI, -10.1 to 5.4; P = .55). Among children with vs without non-vaccine-targeted infections from 24 through 47 months of age, the matched odds ratio for estimated cumulative antigen exposure through age 23 months was not significant (matched odds ratio, 0.94; 95% CI, 0.84 to 1.07). CONCLUSIONS AND RELEVANCE Among children from 24 through 47 months of age with emergency department and inpatient visits for infectious diseases not targeted by vaccines, compared with children without such visits, there was no significant difference in estimated cumulative vaccine antigen exposure through the first 23 months of life.
Collapse
Affiliation(s)
- Jason M Glanz
- Institute for Health Research, Kaiser Permanente Colorado, Denver
- Department of Epidemiology, Colorado School of Public Health, Aurora
| | | | - Matthew F Daley
- Institute for Health Research, Kaiser Permanente Colorado, Denver
| | - Frank DeStefano
- Immunization Safety Office (Vaccine Safety Datalink), Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Holly C Groom
- Kaiser Permanente Center for Health Research, Northwest Kaiser Permanente, Portland, Oregon
| | - Michael L Jackson
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle
| | - Bruno J Lewin
- Kaiser Permanente Department of Research and Evaluation, Kaiser Permanente of Southern California, Pasadena
| | - Natalie L McCarthy
- Immunization Safety Office (Vaccine Safety Datalink), Centers for Disease Control and Prevention, Atlanta, Georgia
| | - David L McClure
- Marshfield Clinic Research Foundation, Marshfield, Wisconsin
| | - Komal J Narwaney
- Institute for Health Research, Kaiser Permanente Colorado, Denver
| | | | - Ousseny Zerbo
- Kaiser Permanente Division of Research, Kaiser Permanente of Northern California, Oakland
| |
Collapse
|
27
|
Kharchenko EP. OCCURRENCE OF SMALL HOMOLOGOUS AND COMPLEMENTARY FRAGMENTS IN HUMAN VIRUS GENOMES AND THEIR POSSIBLE ROLE. RUSSIAN JOURNAL OF INFECTION AND IMMUNITY 2018. [DOI: 10.15789/2220-7619-2017-4-393-404] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
With computer analysis occurrence of small homologous and complementary fragments (21 nucleotides in length) has been studied in genomes of 14 human viruses causing most dangerous infections. The sample includes viruses with (+) and (–) single stranded RNA and DNA-containing hepatitis A virus. Analysis of occurrence of homologous sequences has shown the existence two extreme situations. On the one hand, the same virus contains homologous sequences to almost all other viruses (for example, Ebola virus, severe acute respiratory syndrome-related coronavirus, and mumps virus), and numerous homologous sequences to the same other virus (especially in severe acute respiratory syndrome-related coronavirus to Dengue virus and in Ebola virus to poliovirus). On the other hand, there are rare occurrence and not numerous homologous sequences in genomes of other viruses (rubella virus, hepatitis A virus, and hepatitis B virus). Similar situation exists for occurrence of complementary sequences. Rubella virus, the genome of which has the high content of guanine and cytosine, has no complementary sequences to almost all other viruses. Most viruses have moderate level of occurrence for homologous and complementary sequences. Autocomplementary sequences are numerous in most viruses and one may suggest that the genome of single stranded RNA viruses has branched secondary structure. In addition to possible role in recombination among strains autocomplementary sequences could be regulators of translation rate of virus proteins and determine its optimal proportion in virion assembly with genome and mRNA folding. Occurrence of small homologous and complementary sequences in RNA- and DNA-containing viruses may be the result of multiple recombinations in the past and the present and determine their adaptation and variability. Recombination may take place in coinfection of human and/or common hosts. Inclusion of homologous and complementary sequences into genome could not only renew viruses but also serve as memory of existence of a competitor for host and means of counteraction against a competitor in coinfection being an analogy of the bacterial CRISPR/Cas system.
Collapse
|
28
|
Antunes DA, Rigo MM, Freitas MV, Mendes MFA, Sinigaglia M, Lizée G, Kavraki LE, Selin LK, Cornberg M, Vieira GF. Interpreting T-Cell Cross-reactivity through Structure: Implications for TCR-Based Cancer Immunotherapy. Front Immunol 2017; 8:1210. [PMID: 29046675 PMCID: PMC5632759 DOI: 10.3389/fimmu.2017.01210] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/12/2017] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy has become one of the most promising avenues for cancer treatment, making use of the patient’s own immune system to eliminate cancer cells. Clinical trials with T-cell-based immunotherapies have shown dramatic tumor regressions, being effective in multiple cancer types and for many different patients. Unfortunately, this progress was tempered by reports of serious (even fatal) side effects. Such therapies rely on the use of cytotoxic T-cell lymphocytes, an essential part of the adaptive immune system. Cytotoxic T-cells are regularly involved in surveillance and are capable of both eliminating diseased cells and generating protective immunological memory. The specificity of a given T-cell is determined through the structural interaction between the T-cell receptor (TCR) and a peptide-loaded major histocompatibility complex (MHC); i.e., an intracellular peptide–ligand displayed at the cell surface by an MHC molecule. However, a given TCR can recognize different peptide–MHC (pMHC) complexes, which can sometimes trigger an unwanted response that is referred to as T-cell cross-reactivity. This has become a major safety issue in TCR-based immunotherapies, following reports of melanoma-specific T-cells causing cytotoxic damage to healthy tissues (e.g., heart and nervous system). T-cell cross-reactivity has been extensively studied in the context of viral immunology and tissue transplantation. Growing evidence suggests that it is largely driven by structural similarities of seemingly unrelated pMHC complexes. Here, we review recent reports about the existence of pMHC “hot-spots” for cross-reactivity and propose the existence of a TCR interaction profile (i.e., a refinement of a more general TCR footprint in which some amino acid residues are more important than others in triggering T-cell cross-reactivity). We also make use of available structural data and pMHC models to interpret previously reported cross-reactivity patterns among virus-derived peptides. Our study provides further evidence that structural analyses of pMHC complexes can be used to assess the intrinsic likelihood of cross-reactivity among peptide-targets. Furthermore, we hypothesize that some apparent inconsistencies in reported cross-reactivities, such as a preferential directionality, might also be driven by particular structural features of the targeted pMHC complex. Finally, we explain why TCR-based immunotherapy provides a special context in which meaningful T-cell cross-reactivity predictions can be made.
Collapse
Affiliation(s)
- Dinler A Antunes
- Núcleo de Bioinformática do Laboratório de Imunogenética (NBLI), Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Kavraki Lab, Department of Computer Science, Rice University, Houston, TX, United States
| | - Maurício M Rigo
- Núcleo de Bioinformática do Laboratório de Imunogenética (NBLI), Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Laboratório de Imunologia Celular e Molecular, Instituto de Pesquisas Biomédicas (IPB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Martiela V Freitas
- Núcleo de Bioinformática do Laboratório de Imunogenética (NBLI), Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Marcus F A Mendes
- Núcleo de Bioinformática do Laboratório de Imunogenética (NBLI), Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Marialva Sinigaglia
- Núcleo de Bioinformática do Laboratório de Imunogenética (NBLI), Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Gregory Lizée
- Lizée Lab, Department of Melanoma Medical Oncology - Research, The University of Texas M. D. Anderson Cancer Center, Houston, TX, United States
| | - Lydia E Kavraki
- Kavraki Lab, Department of Computer Science, Rice University, Houston, TX, United States
| | - Liisa K Selin
- Selin Lab, Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Markus Cornberg
- Cornberg Lab, Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.,German Center for Infection Research (DZIF), Partner-Site Hannover-Braunschweig, Hannover, Germany
| | - Gustavo F Vieira
- Núcleo de Bioinformática do Laboratório de Imunogenética (NBLI), Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Saúde e Desenvolvimento Humano, Universidade La Salle, Porto Alegre, Brazil
| |
Collapse
|
29
|
Knobel DL, Arega S, Reininghaus B, Simpson GJG, Gessner BD, Stryhn H, Conan A. Rabies vaccine is associated with decreased all-cause mortality in dogs. Vaccine 2017; 35:3844-3849. [PMID: 28602607 DOI: 10.1016/j.vaccine.2017.05.095] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 01/23/2023]
Abstract
Evidence suggests that rabies vaccine may have non-specific protective effects in animals and children. We analyzed four years of data (2012-2015) from an observational study of the health and demographics of a population of owned, free-roaming dogs in a low-income community in South Africa. The objective of this analysis was to assess the association between rabies vaccine and all-cause mortality in dogs, stratified by age group (0-3months, 4-11months, and 12months and older), and controlling for the effects of sex and number of dogs in the residence. Rabies vaccination reduced the risk of death from any cause by 56% (95% CI=16-77%) in dogs aged 0-3months, by 44% (95% CI=21-60%) in dogs aged 4-11months and by 16% (95% CI=0-29%) in dogs aged 12months and older. We hypothesize that the protective association between rabies vaccination status and all-cause mortality is due to a protective effect of rabies vaccine against diseases other than rabies. Existence of a strong non-specific protective effect of rabies vaccine on mortality in dogs would have implications for the design of dog rabies control programs that aim to eliminate dog-mediated human rabies cases. Further, we propose that owned domestic dogs in high mortality settings provide a useful animal model to better understand any non-specific protective effect of rabies vaccine in children, due to dogs' high numbers, high morbidity and mortality rates, relatively short lifespan, exposure to a variety of infectious and parasitic diseases, and shared environment with people.
Collapse
Affiliation(s)
- Darryn L Knobel
- Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis.
| | - Sintayehu Arega
- Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| | - Bjorn Reininghaus
- Mpumalanga Veterinary Services, Department of Agriculture, Rural Development, Land and Environmental Affairs, Thulamahashe, South Africa
| | - Gregory J G Simpson
- Department of Production Animal Studies, University of Pretoria, Pretoria, South Africa
| | | | - Henrik Stryhn
- Centre for Veterinary Epidemiological Research, University of Prince Edward Island, Charlottetown, Canada
| | - Anne Conan
- Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| |
Collapse
|
30
|
Noho-Konteh F, Adetifa JU, Cox M, Hossin S, Reynolds J, Le MT, Sanyang LC, Drammeh A, Plebanski M, Forster T, Dickinson P, Ghazal P, Whittle H, Rowland-Jones SL, Sutherland JS, Flanagan KL. Sex-Differential Non-Vaccine-Specific Immunological Effects of Diphtheria-Tetanus-Pertussis and Measles Vaccination. Clin Infect Dis 2016; 63:1213-1226. [PMID: 27436422 DOI: 10.1093/cid/ciw492] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/07/2016] [Indexed: 12/31/2022] Open
Abstract
Vaccines can have nontargeted heterologous effects that manifest as increased protection against nonvaccine infections, as described for measles vaccine (MV), or increased susceptibility to infections and death, as described following diphtheria-tetanus-whole cell pertussis (DTP) vaccination. The mechanisms are unknown, and high-quality immunological studies are lacking. This study was designed to investigate the heterologous effects of MV and DTP in 302 Gambian infants. The results support a sex-differential immunosuppressive effect of DTP on innate proinflammatory responses and T-cell immunity. Males but not females receiving MV had enhanced proinflammatory innate responses. The results point to modified signaling via Toll-like receptor 4 (TLR4) as a possible mechanism for the effects on innate immunity. When both vaccines were administered together, purified protein derivative responses were enhanced in females but downregulated in males. Collectively, these data indicate immunological effects that could account for heterologous effects of MV and DTP, to take forward into prospective trials.
Collapse
Affiliation(s)
- Fatou Noho-Konteh
- Infant Immunology Group, Vaccines and Immunity Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Jane U Adetifa
- Infant Immunology Group, Vaccines and Immunity Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Momodou Cox
- Infant Immunology Group, Vaccines and Immunity Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Safayet Hossin
- Infant Immunology Group, Vaccines and Immunity Theme, Medical Research Council Unit, Fajara, The Gambia
| | - John Reynolds
- Biostatistics Consulting Platform, Faculty of Medicine, Nursing and Health Sciences
| | - My Thanh Le
- Infant Immunology Group, Vaccines and Immunity Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Lady Chilel Sanyang
- Infant Immunology Group, Vaccines and Immunity Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Abdoulie Drammeh
- Infant Immunology Group, Vaccines and Immunity Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Magdalena Plebanski
- Department of Immunology and Pathology Monash Institute of Medical Engineering, Monash University, Prahran, Victoria, Australia
| | - Thorsten Forster
- Division of Infection and Pathway Medicine, University of Edinburgh
| | - Paul Dickinson
- Division of Infection and Pathway Medicine, University of Edinburgh
| | - Peter Ghazal
- Division of Infection and Pathway Medicine, University of Edinburgh
| | - Hilton Whittle
- Infant Immunology Group, Vaccines and Immunity Theme, Medical Research Council Unit, Fajara, The Gambia London School of Hygiene and Tropical Medicine
| | - Sarah L Rowland-Jones
- Infant Immunology Group, Vaccines and Immunity Theme, Medical Research Council Unit, Fajara, The Gambia Nuffield Department of Medicine, University of Oxford, United Kingdom
| | - Jayne S Sutherland
- Infant Immunology Group, Vaccines and Immunity Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Katie L Flanagan
- Infant Immunology Group, Vaccines and Immunity Theme, Medical Research Council Unit, Fajara, The Gambia Department of Immunology and Pathology
| |
Collapse
|
31
|
Flanagan KL, Plebanski M. Sex-differential heterologous (non-specific) effects of vaccines: an emerging public health issue that needs to be understood and exploited. Expert Rev Vaccines 2016; 16:5-13. [DOI: 10.1080/14760584.2016.1203260] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Katie L. Flanagan
- Vaccine and Infectious Diseases Laboratory, Department of Immunology and Pathology, Monash University, Prahran, Australia
| | - Magdalena Plebanski
- Vaccine and Infectious Diseases Laboratory, Department of Immunology and Pathology, Monash University, Prahran, Australia
- Monash Institute of Medical Engineering, Monash University, Prahran, Australia
| |
Collapse
|
32
|
Unravelling the nature of non-specific effects of vaccines-A challenge for innate immunologists. Semin Immunol 2016; 28:377-83. [PMID: 27354354 DOI: 10.1016/j.smim.2016.05.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 01/29/2023]
Abstract
Epidemiological observations have shown that vaccines can influence morbidity and mortality more than can be ascribed to target-disease immunity. A growing number of immunological studies have helped identify possible biological mechanisms to explain these so-called nonspecific effects (NSE) of vaccines, including heterologous T-cell reactivity and innate immune memory or 'trained innate immunity', which involves epigenetic reprogramming of innate immune cells. Here, we review the epidemiological evidence for NSE as well as human, animal and in vitro immunological data that could explain these NSE, and discuss priorities for future epidemiologic and immunologic studies to further unravel the biology and optimize the benefits of current and new vaccines.
Collapse
|
33
|
Flanagan KL, Wilson KL, Plebanski M. Polymorphism in liver-stage malaria vaccine candidate proteins: immune evasion and implications for vaccine design. Expert Rev Vaccines 2015; 15:389-99. [PMID: 26610026 DOI: 10.1586/14760584.2016.1125785] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pre-erythrocytic stage of infection by malaria parasites represents a key target for vaccines that aim to eradicate malaria. Two important broad immune evasion strategies that can interfere with vaccine efficacy include the induction of dendritic cell (DC) dysfunction and regulatory T cells (Tregs) by blood-stage malaria parasites, leading to inefficient priming of T cells targeting liver-stage infections. The parasite also uses 'surgical strike' strategies, whereby polymorphism in pre-erythrocytic antigens can interfere with host immunity. Specifically, we review how even single amino acid changes in T cell epitopes can lead to loss of binding to major histocompatibility complex (MHC), lack of cross-reactivity, or antagonism and immune interference, where simultaneous or sequential stimulation with related variants of the same T cell epitope can cause T cell anergy or the conversion of effector to immunosuppressive T cell phenotypes.
Collapse
Affiliation(s)
- Katie L Flanagan
- a Vaccine and Infectious Diseases Laboratory, Department of Immunology , Monash University , Melbourne , VIC , Australia
| | - Kirsty L Wilson
- a Vaccine and Infectious Diseases Laboratory, Department of Immunology , Monash University , Melbourne , VIC , Australia
| | - Magdalena Plebanski
- a Vaccine and Infectious Diseases Laboratory, Department of Immunology , Monash University , Melbourne , VIC , Australia
| |
Collapse
|
34
|
Blok BA, Arts RJW, van Crevel R, Benn CS, Netea MG. Trained innate immunity as underlying mechanism for the long-term, nonspecific effects of vaccines. J Leukoc Biol 2015; 98:347-56. [DOI: 10.1189/jlb.5ri0315-096r] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/19/2015] [Indexed: 12/31/2022] Open
|
35
|
Flanagan KL. Vaccines have sex differential non-targeted heterologous effects: a new dawn in vaccine research. Trans R Soc Trop Med Hyg 2015; 109:1-2. [DOI: 10.1093/trstmh/tru188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|