1
|
Hesnard L, Thériault C, Cahuzac M, Durette C, Vincent K, Hardy MP, Lanoix J, Lavallée GO, Humeau J, Thibault P, Perreault C. Immunogenicity of Non-Mutated Ovarian Cancer-Specific Antigens. Curr Oncol 2024; 31:3099-3121. [PMID: 38920720 PMCID: PMC11203340 DOI: 10.3390/curroncol31060236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
Epithelial ovarian cancer (EOC) has not significantly benefited from advances in immunotherapy, mainly because of the lack of well-defined actionable antigen targets. Using proteogenomic analyses of primary EOC tumors, we previously identified 91 aberrantly expressed tumor-specific antigens (TSAs) originating from unmutated genomic sequences. Most of these TSAs derive from non-exonic regions, and their expression results from cancer-specific epigenetic changes. The present study aimed to evaluate the immunogenicity of 48 TSAs selected according to two criteria: presentation by highly prevalent HLA allotypes and expression in a significant fraction of EOC tumors. Using targeted mass spectrometry analyses, we found that pulsing with synthetic TSA peptides leads to a high-level presentation on dendritic cells. TSA abundance correlated with the predicted binding affinity to the HLA allotype. We stimulated naïve CD8 T cells from healthy blood donors with TSA-pulsed dendritic cells and assessed their expansion with two assays: MHC-peptide tetramer staining and TCR Vβ CDR3 sequencing. We report that these TSAs can expand sizeable populations of CD8 T cells and, therefore, represent attractive targets for EOC immunotherapy.
Collapse
Affiliation(s)
- Leslie Hesnard
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada; (L.H.); (C.T.); (M.C.); (C.D.); (K.V.); (M.-P.H.); (J.L.); (G.O.L.); (J.H.); (P.T.)
| | - Catherine Thériault
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada; (L.H.); (C.T.); (M.C.); (C.D.); (K.V.); (M.-P.H.); (J.L.); (G.O.L.); (J.H.); (P.T.)
| | - Maxime Cahuzac
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada; (L.H.); (C.T.); (M.C.); (C.D.); (K.V.); (M.-P.H.); (J.L.); (G.O.L.); (J.H.); (P.T.)
| | - Chantal Durette
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada; (L.H.); (C.T.); (M.C.); (C.D.); (K.V.); (M.-P.H.); (J.L.); (G.O.L.); (J.H.); (P.T.)
| | - Krystel Vincent
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada; (L.H.); (C.T.); (M.C.); (C.D.); (K.V.); (M.-P.H.); (J.L.); (G.O.L.); (J.H.); (P.T.)
| | - Marie-Pierre Hardy
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada; (L.H.); (C.T.); (M.C.); (C.D.); (K.V.); (M.-P.H.); (J.L.); (G.O.L.); (J.H.); (P.T.)
| | - Joël Lanoix
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada; (L.H.); (C.T.); (M.C.); (C.D.); (K.V.); (M.-P.H.); (J.L.); (G.O.L.); (J.H.); (P.T.)
| | - Gabriel Ouellet Lavallée
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada; (L.H.); (C.T.); (M.C.); (C.D.); (K.V.); (M.-P.H.); (J.L.); (G.O.L.); (J.H.); (P.T.)
| | - Juliette Humeau
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada; (L.H.); (C.T.); (M.C.); (C.D.); (K.V.); (M.-P.H.); (J.L.); (G.O.L.); (J.H.); (P.T.)
| | - Pierre Thibault
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada; (L.H.); (C.T.); (M.C.); (C.D.); (K.V.); (M.-P.H.); (J.L.); (G.O.L.); (J.H.); (P.T.)
- Department of Chemistry, University of Montreal, Montreal, QC H2V 0B3, Canada
| | - Claude Perreault
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada; (L.H.); (C.T.); (M.C.); (C.D.); (K.V.); (M.-P.H.); (J.L.); (G.O.L.); (J.H.); (P.T.)
- Department of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| |
Collapse
|
2
|
Schaerlaekens S, Jacobs L, Stobbelaar K, Cos P, Delputte P. All Eyes on the Prefusion-Stabilized F Construct, but Are We Missing the Potential of Alternative Targets for Respiratory Syncytial Virus Vaccine Design? Vaccines (Basel) 2024; 12:97. [PMID: 38250910 PMCID: PMC10819635 DOI: 10.3390/vaccines12010097] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/12/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
Respiratory Syncytial Virus (RSV) poses a significant global health concern as a major cause of lower respiratory tract infections (LRTIs). Over the last few years, substantial efforts have been directed towards developing vaccines and therapeutics to combat RSV, leading to a diverse landscape of vaccine candidates. Notably, two vaccines targeting the elderly and the first maternal vaccine have recently been approved. The majority of the vaccines and vaccine candidates rely solely on a prefusion-stabilized conformation known for its highly neutralizing epitopes. Although, so far, this antigen design appears to be successful for the elderly, our current understanding remains incomplete, requiring further improvement and refinement in this field. Pediatric vaccines still have a long journey ahead, and we must ensure that vaccines currently entering the market do not lose efficacy due to the emergence of mutations in RSV's circulating strains. This review will provide an overview of the current status of vaccine designs and what to focus on in the future. Further research into antigen design is essential, including the exploration of the potential of alternative RSV proteins to address these challenges and pave the way for the development of novel and effective vaccines, especially in the pediatric population.
Collapse
Affiliation(s)
- Sofie Schaerlaekens
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (S.S.); (L.J.); (K.S.); (P.C.)
| | - Lotte Jacobs
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (S.S.); (L.J.); (K.S.); (P.C.)
| | - Kim Stobbelaar
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (S.S.); (L.J.); (K.S.); (P.C.)
- Pediatrics Department, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (S.S.); (L.J.); (K.S.); (P.C.)
- Infla-Med Centre of Excellence, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium
| | - Peter Delputte
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (S.S.); (L.J.); (K.S.); (P.C.)
- Infla-Med Centre of Excellence, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium
| |
Collapse
|
3
|
Šantak M, Matić Z. The Role of Nucleoprotein in Immunity to Human Negative-Stranded RNA Viruses—Not Just Another Brick in the Viral Nucleocapsid. Viruses 2022; 14:v14030521. [PMID: 35336928 PMCID: PMC8955406 DOI: 10.3390/v14030521] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 12/21/2022] Open
Abstract
Negative-stranded RNA viruses (NSVs) are important human pathogens, including emerging and reemerging viruses that cause respiratory, hemorrhagic and other severe illnesses. Vaccine design traditionally relies on the viral surface glycoproteins. However, surface glycoproteins rarely elicit effective long-term immunity due to high variability. Therefore, an alternative approach is to include conserved structural proteins such as nucleoprotein (NP). NP is engaged in myriad processes in the viral life cycle: coating and protection of viral RNA, regulation of transcription/replication processes and induction of immunosuppression of the host. A broad heterosubtypic T-cellular protection was ascribed very early to this protein. In contrast, the understanding of the humoral immunity to NP is very limited in spite of the high titer of non-neutralizing NP-specific antibodies raised upon natural infection or immunization. In this review, the data with important implications for the understanding of the role of NP in the immune response to human NSVs are revisited. Major implications of the elicited T-cell immune responses to NP are evaluated, and the possible multiple mechanisms of the neglected humoral response to NP are discussed. The intention of this review is to remind that NP is a very promising target for the development of future vaccines.
Collapse
|
4
|
Green C, Sande C, de Lara C, Thompson A, Silva-Reyes L, Napolitano F, Pierantoni A, Capone S, Vitelli A, Klenerman P, Pollard A. Humoral and cellular immunity to RSV in infants, children and adults. Vaccine 2018; 36:6183-6190. [DOI: 10.1016/j.vaccine.2018.08.056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/08/2018] [Accepted: 08/21/2018] [Indexed: 12/11/2022]
|
5
|
Burbulla D, Günther PS, Peper JK, Jahn G, Dennehy KM. Human CD8(+) T Cells Target Multiple Epitopes in Respiratory Syncytial Virus Polymerase. Viral Immunol 2016; 29:307-14. [PMID: 27070377 DOI: 10.1089/vim.2015.0091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection is a serious health problem in young children, immunocompromised patients, and the elderly. The development of novel prevention strategies, such as a vaccine to RSV, is a high priority. One strategy is to design a peptide-based vaccine that activates appropriate CD8(+) T-cell responses. However, this approach is limited by the low number of RSV peptide epitopes defined to date that activate CD8(+) T cells. We aimed to identify peptide epitopes that are presented by common human leukocyte antigen types (HLA-A*01, -A*02, and -B*07). We identify one novel HLA-A*02-restricted and two novel HLA-A*01-restricted peptide epitopes from RSV polymerase. Peptide-HLA multimer staining of specific T cells from healthy donor peripheral blood mononuclear cell, the memory phenotype of such peptide-specific T cells ex vivo, and functional IFNγ responses in short-term stimulation assays suggest that these peptides are recognized during RSV infection. Such peptides are candidates for inclusion into a peptide-based RSV vaccine designed to stimulate defined CD8(+) T-cell responses.
Collapse
Affiliation(s)
- Daniel Burbulla
- 1 Institute for Medical Virology, University Hospital Tübingen , Tübingen, Germany
| | - Patrick S Günther
- 1 Institute for Medical Virology, University Hospital Tübingen , Tübingen, Germany
| | - Janet K Peper
- 2 Department of Immunology, University of Tübingen , Tübingen, Germany
| | - Gerhard Jahn
- 1 Institute for Medical Virology, University Hospital Tübingen , Tübingen, Germany
| | - Kevin M Dennehy
- 1 Institute for Medical Virology, University Hospital Tübingen , Tübingen, Germany
| |
Collapse
|
6
|
Jozwik A, Habibi MS, Paras A, Zhu J, Guvenel A, Dhariwal J, Almond M, Wong EHC, Sykes A, Maybeno M, Del Rosario J, Trujillo-Torralbo MB, Mallia P, Sidney J, Peters B, Kon OM, Sette A, Johnston SL, Openshaw PJ, Chiu C. RSV-specific airway resident memory CD8+ T cells and differential disease severity after experimental human infection. Nat Commun 2015; 6:10224. [PMID: 26687547 PMCID: PMC4703893 DOI: 10.1038/ncomms10224] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 11/16/2015] [Indexed: 12/30/2022] Open
Abstract
In animal models, resident memory CD8+ T (Trm) cells assist in respiratory virus elimination but their importance in man has not been determined. Here, using experimental human respiratory syncytial virus (RSV) infection, we investigate systemic and local virus-specific CD8+ T-cell responses in adult volunteers. Having defined the immunodominance hierarchy, we analyse phenotype and function longitudinally in blood and by serial bronchoscopy. Despite rapid clinical recovery, we note surprisingly extensive lower airway inflammation with persistent viral antigen and cellular infiltrates. Pulmonary virus-specific CD8+ T cells display a CD69+CD103+ Trm phenotype and accumulate to strikingly high frequencies into convalescence without continued proliferation. While these have a more highly differentiated phenotype, they express fewer cytotoxicity markers than in blood. Nevertheless, their abundance before infection correlates with reduced symptoms and viral load, implying that CD8+ Trm cells in the human lung can confer protection against severe respiratory viral disease when humoral immunity is overcome.
Collapse
Affiliation(s)
- Agnieszka Jozwik
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | | | - Allan Paras
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Jie Zhu
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Aleks Guvenel
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Jaideep Dhariwal
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Mark Almond
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Ernie H. C. Wong
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Annemarie Sykes
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Matthew Maybeno
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute of Allergy and Immunology, 9420 Athena Circle, La Jolla, California 92037, USA
| | - Jerico Del Rosario
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | | | - Patrick Mallia
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - John Sidney
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute of Allergy and Immunology, 9420 Athena Circle, La Jolla, California 92037, USA
| | - Bjoern Peters
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute of Allergy and Immunology, 9420 Athena Circle, La Jolla, California 92037, USA
| | - Onn Min Kon
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Alessandro Sette
- Centre for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute of Allergy and Immunology, 9420 Athena Circle, La Jolla, California 92037, USA
| | | | - Peter J. Openshaw
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| | - Christopher Chiu
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
| |
Collapse
|
7
|
Jaberolansar N, Toth I, Young PR, Skwarczynski M. Recent advances in the development of subunit-based RSV vaccines. Expert Rev Vaccines 2015; 15:53-68. [PMID: 26506139 DOI: 10.1586/14760584.2016.1105134] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections causing pneumonia and bronchiolitis in infants. RSV also causes serious illness in elderly populations, immunocompromised patients and individuals with pulmonary or cardiac problems. The significant morbidity and mortality associated with RSV infection have prompted interest in RSV vaccine development. In the 1960s, a formalin-inactivated vaccine trial failed to protect children, and indeed enhanced pathology when naturally infected later with RSV. Hence, an alternative approach to traditional killed virus vaccines, which can induce protective immunity without serious adverse events, is desired. Several strategies have been explored in attempts to produce effective vaccine candidates including gene-based and subunit vaccines. Subunit-based vaccine approaches have shown promising efficacy in animal studies and several have reached clinical trials. The current stage of development of subunit-based vaccines against RSV is reviewed in this article.
Collapse
Affiliation(s)
- Noushin Jaberolansar
- a School of Chemistry and Molecular Biosciences , The University of Queensland , St Lucia , Queensland , Australia
| | - Istvan Toth
- a School of Chemistry and Molecular Biosciences , The University of Queensland , St Lucia , Queensland , Australia.,b Institute for Molecular Bioscience , The University of Queensland , St Lucia , Queensland , Australia.,c School of Pharmacy , The University of Queensland , Woolloongabba , Queensland , Australia
| | - Paul R Young
- a School of Chemistry and Molecular Biosciences , The University of Queensland , St Lucia , Queensland , Australia.,b Institute for Molecular Bioscience , The University of Queensland , St Lucia , Queensland , Australia.,d Australian Infectious Diseases Research Centre , The University of Queensland , St Lucia , Queensland , Australia
| | - Mariusz Skwarczynski
- a School of Chemistry and Molecular Biosciences , The University of Queensland , St Lucia , Queensland , Australia
| |
Collapse
|
8
|
Green CA, Scarselli E, Sande CJ, Thompson AJ, de Lara CM, Taylor KS, Haworth K, Del Sorbo M, Angus B, Siani L, Di Marco S, Traboni C, Folgori A, Colloca S, Capone S, Vitelli A, Cortese R, Klenerman P, Nicosia A, Pollard AJ. Chimpanzee adenovirus- and MVA-vectored respiratory syncytial virus vaccine is safe and immunogenic in adults. Sci Transl Med 2015; 7:300ra126. [PMID: 26268313 PMCID: PMC4669850 DOI: 10.1126/scitranslmed.aac5745] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Respiratory syncytial virus (RSV) causes respiratory infection in annual epidemics, with infants and the elderly at particular risk of developing severe disease and death. However, despite its importance, no vaccine exists. The chimpanzee adenovirus, PanAd3-RSV, and modified vaccinia virus Ankara, MVA-RSV, are replication-defective viral vectors encoding the RSV fusion (F), nucleocapsid (N), and matrix (M2-1) proteins for the induction of humoral and cellular responses. We performed an open-label, dose escalation, phase 1 clinical trial in 42 healthy adults in which four different combinations of prime/boost vaccinations were investigated for safety and immunogenicity, including both intramuscular (IM) and intranasal (IN) administration of the adenovirus-vectored vaccine. The vaccines were safe and well tolerated, with the most common reported adverse events being mild injection site reactions. No vaccine-related serious adverse events occurred. RSV neutralizing antibody titers rose in response to IM prime with PanAd3-RSV and after IM boost for individuals primed by the IN route. Circulating anti-F immunoglobulin G (IgG) and IgA antibody-secreting cells (ASCs) were observed after the IM prime and IM boost. RSV-specific T cell responses were increased after the IM PanAd3-RSV prime and were most efficiently boosted by IM MVA-RSV. Interferon-γ (IFN-γ) secretion after boost was from both CD4(+) and CD8(+) T cells, without detectable T helper cell 2 (TH2) cytokines that have been previously associated with immune pathogenesis following exposure to RSV after the formalin-inactivated RSV vaccine. In conclusion, PanAd3-RSV and MVA-RSV are safe and immunogenic in healthy adults. These vaccine candidates warrant further clinical evaluation of efficacy to assess their potential to reduce the burden of RSV disease.
Collapse
Affiliation(s)
- Christopher A Green
- Oxford Vaccine Group, Department of Paediatrics, and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK.
| | - Elisa Scarselli
- ReiThera SRL (formerly Okairos SRL), Viale Città d'Europa 679, 00144 Rome, Italy
| | - Charles J Sande
- Oxford Vaccine Group, Department of Paediatrics, and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK
| | - Amber J Thompson
- Oxford Vaccine Group, Department of Paediatrics, and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK
| | - Catherine M de Lara
- Experimental Medicine Division, Nuffield Department of Medicine, Peter Medawar Building, University of Oxford, Oxford OX1 3SY, UK
| | - Kathryn S Taylor
- Oxford Vaccine Group, Department of Paediatrics, and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK
| | - Kathryn Haworth
- Oxford Vaccine Group, Department of Paediatrics, and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK
| | | | - Brian Angus
- Oxford Vaccine Group, Department of Paediatrics, and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK
| | - Loredana Siani
- ReiThera SRL (formerly Okairos SRL), Viale Città d'Europa 679, 00144 Rome, Italy
| | - Stefania Di Marco
- ReiThera SRL (formerly Okairos SRL), Viale Città d'Europa 679, 00144 Rome, Italy
| | - Cinzia Traboni
- ReiThera SRL (formerly Okairos SRL), Viale Città d'Europa 679, 00144 Rome, Italy
| | - Antonella Folgori
- ReiThera SRL (formerly Okairos SRL), Viale Città d'Europa 679, 00144 Rome, Italy
| | - Stefano Colloca
- ReiThera SRL (formerly Okairos SRL), Viale Città d'Europa 679, 00144 Rome, Italy
| | - Stefania Capone
- ReiThera SRL (formerly Okairos SRL), Viale Città d'Europa 679, 00144 Rome, Italy
| | - Alessandra Vitelli
- ReiThera SRL (formerly Okairos SRL), Viale Città d'Europa 679, 00144 Rome, Italy
| | | | - Paul Klenerman
- Experimental Medicine Division, Nuffield Department of Medicine, Peter Medawar Building, University of Oxford, Oxford OX1 3SY, UK
| | - Alfredo Nicosia
- ReiThera SRL (formerly Okairos SRL), Viale Città d'Europa 679, 00144 Rome, Italy. CEINGE, Via Gaetano Salvatore 486, 80145 Naples, Italy. Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK
| |
Collapse
|
9
|
Abstract
ABSTRACT Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection and hospitalization among infants. Despite the significant healthcare burden, there is no licensed RSV vaccine currently available. This problem is further exacerbated as a natural RSV infection fails to elicit the development of long-lived immunity. It is well established that RSV-specific antibodies play a critical role in mediating protection from severe disease. The CD8 T-cell response is critical for mediating virus clearance following an acute RSV infection. However, the relative contribution of memory CD8 T cells in providing protection against secondary RSV infections remains unclear. In addition, data from animal models indicate that memory CD8 T-cell responses can be pathogenic under certain conditions. Herein, we provide an overview of the CD8 T-cell response elicited by RSV infection and how our current knowledge may impact future studies and vaccine development.
Collapse
Affiliation(s)
- Cory J Knudson
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
10
|
Serologic cross-reactions between nucleocapsid proteins of human respiratory syncytial virus and human metapneumovirus. J Clin Microbiol 2015; 53:1609-15. [PMID: 25740767 DOI: 10.1128/jcm.03649-14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/23/2015] [Indexed: 02/08/2023] Open
Abstract
Human respiratory syncytial virus (hRSV) and human metapneumovirus (hMPV) share virologic and epidemiologic features and cause clinically similar respiratory illness predominantly in young children. In a previous study of acute febrile respiratory illness in Bangladesh, we tested paired serum specimens from 852 children presenting fever and cough for diagnostic increases in titers of antibody to hRSV and hMPV by enzyme immunoassay (EIA). Unexpectedly, of 93 serum pairs that showed a ≥ 4-fold increase in titers of antibody to hRSV, 24 (25.8%) showed a concurrent increase in titers of antibody to hMPV; of 91 pairs showing an increase to hMPV, 13 (14.3%) showed a concurrent increase to hRSV. We speculated that common antigens shared by these viruses explain this finding. Since the nucleocapsid (N) proteins of these viruses show the greatest sequence homology, we tested hyperimmune antisera prepared for each virus against baculovirus-expressed recombinant N (recN) proteins for potential cross-reactivity. The antisera were reciprocally reactive with both proteins. To localize common antigenic regions, we first expressed the carboxy domain of the hMPV N protein that was the most highly conserved region within the hRSV N protein. Although reciprocally reactive with antisera by Western blotting, this truncated protein did not react with hMPV IgG-positive human sera by EIA. Using 5 synthetic peptides that spanned the amino-terminal portion of the hMPV N protein, we identified a single peptide that was cross-reactive with human sera positive for either virus. Antiserum prepared for this peptide was reactive with recN proteins of both viruses, indicating that a common immunoreactive site exists in this region.
Collapse
|
11
|
Johnstone C, Lorente E, Barriga A, Barnea E, Infantes S, Lemonnier FA, David CS, Admon A, López D. The viral transcription group determines the HLA class I cellular immune response against human respiratory syncytial virus. Mol Cell Proteomics 2015; 14:893-904. [PMID: 25635267 DOI: 10.1074/mcp.m114.045401] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Indexed: 11/06/2022] Open
Abstract
The cytotoxic T-lymphocyte-mediated killing of virus-infected cells requires previous recognition of short viral antigenic peptides bound to human leukocyte antigen class I molecules that are exposed on the surface of infected cells. The cytotoxic T-lymphocyte response is critical for the clearance of human respiratory syncytial virus infection. In this study, naturally processed viral human leukocyte antigen class I ligands were identified with mass spectrometry analysis of complex human leukocyte antigen-bound peptide pools isolated from large amounts of human respiratory syncytial virus-infected cells. Acute antiviral T-cell response characterization showed that viral transcription determines both the immunoprevalence and immunodominance of the human leukocyte antigen class I response to human respiratory syncytial virus. These findings have clear implications for antiviral vaccine design.
Collapse
Affiliation(s)
- Carolina Johnstone
- From the ‡Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain
| | - Elena Lorente
- From the ‡Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain
| | - Alejandro Barriga
- From the ‡Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain
| | - Eilon Barnea
- §Department of Biology, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Susana Infantes
- From the ‡Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain
| | - François A Lemonnier
- ¶Unité d'Immunité Cellulaire Antivirale, Département d'Immunologie, Institut Pasteur, Paris Cedex 15, France
| | - Chella S David
- ‖Department of Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Arie Admon
- §Department of Biology, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Daniel López
- From the ‡Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain;
| |
Collapse
|
12
|
Kim YI, DeVincenzo JP, Jones BG, Rudraraju R, Harrison L, Meyers R, Cehelsky J, Alvarez R, Hurwitz JL. Respiratory syncytial virus human experimental infection model: provenance, production, and sequence of low-passaged memphis-37 challenge virus. PLoS One 2014; 9:e113100. [PMID: 25415360 PMCID: PMC4240712 DOI: 10.1371/journal.pone.0113100] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 10/19/2014] [Indexed: 01/11/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in children and is responsible for as many as 199,000 childhood deaths annually worldwide. To support the development of viral therapeutics and vaccines for RSV, a human adult experimental infection model has been established. In this report, we describe the provenance and sequence of RSV Memphis-37, the low-passage clinical isolate used for the model's reproducible, safe, experimental infections of healthy, adult volunteers. The predicted amino acid sequences for major proteins of Memphis-37 are compared to nine other RSV A and B amino acid sequences to examine sites of vaccine, therapeutic, and pathophysiologic interest. Human T- cell epitope sequences previously defined by in vitro studies were observed to be closely matched between Memphis-37 and the laboratory strain RSV A2. Memphis-37 sequences provide baseline data with which to assess: (i) virus heterogeneity that may be evident following virus infection/transmission, (ii) the efficacy of candidate RSV vaccines and therapeutics in the experimental infection model, and (iii) the potential emergence of escape mutants as a consequence of experimental drug treatments. Memphis-37 is a valuable tool for pre-clinical research, and to expedite the clinical development of vaccines, therapeutic immunomodulatory agents, and other antiviral drug strategies for the protection of vulnerable populations against RSV disease.
Collapse
Affiliation(s)
- Young-In Kim
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Children's Foundation Research Institute of Le Bonheur Children's Hospital, Memphis, Tennessee, United States of America
| | - John P. DeVincenzo
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Children's Foundation Research Institute of Le Bonheur Children's Hospital, Memphis, Tennessee, United States of America
| | - Bart G. Jones
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Rajeev Rudraraju
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Lisa Harrison
- Children's Foundation Research Institute of Le Bonheur Children's Hospital, Memphis, Tennessee, United States of America
| | - Rachel Meyers
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Jeff Cehelsky
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Rene Alvarez
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Julia L. Hurwitz
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
13
|
Vaccine-elicited CD8+ T cells protect against respiratory syncytial virus strain A2-line19F-induced pathogenesis in BALB/c mice. J Virol 2012; 86:13016-24. [PMID: 23015695 DOI: 10.1128/jvi.01770-12] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
CD8(+) T cells may contribute to vaccines for respiratory syncytial virus (RSV). Compared to CD8(+) T cells responding to RSV infection, vaccine-elicited anti-RSV CD8(+) T cells are less well defined. We used a peptide vaccine to test the hypothesis that vaccine-elicited RSV-specific CD8(+) T cells are protective against RSV pathogenesis. BALB/c mice were treated with a mixture (previously termed TriVax) of an M2(82-90) peptide representing an immunodominant CD8 epitope, the Toll-like receptor (TLR) agonist poly(I·C), and a costimulatory anti-CD40 antibody. TriVax vaccination induced potent effector anti-RSV CD8(+) cytotoxic T lymphocytes (CTL). Mice were challenged with RSV strain A2-line19F, a model of RSV pathogenesis leading to airway mucin expression. Mice were protected against RSV infection and against RSV-induced airway mucin expression and cellular lung inflammation when challenged 6 days after vaccination. Compared to A2-line19F infection alone, TriVax vaccination followed by challenge resulted in effector CD8(+) T cells with greater cytokine expression and the more rapid appearance of RSV-specific CD8(+) T cells in the lung. When challenged 42 days after TriVax vaccination, memory CD8(+) T cells were elicited with RSV-specific tetramer responses equivalent to TriVax-induced effector CD8(+) T cells. These memory CD8(+) T cells had lower cytokine expression than effector CD8(+) T cells, and protection against A2-line19F was partial during the memory phase. We found that vaccine-elicited effector anti-RSV CD8(+) T cells protected mice against RSV infection and pathogenesis, and waning protection correlated with reduced CD8(+) T cell cytokine expression.
Collapse
|
14
|
Remot A, Roux X, Dubuquoy C, Fix J, Bouet S, Moudjou M, Eléouët JF, Riffault S, Petit-Camurdan A. Nucleoprotein nanostructures combined with adjuvants adapted to the neonatal immune context: a candidate mucosal RSV vaccine. PLoS One 2012; 7:e37722. [PMID: 22655066 PMCID: PMC3359995 DOI: 10.1371/journal.pone.0037722] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 04/23/2012] [Indexed: 12/27/2022] Open
Abstract
Background The human respiratory syncytial virus (hRSV) is the leading cause of severe bronchiolitis in infants worldwide. The most severe RSV diseases occur between 2 and 6 months-of-age, so pediatric vaccination will have to be started within the first weeks after birth, when the immune system is prone to Th2 responses that may turn deleterious upon exposure to the virus. So far, the high risk to prime for immunopathological responses in infants has hampered the development of vaccine. In the present study we investigated the safety and efficacy of ring-nanostructures formed by the recombinant nucleoprotein N of hRSV (NSRS) as a mucosal vaccine candidate against RSV in BALB/c neonates, which are highly sensitive to immunopathological Th2 imprinting. Methodology and Principal Findings A single intranasal administration of NSRS with detoxified E.coli enterotoxin LT(R192G) to 5–7 day old neonates provided a significant reduction of the viral load after an RSV challenge at five weeks of age. However, neonatal vaccination also generated an enhanced lung infiltration by neutrophils and eosinophils following the RSV challenge. Analysis of antibody subclasses and cytokines produced after an RSV challenge or a boost administration of the vaccine suggested that neonatal vaccination induced a Th2 biased local immune memory. This Th2 bias and the eosinophilic reaction could be prevented by adding CpG to the vaccine formulation, which, however did not prevent pulmonary inflammation and neutrophil infiltration upon viral challenge. Conclusions/Significance In conclusion, protective vaccination against RSV can be achieved in neonates but requires an appropriate combination of adjuvants to prevent harmful Th2 imprinting.
Collapse
Affiliation(s)
- Aude Remot
- Molecular Virology and Immunology (UR892), French National Institute for Agricultural Research, Jouy-en-Josas, France
| | - Xavier Roux
- Molecular Virology and Immunology (UR892), French National Institute for Agricultural Research, Jouy-en-Josas, France
| | - Catherine Dubuquoy
- Molecular Virology and Immunology (UR892), French National Institute for Agricultural Research, Jouy-en-Josas, France
| | - Jenna Fix
- Molecular Virology and Immunology (UR892), French National Institute for Agricultural Research, Jouy-en-Josas, France
| | - Stephan Bouet
- Animal Genetics and Integrative Biology (UMR1313), French National Institute for Agricultural Research, Jouy-en-Josas, France
| | - Mohammed Moudjou
- Molecular Virology and Immunology (UR892), French National Institute for Agricultural Research, Jouy-en-Josas, France
| | - Jean-François Eléouët
- Molecular Virology and Immunology (UR892), French National Institute for Agricultural Research, Jouy-en-Josas, France
| | - Sabine Riffault
- Molecular Virology and Immunology (UR892), French National Institute for Agricultural Research, Jouy-en-Josas, France
- * E-mail:
| | - Agnès Petit-Camurdan
- Molecular Virology and Immunology (UR892), French National Institute for Agricultural Research, Jouy-en-Josas, France
| |
Collapse
|
15
|
Shao HY, Lin YW, Yu SL, Lin HY, Chitra E, Chang YC, Sia C, Chong P, Hsu MT, Wei OL, Chow YH. Immunoprotectivity of HLA-A2 CTL peptides derived from respiratory syncytial virus fusion protein in HLA-A2 transgenic mouse. PLoS One 2011; 6:e25500. [PMID: 21980478 PMCID: PMC3183052 DOI: 10.1371/journal.pone.0025500] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 09/06/2011] [Indexed: 11/18/2022] Open
Abstract
Identification of HLA-restricted CD8+ T cell epitopes is important to study RSV-induced immunity and illness. We algorithmically analyzed the sequence of the fusion protein (F) of respiratory syncytial virus (RSV) and generated synthetic peptides that can potentially bind to HLA-A*0201. Four out of the twenty-five 9-mer peptides tested: peptides 3 (F33–41), 13 (F214–222), 14 (F273–281), and 23 (F559–567), were found to bind to HLA-A*0201 with moderate to high affinity and were capable of inducing IFN-γ and IL-2 secretion in lymphocytes from HLA-A*0201 transgenic (HLA-Tg) mice pre-immunized with RSV or recombinant adenovirus expressing RSV F. HLA-Tg mice were immunized with these four peptides and were found to induce both Th1 and CD8+ T cell responses in in vitro secondary recall. Effector responses induced by these peptides were observed to confer differential protection against live RSV challenge. These peptides also caused better recovery of body weight loss induced by RSV. A significant reduction of lung viral load was observed in mice immunized with peptide 23, which appeared to enhance the levels of inflammatory chemokines (CCL17, CCL22, and IL-18) but did not increase eosinophil infiltration in the lungs. Whereas, significant reduction of infiltrated eosinophils induced by RSV infection was found in mice pre-immunized with peptide 13. Our results suggest that HLA-A2-restricted epitopes of RSV F protein could be useful for the development of epitope-based RSV vaccine.
Collapse
Affiliation(s)
- Hsiao-Yun Shao
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Yi-Wen Lin
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
- Graduate Program of Biotechnology in Medicine, Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Shu-Ling Yu
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Hsiang-Yin Lin
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Ebenezer Chitra
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Yung-Chen Chang
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Charles Sia
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Pele Chong
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Ming-Tao Hsu
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Olivia L. Wei
- The Graduate Division of Biological and Biomedical Sciences (GDBBS), Emory University, Atlanta, Georgia, United States of America
| | - Yen-Hung Chow
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
16
|
Rock MT, McKinney BA, Yoder SM, Prudom CE, Wright DW, Crowe JE. Identification of potential human respiratory syncytial virus and metapneumovirus T cell epitopes using computational prediction and MHC binding assays. J Immunol Methods 2011; 374:13-7. [PMID: 21854782 DOI: 10.1016/j.jim.2011.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 08/03/2011] [Accepted: 08/03/2011] [Indexed: 11/25/2022]
Abstract
Human respiratory syncytial virus (RSV) and human metapneumovirus (MPV) are two of the most common causes of serious viral lower respiratory tract illness in humans. CD8+ T cells have been shown to be important in animal models and human clinical studies for the clearance of viral infection, and they may contribute in part to protection against severe disease during reinfections. Precise enumeration and accurate phenotyping of RSV- or MPV-specific CD8+ T cells in humans is currently limited by the relatively small number of T cell epitopes that have been mapped with accompanying identification of MHC restriction patterns. We sought to expand the number of potential RSV and MPV epitopes for use in clinical and translational studies by identifying an expanded set of MHC-binding peptides based on RSV and MPV wild-type virus strain protein sequences. We interrogated the full protein sequences of all 9 or 11 proteins of MPV or RSV respectively using four established epitope prediction algorithms for human HLA A*0101, A*0201, or B*0702 binding and attempted to synthesize the top-scoring 150-152 peptides for each of the two viruses. Synthesis resulted in 442 synthesized and soluble peptides of the 452 predicted epitopes for MPV or RSV. We then determined the binding of the synthetic peptides to recombinant human HLA A*0101, A*0201 or B*0702 molecules with the predicted restriction using a commercially available plate-based assay, iTopia. A total of 230 of the 442 peptides tested exhibited binding to the appropriate MHC molecule. The binding results suggested that existing algorithms for prediction of MHC A*0201 binding are particularly robust. The binding results also provided a large benchmarking data collection for comparison of new prediction algorithms.
Collapse
Affiliation(s)
- Michael T Rock
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | |
Collapse
|
17
|
Cautivo KM, Bueno SM, Cortes CM, Wozniak A, Riedel CA, Kalergis AM. Efficient lung recruitment of respiratory syncytial virus-specific Th1 cells induced by recombinant bacillus Calmette-Guérin promotes virus clearance and protects from infection. THE JOURNAL OF IMMUNOLOGY 2010; 185:7633-45. [PMID: 21084664 DOI: 10.4049/jimmunol.0903452] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infection by the respiratory syncytial virus (RSV) can cause extensive inflammation and lung damage in susceptible hosts due to a Th2-biased immune response. Such a deleterious inflammatory response can be enhanced by immunization with formalin- or UV-inactivated RSV, as well as with vaccinia virus expressing the RSV-G protein. Recently, we have shown that vaccination with rBCG-expressing RSV Ags can prevent the disease in the mouse. To further understand the immunological mechanisms responsible for protection against RSV, we have characterized the T cell populations contributing to virus clearance in mice immunized with this BCG-based vaccine. We found that both CD4(+) and CD8(+) T cells were recruited significantly earlier to the lungs of infected mice that were previously vaccinated. Furthermore, we observed that simultaneous adoptive transfer of CD8(+) and CD4(+) RSV-specific T cells from vaccinated mice was required to confer protection against virus infection in naive recipients. In addition, CD4(+) T cells induced by vaccination released IFN-γ after RSV challenge, indicating that protection is mediated by a Th1 immune response. These data suggest that vaccination with rBCG-expressing RSV Ags can induce a specific effector/memory Th1 immune response consisting on CD4(+) and CD8(+) T cells, both necessary for a fully protective response against RSV. These results support the notion that an effective induction of Th1 T cell immunity against RSV during childhood could counteract the unbalanced Th2-like immune response triggered by the natural RSV infection.
Collapse
Affiliation(s)
- Kelly M Cautivo
- Millennium Nucleus on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | | | | | | | | | | |
Collapse
|
18
|
Kruijsen D, Bakkers MJ, van Uden NO, Viveen MC, van der Sluis TC, Kimpen JL, Leusen JH, Coenjaerts FE, van Bleek GM. Serum antibodies critically affect virus-specific CD4+/CD8+ T cell balance during respiratory syncytial virus infections. THE JOURNAL OF IMMUNOLOGY 2010; 185:6489-98. [PMID: 20971927 DOI: 10.4049/jimmunol.1002645] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Following infection with respiratory syncytial virus (RSV), reinfection in healthy individuals is common and presumably due to ineffective memory T cell responses. In peripheral blood of healthy adults, a higher CD4(+)/CD8(+) memory T cell ratio was observed compared with the ratio of virus-specific effector CD4(+)/CD8(+) T cells that we had found in earlier work during primary RSV infections. In mice, we show that an enhanced ratio of RSV-specific neutralizing to nonneutralizing Abs profoundly enhanced the CD4(+) T cell response during RSV infection. Moreover, FcγRs and complement factor C1q contributed to this Ab-mediated enhancement. Therefore, the increase in CD4(+) memory T cell response likely occurs through enhanced endosomal Ag processing dependent on FcγRs. The resulting shift in memory T cell response was likely amplified by suppressed T cell proliferation caused by RSV infection of APCs, a route important for Ag presentation via MHC class I molecules leading to CD8(+) T cell activation. Decreasing memory CD8(+) T cell numbers could explain the inadequate immunity during repeated RSV infections. Understanding this interplay of Ab-mediated CD4(+) memory T cell response enhancement and infection mediated CD8(+) memory T cell suppression is likely critical for development of effective RSV vaccines.
Collapse
Affiliation(s)
- Debby Kruijsen
- Department of Pediatrics, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Anderson R, Huang Y, Langley JM. Prospects for defined epitope vaccines for respiratory syncytial virus. Future Microbiol 2010; 5:585-602. [DOI: 10.2217/fmb.10.22] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The history of vaccines for respiratory syncytial virus (RSV) illustrates the complex immunity and immunopathology to this ubiquitous virus, starting from the failed formalin-inactivated vaccine trials performed in the 1960s. An attractive alternative to traditional live or killed virus vaccines is a defined vaccine composed of discrete antigenic epitopes for which immunological activities have been characterized as comprehensively as possible. Here we present cumulative data on murine and human CD4, CD8 and neutralization epitopes identified in RSV proteins along with information regarding their associated immune responses and host-dependent variability. Identification and characterization of RSV epitopes is a rapidly expanding topic of research with potential contributions to the tailored design of improved safe and effective vaccines.
Collapse
Affiliation(s)
- Robert Anderson
- Department of Microbiology & Immunology, Pediatrics and Canadian Center for Vaccinology, Dalhousie University, Halifax, Nova Scotia, B3H 1X5, Canada
| | - Yan Huang
- Department of Microbiology & Immunology and Canadian Center for Vaccinology, Dalhousie University, Halifax, Nova Scotia, B3H 1X5, Canada
| | - Joanne M Langley
- Department of Pediatrics, Community Health & Epidemiology and Canadian Center for Vaccinology, Dalhousie University, Halifax, Nova Scotia, B3H 1X5, Canada
| |
Collapse
|
20
|
A new subunit vaccine based on nucleoprotein nanoparticles confers partial clinical and virological protection in calves against bovine respiratory syncytial virus. Vaccine 2010; 28:3722-34. [PMID: 20307593 PMCID: PMC7115569 DOI: 10.1016/j.vaccine.2010.03.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 03/01/2010] [Accepted: 03/05/2010] [Indexed: 11/15/2022]
Abstract
Human and bovine respiratory syncytial viruses (HRSV and BRSV) are two closely related, worldwide prevalent viruses that are the leading cause of severe airway disease in children and calves, respectively. Efficacy of commercial bovine vaccines needs improvement and no human vaccine is licensed yet. We reported that nasal vaccination with the HRSV nucleoprotein produced as recombinant ring-shaped nanoparticles (N(SRS)) protects mice against a viral challenge with HRSV. The aim of this work was to evaluate this new vaccine that uses a conserved viral antigen, in calves, natural hosts for BRSV. Calves, free of colostral or natural anti-BRSV antibodies, were vaccinated with N(SRS) either intramuscularly, or both intramuscularly and intranasally using Montanide ISA71 and IMS4132 as adjuvants and challenged with BRSV. All vaccinated calves developed anti-N antibodies in blood and nasal secretions and N-specific cellular immunity in local lymph nodes. Clinical monitoring post-challenge demonstrated moderate respiratory pathology with local lung tissue consolidations for the non-vaccinated calves that were significantly reduced in the vaccinated calves. Vaccinated calves had lower viral loads than the non-vaccinated control calves. Thus N(SRS) vaccination in calves provided cross-protective immunity against BRSV infection without adverse inflammatory reaction.
Collapse
|
21
|
Abstract
Respiratory syncytial virus (RSV) is the leading cause of severe respiratory disease in infants and is an important source of morbidity and mortality in the elderly and immunocompromised. This review will discuss the humoral and cellular adaptive immune responses to RSV infection and how these responses are shaped in the immature immune system of the infant and the aged environment of the elderly. Furthermore, we will provide an overview of our current understanding of the role the various arms of the adaptive immune response play in mediating the delicate balance between the successful elimination of the virus from the host and the induction of immunopathology. Efficacious immunization against RSV remains a high priority within the field and we will highlight recent advances made in vaccine design.
Collapse
Affiliation(s)
- Matthew R Olson
- Department of Microbiology, 51 Newton Road, 3−532 Bowen Science Building, University of Iowa, Iowa City, IA 52242, USA Tel.: +1 319 335 8433 Fax: +1 319 335 9006
| | - Steven M Varga
- Department of Microbiology, Interdisciplinary Graduate Program in Immunology, 51 Newton Road, 3−532 Bowen Science Building, University of Iowa, Iowa City, IA 52242, USA Tel.: +1 319 335 7784 Fax: +1 319 335 9006
| |
Collapse
|
22
|
Herd KA, Nissen MD, Hopkins PM, Sloots TP, Tindle RW. Major histocompatibility complex class I cytotoxic T lymphocyte immunity to human metapneumovirus (hMPV) in individuals with previous hMPV infection and respiratory disease. J Infect Dis 2008; 197:584-92. [PMID: 18240952 DOI: 10.1086/526536] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Recently identified human metapneumovirus (hMPV) is an important respiratory pathogen in children and adults worldwide. Little is known about cytotoxic T lymphocyte (CTL) responses that may control hMPV infection in humans. To address this, we evaluated major histocompatibility complex (MHC) class I T cell immunity in 7 patients with previous hMPV respiratory disease. CTL responses were evident in most patients and to most proteins of hMPV. Individual patients had responses to at least 2 hMPV proteins (particularly the M protein) and had multiallele responses. In addition, we identified 9 CTL epitopes that are presented by human leukocyte antigen alleles of the most common MHC "supertypes." Many of these CTL epitopes are conserved across hMPV types, and there is epitope similarity between hMPV and human respiratory syncytial virus. This study provides the first report of MHC class I T cell immunity to hMPV in humans. These findings have significance for understanding cellular immunity to hMPV infection and for future vaccine development.
Collapse
Affiliation(s)
- Karen A Herd
- Royal Children's Hospital, Herston, Queensland, Australia
| | | | | | | | | |
Collapse
|
23
|
Roux X, Dubuquoy C, Durand G, Tran-Tolla TL, Castagné N, Bernard J, Petit-Camurdan A, Eléouët JF, Riffault S. Sub-nucleocapsid nanoparticles: a nasal vaccine against respiratory syncytial virus. PLoS One 2008; 3:e1766. [PMID: 18335041 PMCID: PMC2262139 DOI: 10.1371/journal.pone.0001766] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Accepted: 02/06/2008] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Bronchiolitis caused by the respiratory syncytial virus (RSV) in infants less than two years old is a growing public health concern worldwide, and there is currently no safe and effective vaccine. A major component of RSV nucleocapsid, the nucleoprotein (N), has been so far poorly explored as a potential vaccine antigen, even though it is a target of protective anti-viral T cell responses and is remarkably conserved between human RSV A and B serotypes. We recently reported a method to produce recombinant N assembling in homogenous rings composed of 10-11 N subunits enclosing a bacterial RNA. These nanoparticles were named sub-nucleocapsid ring structure (N SRS). METHODOLOGY AND PRINCIPAL FINDINGS The vaccine potential of N SRS was evaluated in a well-characterized and widely acknowledged mouse model of RSV infection. BALB/c adult mice were immunized intranasally with N SRS adjuvanted with the detoxified E. coli enterotoxin LT(R192G). Upon RSV challenge, vaccinated mice were largely protected against virus replication in the lungs, with a mild inflammatory lymphocytic and neutrophilic reaction in their airways. Mucosal immunization with N SRS elicited strong local and systemic immunity characterized by high titers of IgG1, IgG2a and IgA anti-N antibodies, antigen-specific CD8(+) T cells and IFN-gamma-producing CD4(+) T cells. CONCLUSIONS/SIGNIFICANCE This is the first report of using nanoparticles formed by the recombinant nucleocapsid protein as an efficient and safe intra-nasal vaccine against RSV.
Collapse
Affiliation(s)
- Xavier Roux
- Unité de Virologie et Immunologie Moléculaires (UR892), INRA, Jouy-en-Josas, France
| | - Catherine Dubuquoy
- Unité de Virologie et Immunologie Moléculaires (UR892), INRA, Jouy-en-Josas, France
| | - Guillaume Durand
- Unité de Virologie et Immunologie Moléculaires (UR892), INRA, Jouy-en-Josas, France
| | - Thi-Lan Tran-Tolla
- Unité de Virologie et Immunologie Moléculaires (UR892), INRA, Jouy-en-Josas, France
| | - Nathalie Castagné
- Unité de Virologie et Immunologie Moléculaires (UR892), INRA, Jouy-en-Josas, France
| | - Julie Bernard
- Unité de Virologie et Immunologie Moléculaires (UR892), INRA, Jouy-en-Josas, France
| | - Agnès Petit-Camurdan
- Unité de Virologie et Immunologie Moléculaires (UR892), INRA, Jouy-en-Josas, France
| | | | - Sabine Riffault
- Unité de Virologie et Immunologie Moléculaires (UR892), INRA, Jouy-en-Josas, France
- * E-mail:
| |
Collapse
|
24
|
Heidema J, Lukens MV, van Maren WWC, van Dijk MEA, Otten HG, van Vught AJ, van der Werff DBM, van Gestel SJP, Semple MG, Smyth RL, Kimpen JLL, van Bleek GM. CD8+ T cell responses in bronchoalveolar lavage fluid and peripheral blood mononuclear cells of infants with severe primary respiratory syncytial virus infections. THE JOURNAL OF IMMUNOLOGY 2008; 179:8410-7. [PMID: 18056387 DOI: 10.4049/jimmunol.179.12.8410] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A protective role for CD8+ T cells during viral infections is generally accepted, but little is known about how CD8+ T cell responses develop during primary infections in infants, their efficacy, and how memory is established after viral clearance. We studied CD8+ T cell responses in bronchoalveolar lavage (BAL) samples and blood of infants with a severe primary respiratory syncytial virus (RSV) infection. RSV-specific CD8+ T cells with an activated effector cell phenotype: CD27+CD28+CD45RO+CCR7-CD38+HLA-DR+Granzyme B+CD127- could be identified in BAL and blood. A high proportion of these CD8+ T cells proliferated and functionally responded upon in vitro stimulation with RSV Ag. Thus, despite the very young age of the patients, a robust systemic virus-specific CD8+ T cell response was elicited against a localized respiratory infection. RSV-specific T cell numbers as well as the total number of activated effector type CD8+ T cells peaked in blood around day 9-12 after the onset of primary symptoms, i.e., at the time of recovery. The lack of a correlation between RSV-specific T cell numbers and parameters of disease severity make a prominent role in immune pathology unlikely, in contrast the T cells might be involved in the recovery process.
Collapse
Affiliation(s)
- Jojanneke Heidema
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Terrosi C, Di Genova G, Savellini GG, Correale P, Blardi P, Cusi MG. Immunological Characterization of Respiratory Syncytial Virus N Protein Epitopes Recognized by Human Cytotoxic T Lymphocytes. Viral Immunol 2007; 20:399-406. [DOI: 10.1089/vim.2007.0041] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Chiara Terrosi
- Microbiology Section, Department of Molecular Biology, University of Siena, Siena, Italy
| | - Giuseppa Di Genova
- Microbiology Section, Department of Molecular Biology, University of Siena, Siena, Italy
| | - Gianni Gori Savellini
- Microbiology Section, Department of Molecular Biology, University of Siena, Siena, Italy
| | - Pierpaolo Correale
- Medical Oncology Section, Department of Human Pathology and Oncology, University of Siena, Siena, Italy
| | - Patrizia Blardi
- Department of Clinical Medicine and Immunological Sciences, University of Siena, Siena, Italy
| | - Maria G. Cusi
- Microbiology Section, Department of Molecular Biology, University of Siena, Siena, Italy
| |
Collapse
|
26
|
Meyer G, Deplanche M, Schelcher F. Human and bovine respiratory syncytial virus vaccine research and development. Comp Immunol Microbiol Infect Dis 2007; 31:191-225. [PMID: 17720245 DOI: 10.1016/j.cimid.2007.07.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2007] [Indexed: 11/23/2022]
Abstract
Human (HRSV) and bovine (BRSV) respiratory syncytial viruses (RSV) are two closely related viruses, which are the most important causative agents of respiratory tract infections of young children and calves, respectively. BRSV vaccines have been available for nearly 2 decades. They probably have reduced the prevalence of RSV infection but their efficacy needs improvement. In contrast, despite decades of research, there is no currently licensed vaccine for the prevention of HRSV disease. Development of a HRSV vaccine for infants has been hindered by the lack of a relevant animal model that develops disease, the need to immunize immunologically immature young infants, the difficulty for live vaccines to find the right balance between attenuation and immunogenicity, and the risk of vaccine-associated disease. During the past 15 years, intensive research into a HRSV vaccine has yielded vaccine candidates, which have been evaluated in animal models and, for some of them, in clinical trials in humans. Recent formulations have focused on subunit vaccines with specific CD4+ Th-1 immune response-activating adjuvants and on genetically engineered live attenuated vaccines. It is likely that different HRSV vaccines and/or combinations of vaccines used sequentially will be needed for the various populations at risk. This review discusses the recent advances in RSV vaccine development.
Collapse
Affiliation(s)
- Gilles Meyer
- INRA-ENVT, UMR1225 IHAP, Interactions Hôtes-Virus et Vaccinologie, Ecole Nationale Vétérinaire, 23 Chemin des Capelles, BP 87614, 31076 Toulouse Cedex, France.
| | | | | |
Collapse
|
27
|
de Waal L, Süzer Y, Wyatt LS, Sintnicolaas K, Sutter G, Moss B, Osterhaus ADME, de Swart RL. T Cell Responses to Respiratory Syncytial Virus Fusion and Attachment Proteins in Human Peripheral Blood Mononuclear Cells. Viral Immunol 2006; 19:669-78. [PMID: 17201662 DOI: 10.1089/vim.2006.19.669] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The cellular immune response to respiratory syncytial virus (RSV) is considered important in both protection and immunopathogenesis. We have studied the HLA class I- and class II-restricted T cell responses to RSV fusion (F) and attachment (G) proteins in peripheral blood mononuclear cells (PBMCs) obtained from healthy young adults. PBMCs were stimulated with autologous cells infected with recombinant modified vaccinia virus Ankara (rMVA) expressing RSV F (rMVA-F) or G (rMVA-G). In rMVA-F-stimulated bulk cultures F-specific CD4(+) and CD8(+) T cell responses were demonstrated, whereas in rMVA-G-stimulated cultures only G-specific CD4(+) T cell responses were detected. Using a set of overlapping peptides spanning the F protein, a number of the F-specific T cell responses could be mapped to different antigenic regions, whereas for the G protein only CD4(+) T cell responses recognizing the central conserved domain could be detected. These results suggest that the RSV glycoprotein-specific T cell response is directed to a number of different epitopes. Further studies must be performed to confirm the apparent inability of the RSV G protein to induce CD8(+) T cell responses. The rMVA-based in vitro stimulation protocol will be useful to define protein-specific T cell responses in different viral systems.
Collapse
Affiliation(s)
- Leon de Waal
- Department of Virology, Erasmus MC, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Meiring HD, Soethout EC, Poelen MCM, Mooibroek D, Hoogerbrugge R, Timmermans H, Boog CJ, Heck AJR, de Jong APJM, van Els CACM. Stable isotope tagging of epitopes: a highly selective strategy for the identification of major histocompatibility complex class I-associated peptides induced upon viral infection. Mol Cell Proteomics 2006; 5:902-13. [PMID: 16432254 DOI: 10.1074/mcp.t500014-mcp200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Identification of peptides presented in major histocompatibility complex (MHC) class I molecules after viral infection is of strategic importance for vaccine development. Until recently, mass spectrometric identification of virus-induced peptides was based on comparative analysis of peptide pools isolated from uninfected and virus-infected cells. Here we report on a powerful strategy aiming at the rapid, unambiguous identification of naturally processed MHC class I-associated peptides, which are induced by viral infection. The methodology, stable isotope tagging of epitopes (SITE), is based on metabolic labeling of endogenously synthesized proteins during infection. This is accomplished by culturing virus-infected cells with stable isotope-labeled amino acids that are expected to be anchor residues (i.e. residues of the peptide that have amino acid side chains that bind into pockets lining the peptide-binding groove of the MHC class I molecule) for the human leukocyte antigen allele of interest. Subsequently these cells are mixed with an equal number of non-infected cells, which are cultured in normal medium. Finally peptides are acid-eluted from immunoprecipitated MHC molecules and subjected to two-dimensional nanoscale LC-MS analysis. Virus-induced peptides are identified through computer-assisted detection of characteristic, binomially distributed ratios of labeled and unlabeled molecules. Using this approach we identified novel measles virus and respiratory syncytial virus epitopes as well as infection-induced self-peptides in several cell types, showing that SITE is a unique and versatile method for unequivocal identification of disease-related MHC class I epitopes.
Collapse
Affiliation(s)
- Hugo D Meiring
- Laboratory for Vaccine Research, Unit Research and Development, Netherlands Vaccine Institute, 3720 AL Bilthoven, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Appay V. Virus Specific T-Cell Responses. ANALYZING T CELL RESPONSES 2005. [PMCID: PMC7120106 DOI: 10.1007/1-4020-3623-x_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
CD8+ and CD4+ T-cells play a key role in the maintenance of our immunity against viruses. Recent technological developments, such as the use of MHC-peptide tetrameric complexes, have permitted significant improvements in the study of these cells. It is now possible to assess precisely frequencies as well as phenotypic and functional features of virus specific T-cells from the onset of many viral infections onwards. Different virus specific T-cell populations exhibit distinct functional characteristics and can be positioned at different stages of a process of post-thymic development, which we are drawing near to understanding the significance. Still, further work is needed before consensus is reached as regards what defines and how to induce the optimal virus specific T-cell response which will confer long lasting immunological protection in humans.
Collapse
|
30
|
Heidema J, de Bree GJ, de Graaff PMA, van Maren WWC, Hoogerhout P, Out TA, Kimpen JLL, van Bleek GM. Human CD8(+) T cell responses against five newly identified respiratory syncytial virus-derived epitopes. J Gen Virol 2004; 85:2365-2374. [PMID: 15269378 DOI: 10.1099/vir.0.80131-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
CD8(+) T lymphocytes play a major role in the clearance of respiratory syncytial virus (RSV) infections. To be able to study the primary CTL response in RSV-infected children, epitopes presented by a set of commonly used HLA alleles (HLA-A1, -A3, -B44 and -B51) were searched for. Five epitopes were characterized derived from the matrix (M), non-structural (NS2) and second matrix (M2) proteins of RSV. All epitopes were shown to be processed and presented by RSV-infected antigen-presenting cells. HLA-A1 tetramers for one of these epitopes derived from the M protein were constructed and used to quantify and phenotype the memory CD8(+) T cell pool in a panel of healthy adult donors. In about 60 % of the donors, CD8(+) T cells specific for the M protein could be identified. These cells belonged to the memory T cell subset characterized by expression of CD27 and CD28, and down-regulation of CCR7 and CD45RA. The frequency of tetramer-positive cells varied between 0.4 and 3 per 10(4) CD8(+) T cells in PBMC of healthy asymptomatic adult donors.
Collapse
Affiliation(s)
- Jojanneke Heidema
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center, KE 04.133.1, PO Box 85500, 3508 AB Utrecht, The Netherlands
| | - Godelieve J de Bree
- Division of Pulmonology and the Department of Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands
| | - Patricia M A de Graaff
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center, KE 04.133.1, PO Box 85500, 3508 AB Utrecht, The Netherlands
| | - Wendy W C van Maren
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center, KE 04.133.1, PO Box 85500, 3508 AB Utrecht, The Netherlands
| | | | - Theo A Out
- Division of Clinical Immunology, Academic Medical Center, Amsterdam, The Netherlands
| | - Jan L L Kimpen
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center, KE 04.133.1, PO Box 85500, 3508 AB Utrecht, The Netherlands
| | - Grada M van Bleek
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center, KE 04.133.1, PO Box 85500, 3508 AB Utrecht, The Netherlands
| |
Collapse
|
31
|
de Waal L, Yüksel S, Brandenburg AH, Langedijk JPM, Sintnicolaas K, Verjans GMGM, Osterhaus ADME, de Swart RL. Identification of a common HLA-DP4-restricted T-cell epitope in the conserved region of the respiratory syncytial virus G protein. J Virol 2004; 78:1775-81. [PMID: 14747542 PMCID: PMC369497 DOI: 10.1128/jvi.78.4.1775-1781.2004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The cellular immune response to respiratory syncytial virus (RSV) is important in both protection and immunopathogenesis. In contrast to HLA class I, HLA class II-restricted RSV-specific T-cell epitopes have not been identified. Here, we describe the generation and characterization of two human RSV-specific CD4(+)-T-cell clones (TCCs) associated with type 0-like cytokine profiles. TCC 1 was specific for the matrix protein and restricted over HLA-DPB1*1601, while TCC 2 was specific for the attachment protein G and restricted over either HLA-DPB1*0401 or -0402. Interestingly, the latter epitope is conserved in both RSV type A and B viruses. Given the high allele frequencies of HLA-DPB1*0401 and -0402 worldwide, this epitope could be widely recognized and boosted by recurrent RSV infections. Indeed, peptide stimulation of peripheral blood mononuclear cells from healthy adults resulted in the detection of specific responses in 8 of 13 donors. Additional G-specific TCCs were generated from three of these cultures, which recognized the identical (n = 2) or almost identical (n = 1) HLA-DP4-restricted epitope as TCC 2. No significant differences were found between the capacities of cell lines obtained from infants with severe (n = 41) or mild (n = 46) RSV lower respiratory tract infections to function as antigen-presenting cells to the G-specific TCCs, suggesting that the severity of RSV disease is not linked to the allelic frequency of HLA-DP4. In conclusion, we have identified an RSV G-specific human T helper cell epitope restricted by the widely expressed HLA class II alleles DPB1*0401 and -0402. Its putative role in protection and/or immunopathogenesis remains to be determined.
Collapse
Affiliation(s)
- L de Waal
- Department of Virology, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Bollard CM, Kuehnle I, Leen A, Rooney CM, Heslop HE. Adoptive immunotherapy for posttransplantation viral infections. Biol Blood Marrow Transplant 2004; 10:143-55. [PMID: 14993880 DOI: 10.1016/j.bbmt.2003.09.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Viral diseases are a major cause of morbidity and mortality after hemopoietic stem cell transplantation. Because viral complications in these patients are clearly associated with the lack of recovery of virus-specific cellular immune responses, reconstitution of the host with in vitro expanded cytotoxic T lymphocytes is a potential approach to prevent and treat these diseases. Initial clinical studies of cytomegalovirus and Epstein-Barr virus in human stem cell transplant patients have shown that adoptively transferred donor-derived virus-specific T cells may restore protective immunity and control established infections. Preclinical studies are evaluating this approach for other viruses while strategies for generating T cells specific for multiple viruses to provide broader protection are being evaluated in clinical trials. The use of genetically modified T cells or the use of newer suicide genes may result in improved safety and efficacy.
Collapse
Affiliation(s)
- Catherine M Bollard
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
33
|
Yui I, Hoshi A, Shigeta Y, Takami T, Nakayama T. Detection of human respiratory syncytial virus sequences in peripheral blood mononuclear cells. J Med Virol 2003; 70:481-9. [PMID: 12767015 DOI: 10.1002/jmv.10421] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Peripheral blood mononuclear cells (PBMC) obtained from patients with lower respiratory infections were examined for the detection of human respiratory syncytial virus (RSV) sequences in the N region using the reverse transcription polymerase chain reaction (RT-PCR). RSV infection was confirmed by at least one method, i.e., virus isolation, enzyme immunoassay for viral antigen, and RT-PCR of nasopharyngeal secretions (NPS) samples. The detection rate for RSV RNA in PBMC obtained from RSV-infected patients was 40% (38/94), compared to 5% (1/20) in controls (P = 0.002). Between the groups positive (38) and negative (56) for RSV RNA in PBMC, there was no significant difference in clinical parameters. Seven patients had eight episodes of reinfection and RSV RNA was detected in 50% (4/8) during consecutive infections. Sequences of their PBMC samples were distinct from those of prototype strains of subgroup A and B. However, they were not always consistent with those of paired NPS samples. The findings suggested that RSV RNA could be detected in PBMC even during reinfection and as might have the possibility of quasispecies dynamics, reflecting the nature of RNA viruses.
Collapse
Affiliation(s)
- Ikuko Yui
- Kitasato Institute for Life Sciences, Laboratory of Viral Infection, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
34
|
Venter M, Rock M, Puren AJ, Tiemessen CT, Crowe JE. Respiratory syncytial virus nucleoprotein-specific cytotoxic T-cell epitopes in a South African population of diverse HLA types are conserved in circulating field strains. J Virol 2003; 77:7319-29. [PMID: 12805430 PMCID: PMC164818 DOI: 10.1128/jvi.77.13.7319-7329.2003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This study identifies memory cytotoxic T lymphocyte (CTL) epitopes to respiratory syncytial virus (RSV) in healthy South African adults and demonstrates the conservation of those epitopes in circulating field strains of RSV in South Africa. Thirty-seven healthy adults from a population with diverse HLA backgrounds were screened by gamma interferon (IFN-gamma) enzyme-linked immunospot for memory CTL activity in response to overlapping peptides representing the complete nucleoprotein (N) of RSV. Responses of more than 40 spot-forming cells/million cells were detectable in 21 individuals. The significant responses were further characterized, and 14-mer peptides were identified that induced cytolytic activity. Fine mapping of peptides with the highest cytolytic activity identified an HLA-B(*)08-restricted RSV-specific CTL epitope. The extended 14-mer peptide containing this epitope also induced lysis in the context of A(*)02-restricted target cells in some individuals. These HLA types are common in the target population; thus, the epitope is useful for studies of CTL responses to RSV in humans. The epitope was detected in healthy adults, reflecting the response generated in the course of previous natural RSV infection. We obtained a large panel of naturally occurring isolates of RSV to determine whether there was evidence of escape from CTL activity in circulating strains. We found that this epitope and a previously identified B(*)07-restricted N protein epitope were conserved in RSV field strains representing the diversity of circulating genotypes. This work suggests that escape from CTL activity is not common for this acute respiratory infection.
Collapse
Affiliation(s)
- Marietjie Venter
- National Institute for Communicable Diseases, Sandringham, South Africa
| | | | | | | | | |
Collapse
|
35
|
Rock MT, Crowe JE. Identification of a novel human leucocyte antigen-A*01-restricted cytotoxic T-lymphocyte epitope in the respiratory syncytial virus fusion protein. Immunology 2003; 108:474-80. [PMID: 12667209 PMCID: PMC1782928 DOI: 10.1046/j.1365-2567.2003.01619.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Virus-specific cytotoxic T lymphocytes (CTL) play a major role in the clearance of respiratory syncytial virus (RSV) infection. To begin monitoring the immunological response to infection, especially in infants, it is important to identify human leucocyte antigen (HLA)-restricted CTL epitopes. Herein, we used a novel, comprehensive peptide panel containing all possible 8-, 9- and 10-mer peptides spanning the RSV fusion protein to screen for novel HLA-restricted T-cell epitopes. These peptide sets were synthesized as 10-mer peptides overlapping by nine amino acids and contained corresponding 8- and 9-mer peptides generated by C-terminal truncation. Unselected and uncultured peripheral blood mononuclear cells from healthy adult subjects were screened by interferon-gamma (IFN-gamma) Elispot assays against the peptide panel. Seven of 19 subjects displayed positive responses against 10 of the 565 peptides analysed. An HLA-A*01-restricted CTL epitope detected in three healthy adult subjects is characterized. This is the first RSV-specific memory CTL response identified in the fusion protein of RSV.
Collapse
Affiliation(s)
- Michael T Rock
- Departments of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee 37205, USA
| | | |
Collapse
|
36
|
Gaddum RM, Cook RS, Furze JM, Ellis SA, Taylor G. Recognition of bovine respiratory syncytial virus proteins by bovine CD8+ T lymphocytes. Immunology 2003; 108:220-9. [PMID: 12562331 PMCID: PMC1782889 DOI: 10.1046/j.1365-2567.2003.01566.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD8+ T lymphocytes play a major role in the clearance of bovine respiratory syncytial virus (BRSV), an important respiratory pathogen of young calves that shares many of the epidemiological and pathological features of human respiratory syncytial virus (HRSV) in infants. Recombinant vaccinia virus (rVV) and recombinant fowlpox virus (rFPV), expressing individual BRSV proteins, were used to demonstrate that the F, N and M2 proteins were the major antigens recognized by bovine CD8+ T cells in major histocompatibility complex (MHC)-defined cattle. BRSV protein recognition by CD8+ T cells was analysed using cytotoxic T lymphocyte (CTL) assays or by the production of interferon-gamma (IFN-gamma) following restimulation with BRSV proteins. Strong recognition of the G protein by CD8+ T cells was observed in cattle that had been vaccinated with rVV expressing this protein and subsequently challenged with BRSV. Although there is variation in the number of expressed MHC genes in cattle with different class I haplotypes, this did not appear to influence BRSV protein recognition by CD8+ T cells. Knowledge of the antigenic specificity of BRSV-specific CD8+ T cells will facilitate the qualitative and quantitative analysis of BRSV-specific CD8+ T-cell memory in cattle and help to ensure that potential vaccines induce a qualitatively appropriate CD8+ T-cell response.
Collapse
Affiliation(s)
- Ruth M Gaddum
- Institute for Animal Health, Compton, Newbury, Berkshire, UK
| | | | | | | | | |
Collapse
|
37
|
van Bleek GM, Poelen MC, van der Most R, Brugghe HF, Timmermans HAM, Boog CJ, Hoogerhout P, Otten HG, van Els CACM. Identification of immunodominant epitopes derived from the respiratory syncytial virus fusion protein that are recognized by human CD4 T cells. J Virol 2003; 77:980-8. [PMID: 12502814 PMCID: PMC140824 DOI: 10.1128/jvi.77.2.980-988.2003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Memory CD4 T-cell responses against respiratory syncytial virus (RSV) were evaluated in peripheral blood mononuclear cells of healthy blood donors with gamma interferon enzyme-linked immunospot (Elispot) assays. RSV-specific responses were detected in every donor at levels varying between 0.05 and 0.3% of CD4 T cells. For all donors tested, a considerable component of the CD4 T-cell response was directed against the fusion (F) protein of RSV. We characterized a set of 31 immunodominant antigenic peptides targeted by CD4 T cells in the context of the most prevalent HLA class II molecules within the Caucasian population. Most antigenic peptides were HLA-DR restricted, whereas two dominant DQ peptides were also identified. The antigenic peptides identified were located across the entire sequence of the F protein. Several peptides were presented by more than one major histocompatibility complex class II molecule. Furthermore, most donors recognized several F peptides. Detailed knowledge about immunodominant antigenic peptides will facilitate the ability to monitor CD4 T-cell responses in patients and the measurement of correlates of protection in vaccinated subjects.
Collapse
Affiliation(s)
- Grada M van Bleek
- Laboratory for Vaccine Research, National Institute for Public Health and the Environment, Bilthoven, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Klenerman P, Cerundolo V, Dunbar PR. Tracking T cells with tetramers: new tales from new tools. Nat Rev Immunol 2002; 2:263-72. [PMID: 12001997 DOI: 10.1038/nri777] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
To understand the success or failure of immune responses against pathogens or tumours requires the direct measurement of specific lymphocytes. Recently, there has been an explosion of data in this field through the use of several new tools for measuring the number and function of T cells. This has allowed immunologists who study human disease and mouse models of infection and cancer to readily track specific T cells--in both time and space. Although there are common patterns, over time, each host-pathogen relationship seems to develop unique characteristics, as reflected in the quality of the T-cell response.
Collapse
Affiliation(s)
- Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, UK.
| | | | | |
Collapse
|
39
|
Abstract
Defects in cytotoxic T-lymphocyte (CTL) function after hemopoietic stem cell transplantation (HSCT) are associated with an increased frequency and severity of viral diseases. Initial investigations of viral infections in immunosuppressed mice and subsequent clinical studies of cytomegalovirus (CMV) and Epstein-Barr virus (EBV) in human stem cell transplant patients have suggested that adoptive transfer of virus-specific T cells may restore protective immunity and control established infections. Current efforts focus on optimizing adoptive immunotherapy approaches and developing strategies for generating T cells specific for multiple viruses to provide broader protection.
Collapse
Affiliation(s)
- Benedikt Gahn
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
40
|
Chang J, Srikiatkhachorn A, Braciale TJ. Visualization and characterization of respiratory syncytial virus F-specific CD8(+) T cells during experimental virus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:4254-60. [PMID: 11591747 DOI: 10.4049/jimmunol.167.8.4254] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CTL play a major role in the clearance of respiratory syncytial virus (RSV) during experimental pulmonary infection. The fusion (F) glycoprotein of RSV is a protective Ag that elicits CTL and Ab response against RSV infection in BALB/c mice. We used the strategy of screening a panel of overlapping synthetic peptides corresponding to the RSV F protein and identified an immunodominant H-2K(d)-restricted epitope (F(85-93); KYKNAVTEL) recognized by CD8(+) T cells from BALB/c mice. We enumerated the F-specific CD8(+) T cell response in the lungs of infected mice by flow cytometry using tetramer staining and intracellular cytokine synthesis. During primary infection, F(85-93)-specific effector CD8(+) T cells constitute approximately 4.8% of pulmonary CD8(+) T cells at the peak of the primary response (day 8), whereas matrix 2-specific CD8(+) T cells constituted approximately 50% of the responding CD8(+) T cell population in the lungs. When RSV F-immune mice undergo a challenge RSV infection, the F-specific CD8(+) T cell response is accelerated and dominates, whereas the primary response to the matrix 2 epitope in the lungs is reduced by approximately 20-fold. In addition, we found that activated F-specific effector CD8(+) T cells isolated from the lungs of RSV-infected mice exhibited a lower than expected frequency of IFN-gamma-producing CD8(+) T cells and were significantly impaired in ex vivo cytolytic activity compared with competent F-specific effector CD8(+) T cells generated in vitro. The significance of these results for the regulation of the CD8(+) T cell response to RSV is discussed.
Collapse
Affiliation(s)
- J Chang
- Beirne B. Carter Center for Immunology Research and Department of Pathology and Microbiology, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
41
|
Schamel K, Staeheli P, Hausmann J. Identification of the immunodominant H-2K(k)-restricted cytotoxic T-cell epitope in the Borna disease virus nucleoprotein. J Virol 2001; 75:8579-88. [PMID: 11507203 PMCID: PMC115103 DOI: 10.1128/jvi.75.18.8579-8588.2001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Borna disease virus (BDV)-induced immunopathology in mice is most prominent in strains carrying the major histocompatibility complex H-2k allele and is mediated by CD8(+) T cells that are directed against the viral nucleoprotein p40. We now identified the highly conserved octamer peptide TELEISSI, located between amino acid residues 129 and 136 of BDV p40, as a potent H-2K(k)-restricted cytotoxic T-cell (CTL) epitope. When added to the culture medium of L929 target cells, TELEISSI conferred sensitivity to lysis by CTLs isolated from brains of BDV-infected MRL mice with acute neurological disease. Vaccinia virus-mediated expression of a p40 variant with mutations in the two K(k)-specific anchor residues of the TELEISSI peptide (p40(E130K,I136T)) did not sensitize L929 target cells for lysis by BDV-specific CTLs, whereas expression of wild-type p40 did. Furthermore, unlike vaccination with wild-type p40, vaccination of persistently infected symptomless B10.BR mice with p40(E130K,I136T) did not result in central nervous system inflammation and neurological disease. These results demonstrate that TELEISSI is the immunodominant CTL epitope of BDV p40 in H-2k mice.
Collapse
Affiliation(s)
- K Schamel
- Abteilung Virologie, Institut für Medizinische Mikrobiologie und Hygiene, Universität Freiburg, D-79104 Freiburg, Germany
| | | | | |
Collapse
|
42
|
Lechner F, Cuero AL, Kantzanou M, Klenerman P. Studies of human antiviral CD8+ lymphocytes using class I peptide tetramers. Rev Med Virol 2001; 11:11-22. [PMID: 11241799 DOI: 10.1002/rmv.295] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Understanding the interactions between a host and a pathogen relies crucially on quantitative measurements of immune responses. Until recently, measurements of the levels of cellular immune responses, i.e. those mediated by CD4+ and CD8+ T lymphocytes have depended largely on culture in vitro and subsequent measurement of specific functions (such as cytolysis). More recently, new technologies based around tetrameric class I peptide complexes (tetramers) have allowed immunologists to measure CD8+ T lymphocyte levels directly ex vivo and independently of function. Since CD8+ lymphocytes play a key role in a number of important human viral infections, these tools have yielded useful insights into the dynamics, phenotype and function of human antiviral lymphocyte populations. In this review we describe some of the basic aspects of the biology of virus-specific CD8+ lymphocytes, and the current methods available to detect them. The use of tetramers has, in just four years, transformed our understanding of the immune responses against HIV, HTLV-1, HBV, HCV, CMV and EBV, and holds promise in a number of areas where quantitative analysis of the antiviral response in terms of both number and function is critical.
Collapse
Affiliation(s)
- F Lechner
- Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | | | | | | |
Collapse
|