1
|
Tang X, Zhang W, Zhang Z. Developing T Cell Epitope-Based Vaccines Against Infection: Challenging but Worthwhile. Vaccines (Basel) 2025; 13:135. [PMID: 40006681 PMCID: PMC11861332 DOI: 10.3390/vaccines13020135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
T cell epitope-based vaccines are designed to elicit long-lived pathogen-specific memory T cells that can quickly activate protective effector functions in response to subsequent infections. These vaccines have the potential to provide sustained protection against mutated variants, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which are increasingly capable of evading neutralizing antibodies. Recent advancements in epitope discovery, T cell receptor analysis, and bioinformatics have enabled the precise selection of epitopes and the sophisticated design of epitope-based vaccines. This review outlines the development process for T cell epitope-based vaccines. We summarize the current progress in T cell epitope discovery technologies, highlighting the advantages and disadvantages of each method. We also examine advancements in the design and optimization of epitope-based vaccines, particularly through bioinformatics tools. Additionally, we discuss the challenges of validating the accurate processing and presentation of individual epitopes and establishing suitable rodent models to evaluate vaccine immunogenicity and protective efficacy.
Collapse
Affiliation(s)
- Xian Tang
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen 518112, China; (X.T.); (W.Z.)
| | - Wei Zhang
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen 518112, China; (X.T.); (W.Z.)
| | - Zheng Zhang
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen 518112, China; (X.T.); (W.Z.)
- Guangdong Key Laboratory for Anti-Infection Drug Quality Evaluation, Shenzhen 518112, China
| |
Collapse
|
2
|
Camarena ME, Theunissen P, Ruiz M, Ruiz-Orera J, Calvo-Serra B, Castelo R, Castro C, Sarobe P, Fortes P, Perera-Bel J, Albà MM. Microproteins encoded by noncanonical ORFs are a major source of tumor-specific antigens in a liver cancer patient meta-cohort. SCIENCE ADVANCES 2024; 10:eadn3628. [PMID: 38985879 PMCID: PMC11235171 DOI: 10.1126/sciadv.adn3628] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 06/04/2024] [Indexed: 07/12/2024]
Abstract
The expression of tumor-specific antigens during cancer progression can trigger an immune response against the tumor. Here, we investigate if microproteins encoded by noncanonical open reading frames (ncORFs) are a relevant source of tumor-specific antigens. We analyze RNA sequencing data from 117 hepatocellular carcinoma (HCC) tumors and matched healthy tissue together with ribosome profiling and immunopeptidomics data. Combining human leukocyte antigen-epitope binding predictions and experimental validation experiments, we conclude that around 40% of the tumor-specific antigens in HCC are likely to be derived from ncORFs, including two peptides that can trigger an immune response in humanized mice. We identify a subset of 33 tumor-specific long noncoding RNAs expressing novel cancer antigens shared by more than 10% of the HCC samples analyzed, which, when combined, cover a large proportion of the patients. The results of the study open avenues for extending the range of anticancer vaccines.
Collapse
Affiliation(s)
| | - Patrick Theunissen
- Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain
| | - Marta Ruiz
- Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain
| | - Jorge Ruiz-Orera
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Beatriz Calvo-Serra
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Robert Castelo
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Carla Castro
- Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain
| | - Pablo Sarobe
- Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Cancer Clinic University of Navarra (CCUN), Pamplona, Spain
| | - Puri Fortes
- Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Cancer Clinic University of Navarra (CCUN), Pamplona, Spain
- Spanish Network for Advanced Therapies (TERAV ISCIII), Madrid, Spain
| | | | - M Mar Albà
- Hospital del Mar Research Institute, Barcelona, Spain
- Catalan Institute for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
3
|
Srivastava PK. Cancer neoepitopes viewed through negative selection and peripheral tolerance: a new path to cancer vaccines. J Clin Invest 2024; 134:e176740. [PMID: 38426497 PMCID: PMC10904052 DOI: 10.1172/jci176740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
A proportion of somatic mutations in tumors create neoepitopes that can prime T cell responses that target the MHC I-neoepitope complexes on tumor cells, mediating tumor control or rejection. Despite the compelling centrality of neoepitopes to cancer immunity, we know remarkably little about what constitutes a neoepitope that can mediate tumor control in vivo and what distinguishes such a neoepitope from the vast majority of similar candidate neoepitopes that are inefficacious in vivo. Studies in mice as well as clinical trials have begun to reveal the unexpected paradoxes in this area. Because cancer neoepitopes straddle that ambiguous ground between self and non-self, some rules that are fundamental to immunology of frankly non-self antigens, such as viral or model antigens, do not appear to apply to neoepitopes. Because neoepitopes are so similar to self-epitopes, with only small changes that render them non-self, immune response to them is regulated at least partially the way immune response to self is regulated. Therefore, neoepitopes are viewed and understood here through the clarifying lens of negative thymic selection. Here, the emergent questions in the biology and clinical applications of neoepitopes are discussed critically and a mechanistic and testable framework that explains the complexity and translational potential of these wonderful antigens is proposed.
Collapse
|
4
|
Peri A, Salomon N, Wolf Y, Kreiter S, Diken M, Samuels Y. The landscape of T cell antigens for cancer immunotherapy. NATURE CANCER 2023:10.1038/s43018-023-00588-x. [PMID: 37415076 DOI: 10.1038/s43018-023-00588-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 05/18/2023] [Indexed: 07/08/2023]
Abstract
The remarkable capacity of immunotherapies to induce durable regression in some patients with metastatic cancer relies heavily on T cell recognition of tumor-presented antigens. As checkpoint-blockade therapy has limited efficacy, tumor antigens have the potential to be exploited for complementary treatments, many of which are already in clinical trials. The surge of interest in this topic has led to the expansion of the tumor antigen landscape with the emergence of new antigen categories. Nonetheless, how different antigens compare in their ability to elicit efficient and safe clinical responses remains largely unknown. Here, we review known cancer peptide antigens, their attributes and the relevant clinical data and discuss future directions.
Collapse
Affiliation(s)
- Aviyah Peri
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Nadja Salomon
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Yochai Wolf
- Ella Lemelbaum Institute for Immuno-oncology and Skin Cancer, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Sebastian Kreiter
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany.
| | - Mustafa Diken
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany.
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
5
|
Lozano-Rabella M, Garcia-Garijo A, Palomero J, Yuste-Estevanez A, Erhard F, Farriol-Duran R, Martín-Liberal J, Ochoa-de-Olza M, Matos I, Gartner JJ, Ghosh M, Canals F, Vidal A, Piulats JM, Matías-Guiu X, Brana I, Muñoz-Couselo E, Garralda E, Schlosser A, Gros A. Exploring the Immunogenicity of Noncanonical HLA-I Tumor Ligands Identified through Proteogenomics. Clin Cancer Res 2023; 29:2250-2265. [PMID: 36749875 PMCID: PMC10261919 DOI: 10.1158/1078-0432.ccr-22-3298] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/20/2022] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
PURPOSE Tumor antigens are central to antitumor immunity. Recent evidence suggests that peptides from noncanonical (nonC) aberrantly translated proteins can be presented on HLA-I by tumor cells. Here, we investigated the immunogenicity of nonC tumor HLA-I ligands (nonC-TL) to better understand their contribution to cancer immunosurveillance and their therapeutic applicability. EXPERIMENTAL DESIGN Peptides presented on HLA-I were identified in 9 patient-derived tumor cell lines from melanoma, gynecologic, and head and neck cancer through proteogenomics. A total of 507 candidate tumor antigens, including nonC-TL, neoantigens, cancer-germline, or melanocyte differentiation antigens, were tested for T-cell recognition of preexisting responses in patients with cancer. Donor peripheral blood lymphocytes (PBL) were in vitro sensitized against 170 selected nonC-TL to isolate antigen-specific T-cell receptors (TCR) and evaluate their therapeutic potential. RESULTS We found no recognition of the 507 nonC-TL tested by autologous ex vivo expanded tumor-reactive T-cell cultures while the same cultures demonstrated reactivity to mutated, cancer-germline, or melanocyte differentiation antigens. However, in vitro sensitization of donor PBL against 170 selected nonC-TL, led to the identification of TCRs specific to three nonC-TL, two of which mapped to the 5' UTR regions of HOXC13 and ZKSCAN1, and one mapping to a noncoding spliced variant of C5orf22C. T cells targeting these nonC-TL recognized cancer cell lines naturally presenting their corresponding antigens. Expression of the three immunogenic nonC-TL was shared across tumor types and barely or not detected in normal cells. CONCLUSIONS Our findings predict a limited contribution of nonC-TL to cancer immunosurveillance but demonstrate they may be attractive novel targets for widely applicable immunotherapies. See related commentary by Fox et al., p. 2173.
Collapse
Affiliation(s)
- Maria Lozano-Rabella
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Andrea Garcia-Garijo
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Jara Palomero
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Anna Yuste-Estevanez
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Florian Erhard
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Roc Farriol-Duran
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Juan Martín-Liberal
- Early Drug Development Unit (UITM) Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital, Barcelona, Spain
| | - Maria Ochoa-de-Olza
- Early Drug Development Unit (UITM) Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital, Barcelona, Spain
| | - Ignacio Matos
- Early Drug Development Unit (UITM) Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital, Barcelona, Spain
| | - Jared J. Gartner
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland
| | - Michael Ghosh
- Institute for Cell Biology Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Francesc Canals
- Proteomics, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital, Barcelona, Spain
| | - August Vidal
- Department of Pathology. Hospital Universitari de Bellvitge-IDIBELL, CIBERONC, Barcelona, Spain
| | - Josep Maria Piulats
- Medical Oncology, Catalan Institute of Cancer (ICO), IDIBELL-Oncobell, Hospitalet de Llobregat, Spain
| | - Xavier Matías-Guiu
- Department of Pathology. Hospital Universitari de Bellvitge-IDIBELL, CIBERONC, Barcelona, Spain
| | - Irene Brana
- Early Drug Development Unit (UITM) Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital, Barcelona, Spain
| | - Eva Muñoz-Couselo
- Melanoma and other skin tumors unit, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital, Barcelona, Spain
| | - Elena Garralda
- Early Drug Development Unit (UITM) Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital, Barcelona, Spain
| | - Andreas Schlosser
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Alena Gros
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| |
Collapse
|
6
|
Apcher S, Vojtesek B, Fahraeus R. In search of the cell biology for self- versus non-self- recognition. Curr Opin Immunol 2023; 83:102334. [PMID: 37210933 DOI: 10.1016/j.coi.2023.102334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 05/23/2023]
Abstract
Several of today's cancer treatments are based on the immune system's capacity to detect and destroy cells expressing neoantigens on major histocompatibility class-I molecules (MHC-I). Despite this, we still do not know the cell biology behind how antigenic peptide substrates (APSs) for the MHC-I pathway are produced. Indeed, there are few research fields with so many divergent views as the one concerning the source of APSs. This is quite remarkable considering their fundamental role in the immune systems' capacity to detect and destroy virus-infected or transformed cells. A better understanding of the processes generating APSs and how these are regulated will shed light on the evolution of self-recognition and provide new targets for therapeutic intervention. We discuss the search for the elusive source of MHC-I peptides and highlight the cell biology that is still missing to explain how they are synthesised and where they come from.
Collapse
Affiliation(s)
- Sebastien Apcher
- Institut Gustave Roussy, Université Paris Sud, UMR 1015, Villejuif, France
| | - Borek Vojtesek
- RECAMO, Masaryk Memorial Cancer Institute, Zluty kopec 7, 65653 Brno, Czech Republic
| | - Robin Fahraeus
- Inserm UMRS1131, Institut de Génétique Moléculaire, Université Paris 7, Hôpital St. Louis, France; Department of Medical Biosciences, Building 6M, Umeå University, 901 85 Umeå, Sweden; RECAMO, Masaryk Memorial Cancer Institute, Zluty kopec 7, 65653 Brno, Czech Republic.
| |
Collapse
|
7
|
Galluzzi L, Kepp O, Hett E, Kroemer G, Marincola FM. Immunogenic cell death in cancer: concept and therapeutic implications. J Transl Med 2023; 21:162. [PMID: 36864446 PMCID: PMC9979428 DOI: 10.1186/s12967-023-04017-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/08/2023] [Indexed: 03/04/2023] Open
Abstract
Mammalian cells responding to specific perturbations of homeostasis can undergo a regulated variant of cell death that elicits adaptive immune responses. As immunogenic cell death (ICD) can only occur in a precise cellular and organismal context, it should be conceptually differentiated from instances of immunostimulation or inflammatory responses that do not mechanistically depend on cellular demise. Here, we critically discuss key conceptual and mechanistic aspects of ICD and its implications for cancer (immuno)therapy.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA. .,Sandra and Edward Meyer Cancer Center, New York, NY, USA. .,Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Erik Hett
- Sonata Therapeutics, Boston, MA, USA
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France.,Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | | |
Collapse
|
8
|
Ahn R, Cui Y, White FM. Antigen discovery for the development of cancer immunotherapy. Semin Immunol 2023; 66:101733. [PMID: 36841147 DOI: 10.1016/j.smim.2023.101733] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023]
Abstract
Central to successful cancer immunotherapy is effective T cell antitumor immunity. Multiple targeted immunotherapies engineered to invigorate T cell-driven antitumor immunity rely on identifying the repertoire of T cell antigens expressed on the tumor cell surface. Mass spectrometry-based survey of such antigens ("immunopeptidomics") combined with other omics platforms and computational algorithms has been instrumental in identifying and quantifying tumor-derived T cell antigens. In this review, we discuss the types of tumor antigens that have emerged for targeted cancer immunotherapy and the immunopeptidomics methods that are central in MHC peptide identification and quantification. We provide an overview of the strength and limitations of mass spectrometry-driven approaches and how they have been integrated with other technologies to discover targetable T cell antigens for cancer immunotherapy. We highlight some of the emerging cancer immunotherapies that successfully capitalized on immunopeptidomics, their challenges, and mass spectrometry-based strategies that can support their development.
Collapse
Affiliation(s)
- Ryuhjin Ahn
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yufei Cui
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Forest M White
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
9
|
Alarcon NO, Jaramillo M, Mansour HM, Sun B. Therapeutic Cancer Vaccines—Antigen Discovery and Adjuvant Delivery Platforms. Pharmaceutics 2022; 14:pharmaceutics14071448. [PMID: 35890342 PMCID: PMC9325128 DOI: 10.3390/pharmaceutics14071448] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 12/15/2022] Open
Abstract
For decades, vaccines have played a significant role in protecting public and personal health against infectious diseases and proved their great potential in battling cancers as well. This review focused on the current progress of therapeutic subunit vaccines for cancer immunotherapy. Antigens and adjuvants are key components of vaccine formulations. We summarized several classes of tumor antigens and bioinformatic approaches of identification of tumor neoantigens. Pattern recognition receptor (PRR)-targeting adjuvants and their targeted delivery platforms have been extensively discussed. In addition, we emphasized the interplay between multiple adjuvants and their combined delivery for cancer immunotherapy.
Collapse
Affiliation(s)
- Neftali Ortega Alarcon
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
| | - Maddy Jaramillo
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
| | - Heidi M. Mansour
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
- The University of Arizona Cancer Center, Tucson, AZ 85721, USA
- Department of Medicine, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Bo Sun
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
- The University of Arizona Cancer Center, Tucson, AZ 85721, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
- Correspondence: ; Tel.: +1-520-621-6420
| |
Collapse
|
10
|
Apcher S, Tovar-Fernadez M, Ducellier S, Thermou A, Nascimento M, Sroka E, Fahraeus R. mRNA translation from an antigen presentation perspective: A tribute to the works of Nilabh Shastri. Mol Immunol 2021; 141:305-308. [PMID: 34920325 DOI: 10.1016/j.molimm.2021.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/04/2021] [Accepted: 12/07/2021] [Indexed: 10/19/2022]
Abstract
The field of mRNA translation has witnessed an impressive expansion in the last decade. The once standard model of translation initiation has undergone, and is still undergoing, a major overhaul, partly due to more recent technical advancements detailing, for example, initiation at non-AUG codons. However, some of the pioneering works in this area have come from immunology and more precisely from the field of antigen presentation to the major histocompatibility class I (MHC-I) pathway. Despite early innovative studies from the lab of Nilabh Shastri demonstrating alternative mRNA translation initiation as a source for MHC-I peptide substrates, the mRNA translation field did not include these into their models. It was not until the introduction of the ribo-sequence technique that the extent of non-canonical translation initiation became widely acknowledged. The detection of peptides on MHC-I molecules by CD8 + T cells is extremely sensitive, making this a superior model system for studying alternative mRNA translation initiation from specific mRNAs. In view of this, we give a brief history on alternative initiation from an immunology perspective and its fundamental role in allowing the immune system to distinguish self from non-self and at the same time pay tribute to the works of Nilabh Shastri.
Collapse
Affiliation(s)
- Sebastien Apcher
- Université Paris-Saclay, Institut Gustave Roussy, Inserm UMR1015, Immunologie des tumeurs et Immunothérapie contre le cancer, 94805, Villejuif, France.
| | - Maria Tovar-Fernadez
- ICCVS, University of Gdańsk, Science, ul. Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Sarah Ducellier
- Université Paris-Saclay, Institut Gustave Roussy, Inserm UMR1015, Immunologie des tumeurs et Immunothérapie contre le cancer, 94805, Villejuif, France
| | - Aikaterini Thermou
- ICCVS, University of Gdańsk, Science, ul. Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Megane Nascimento
- Université Paris-Saclay, Institut Gustave Roussy, Inserm UMR1015, Immunologie des tumeurs et Immunothérapie contre le cancer, 94805, Villejuif, France
| | - Ewa Sroka
- ICCVS, University of Gdańsk, Science, ul. Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Robin Fahraeus
- ICCVS, University of Gdańsk, Science, ul. Wita Stwosza 63, 80-308, Gdańsk, Poland; Inserm UMRS1131, Institut de Génétique Moléculaire, Université Paris 7, Hôpital St. Louis, F-75010, Paris, France; RECAMO, Masaryk Memorial Cancer Institute, Zluty kopec 7, 65653, Brno, Czech Republic; Department of Medical Biosciences, Building 6M, Umeå University, 901 85, Umeå, Sweden.
| |
Collapse
|
11
|
Joyce S, Ternette N. Know thy immune self and non-self: Proteomics informs on the expanse of self and non-self, and how and where they arise. Proteomics 2021; 21:e2000143. [PMID: 34310018 PMCID: PMC8865197 DOI: 10.1002/pmic.202000143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/30/2021] [Accepted: 07/19/2021] [Indexed: 12/30/2022]
Abstract
T cells play an important role in the adaptive immune response to a variety of infections and cancers. Initiation of a T cell mediated immune response requires antigen recognition in a process termed MHC (major histocompatibility complex) restri ction. A T cell antigen is a composite structure made up of a peptide fragment bound within the antigen-binding groove of an MHC-encoded class I or class II molecule. Insight into the precise composition and biology of self and non-self immunopeptidomes is essential to harness T cell mediated immunity to prevent, treat, or cure infectious diseases and cancers. T cell antigen discovery is an arduous task! The pioneering work in the early 1990s has made large-scale T cell antigen discovery possible. Thus, advancements in mass spectrometry coupled with proteomics and genomics technologies make possible T cell antigen discovery with ease, accuracy, and sensitivity. Yet we have only begun to understand the breadth and the depth of self and non-self immunopeptidomes because the molecular biology of the cell continues to surprise us with new secrets directly related to the source, and the processing and presentation of MHC ligands. Focused on MHC class I molecules, this review, therefore, provides a brief historic account of T cell antigen discovery and, against a backdrop of key advances in molecular cell biologic processes, elaborates on how proteogenomics approaches have revolutionised the field.
Collapse
Affiliation(s)
- Sebastian Joyce
- Department of Veterans AffairsTennessee Valley Healthcare System and the Department of PathologyMicrobiology and ImmunologyVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Nicola Ternette
- Centre for Cellular and Molecular PhysiologyNuffield Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
12
|
Davis L, Tarduno A, Lu YC. Neoantigen-Reactive T Cells: The Driving Force behind Successful Melanoma Immunotherapy. Cancers (Basel) 2021; 13:cancers13236061. [PMID: 34885172 PMCID: PMC8657037 DOI: 10.3390/cancers13236061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Cancer immunotherapy is a revolutionary type of cancer therapy. It uses the patient’s own immune system to fight and potentially cure cancer. The first major breakthrough of immunotherapy came from successful clinical trials for melanoma treatments. Since then, researchers have focused on understanding the science behind immunotherapy, so that patients with other types of cancer may also benefit. One of the major findings is that the T cells in melanoma patients may recognize a specific type of tumor antigen, called neoantigens, and then kill tumor cells that present these neoantigens. The neoantigens mainly arise from the DNA mutations found in tumor cells. These mutations are translated into mutated proteins that are then distinguished by T cells. In this article, we discuss the critical role of T cells in immunotherapy, as well as the clinical trials that shaped the treatments for melanoma. Abstract Patients with metastatic cutaneous melanoma have experienced significant clinical responses after checkpoint blockade immunotherapy or adoptive cell therapy. Neoantigens are mutated proteins that arise from tumor-specific mutations. It is hypothesized that the neoantigen recognition by T cells is the critical step for T-cell-mediated anti-tumor responses and subsequent tumor regressions. In addition to describing neoantigens, we review the sentinel and ongoing clinical trials that are helping to shape the current treatments for patients with cutaneous melanoma. We also present the existing evidence that establishes the correlations between neoantigen-reactive T cells and clinical responses in melanoma immunotherapy.
Collapse
Affiliation(s)
- Lindy Davis
- Department of Surgery, Albany Medical Center, Albany, NY 12208, USA; (L.D.); (A.T.)
| | - Ashley Tarduno
- Department of Surgery, Albany Medical Center, Albany, NY 12208, USA; (L.D.); (A.T.)
| | - Yong-Chen Lu
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence:
| |
Collapse
|
13
|
Implications of Antigen Selection on T Cell-Based Immunotherapy. Pharmaceuticals (Basel) 2021; 14:ph14100993. [PMID: 34681217 PMCID: PMC8537967 DOI: 10.3390/ph14100993] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 12/15/2022] Open
Abstract
Many immunotherapies rely on CD8+ effector T cells to recognize and kill cognate tumor cells. These T cell-based immunotherapies include adoptive cell therapy, such as CAR T cells or transgenic TCR T cells, and anti-cancer vaccines which expand endogenous T cell populations. Tumor mutation burden and the choice of antigen are among the most important aspects of T cell-based immunotherapies. Here, we highlight various classes of cancer antigens, including self, neojunction-derived, human endogenous retrovirus (HERV)-derived, and somatic nucleotide variant (SNV)-derived antigens, and consider their utility in T cell-based immunotherapies. We further discuss the respective anti-tumor/anti-self-properties that influence both the degree of immunotolerance and potential off-target effects associated with each antigen class.
Collapse
|
14
|
Mahnke YD, Devevre E, Baumgaertner P, Matter M, Rufer N, Romero P, Speiser DE. Human melanoma-specific CD8(+) T-cells from metastases are capable of antigen-specific degranulation and cytolysis directly ex vivo. Oncoimmunology 2021; 1:467-530. [PMID: 22754765 PMCID: PMC3382891 DOI: 10.4161/onci.19856] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The relatively low frequencies of tumor Ag-specific T-cells in PBMC and metastases from cancer patients have long precluded the analysis of their direct ex vivo cytolytic capacity. Using a new composite technique that works well with low cell numbers, we aimed at determining the functional competence of melanoma-specific CD8+ T-cells. A multiparameter flow cytometry based technique was applied to assess the cytolytic function, degranulation and IFNγ production by tumor Ag-specific CD8+ T-cells from PBMC and tumor-infiltrated lymph nodes (TILN) of melanoma patients. We found strong cytotoxicity by T-cells not only when they were isolated from PBMC but also from TILN. Cytotoxicity was observed against peptide-pulsed target cells and melanoma cells presenting the naturally processed endogenous antigen. However, unlike their PBMC-derived counterparts, T-cells from TILN produced only minimal amounts of IFNγ, while exhibiting similar levels of degranulation, revealing a critical functional dichotomy in metastatic lesions. Our finding of partial functional impairment fits well with the current knowledge that T-cells from cancer metastases are so-called exhausted, a state of T-cell hyporesponsiveness also found in chronic viral infections. The identification of responsible mechanisms in the tumor microenvironment is important for improving cancer therapies.
Collapse
Affiliation(s)
- Yolanda D Mahnke
- Ludwig Center for Cancer Research; University of Lausanne; Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
15
|
Leko V, Rosenberg SA. Identifying and Targeting Human Tumor Antigens for T Cell-Based Immunotherapy of Solid Tumors. Cancer Cell 2020; 38:454-472. [PMID: 32822573 PMCID: PMC7737225 DOI: 10.1016/j.ccell.2020.07.013] [Citation(s) in RCA: 268] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 12/20/2022]
Abstract
Cancer elimination in humans can be achieved with immunotherapy that relies on T lymphocyte-mediated recognition of tumor antigens. Several types of these antigens have been recognized based on their cellular origins and expression patterns, while their detection has been greatly facilitated by recent achievements in next-generation sequencing and immunopeptidomics. Some of them have been targeted in clinical trials with various immunotherapy approaches, while many others remain untested. Here, we discuss molecular identification of different tumor antigen types, and the clinical safety and efficacy of targeting them with immunotherapy. Additionally, we suggest strategies to increase the efficacy and availability of antigen-directed immunotherapies for treatment of patients with metastatic cancer.
Collapse
Affiliation(s)
- Vid Leko
- Surgery Branch, National Cancer Institute, National Institutes of Health, Building 10-CRC, Room 3-3942, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Steven A Rosenberg
- Surgery Branch, National Cancer Institute, National Institutes of Health, Building 10-CRC, Room 3-3942, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Boulpicante M, Darrigrand R, Pierson A, Salgues V, Rouillon M, Gaudineau B, Khaled M, Cattaneo A, Bachi A, Cascio P, Apcher S. Tumors escape immunosurveillance by overexpressing the proteasome activator PSME3. Oncoimmunology 2020; 9:1761205. [PMID: 32923122 PMCID: PMC7458623 DOI: 10.1080/2162402x.2020.1761205] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/03/2020] [Indexed: 11/09/2022] Open
Abstract
The success of CD8+ T cell-based cancer immunotherapy emphasizes the importance of understanding the mechanisms of generation of MHC-I peptide ligands and the possible pathways of tumor cell escape from immunosurveillance. Recently, we showed that peptides generated in the nucleus during a pioneer round of mRNA translation (pioneer translation products, or PTPs) are an important source of tumor specific peptides which correlates with the aberrant splicing and transcription events associated with oncogenesis. Here we show that up-regulation of PSME3 proteasome activator in cancer cells results in increased destruction of PTP-derived peptides in the nucleus thus enabling cancer cell to subvert immunosurveillance. These findings unveil a previously unexpected role for PSME3 in antigen processing and identify PSME3 as a druggable target to improve the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Mathilde Boulpicante
- Immunologie des Tumeurs et Immunothérapie, Université Paris-Saclay, Institut Gustave Roussy, Inserm, Villejuif, France
| | - Romain Darrigrand
- Immunologie des Tumeurs et Immunothérapie, Université Paris-Saclay, Institut Gustave Roussy, Inserm, Villejuif, France
| | - Alison Pierson
- Immunologie des Tumeurs et Immunothérapie, Université Paris-Saclay, Institut Gustave Roussy, Inserm, Villejuif, France
| | - Valérie Salgues
- Immunologie des Tumeurs et Immunothérapie, Université Paris-Saclay, Institut Gustave Roussy, Inserm, Villejuif, France
| | - Marine Rouillon
- Immunologie des Tumeurs et Immunothérapie, Université Paris-Saclay, Institut Gustave Roussy, Inserm, Villejuif, France
| | - Benoit Gaudineau
- Dynamique des Cellules Tumorales, Université Paris-Saclay, Institut Gustave Roussy, Inserm, Villejuif, France
| | - Mehdi Khaled
- Dynamique des Cellules Tumorales, Université Paris-Saclay, Institut Gustave Roussy, Inserm, Villejuif, France
| | - Angela Cattaneo
- IFOM, The FIRC Institute of Molecular Oncology, Milano, Italy
| | - Angela Bachi
- IFOM, The FIRC Institute of Molecular Oncology, Milano, Italy
| | - Paolo Cascio
- Department of Veterinary Sciences, University of Turin, 10095, Grugliasco, Turin, Italy
| | - Sébastien Apcher
- Immunologie des Tumeurs et Immunothérapie, Université Paris-Saclay, Institut Gustave Roussy, Inserm, Villejuif, France
| |
Collapse
|
17
|
Janelle V, Rulleau C, Del Testa S, Carli C, Delisle JS. T-Cell Immunotherapies Targeting Histocompatibility and Tumor Antigens in Hematological Malignancies. Front Immunol 2020; 11:276. [PMID: 32153583 PMCID: PMC7046834 DOI: 10.3389/fimmu.2020.00276] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/03/2020] [Indexed: 12/19/2022] Open
Abstract
Over the last decades, T-cell immunotherapy has revealed itself as a powerful, and often curative, strategy to treat blood cancers. In hematopoietic cell transplantation, most of the so-called graft-vs.-leukemia (GVL) effect hinges on the recognition of histocompatibility antigens that reflect immunologically relevant genetic variants between donors and recipients. Whether other variants acquired during the neoplastic transformation, or the aberrant expression of gene products can yield antigenic targets of similar relevance as the minor histocompatibility antigens is actively being pursued. Modern genomics and proteomics have enabled the high throughput identification of candidate antigens for immunotherapy in both autologous and allogeneic settings. As such, these major histocompatibility complex-associated tumor-specific (TSA) and tumor-associated antigens (TAA) can allow for the targeting of multiple blood neoplasms, which is a limitation for other immunotherapeutic approaches, such as chimeric antigen receptor (CAR)-modified T cells. We review the current strategies taken to translate these discoveries into T-cell therapies and propose how these could be introduced in clinical practice. Specifically, we discuss the criteria that are used to select the antigens with the greatest therapeutic value and we review the various T-cell manufacturing approaches in place to either expand antigen-specific T cells from the native repertoire or genetically engineer T cells with minor histocompatibility antigen or TSA/TAA-specific recombinant T-cell receptors. Finally, we elaborate on the current and future incorporation of these therapeutic T-cell products into the treatment of hematological malignancies.
Collapse
Affiliation(s)
- Valérie Janelle
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Caroline Rulleau
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Simon Del Testa
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Cédric Carli
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Jean-Sébastien Delisle
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Médecine, Université de Montréal, Montréal, QC, Canada.,Division Hématologie et Oncologie, Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| |
Collapse
|
18
|
Samimi M, Benlalam H, Aumond P, Gaboriaud P, Fradin D, Kervarrec T, Florenceau L, Vignard V, Blom A, Touzé A, Gervois N, Labarriere N. Viral and tumor antigen-specific CD8 T-cell responses in Merkel cell carcinoma. Cell Immunol 2019; 344:103961. [PMID: 31472938 DOI: 10.1016/j.cellimm.2019.103961] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 07/31/2019] [Indexed: 12/23/2022]
Abstract
Merkel cell carcinoma (MCC) is a rare and aggressive cutaneous cancer, which is immunogenic, regardless of the presence of MCPyV (80% of cases). The identification of MCC-specific epitopes recognized by CD8 T cells is crucial to expand the arsenal of immunotherapeutic treatments. Until now, most efforts focused on the identification of virus-specific epitopes, whereas immune responses directed against shared cellular tumor-specific antigens have not been evidenced. In this study, we measured T-cell responses against viral (n = 3) and tumor antigens (n = 47) from TILs derived from 21 MCC tumors. Virus-specific CD8 T-cell responses dominated MCC-specific immune responses, and we identified two new HLA-peptide complexes derived from the LT antigen, located in a region encompassing 3 previously identified epitopes. Finally, we show that MAGE-A3 antigen, frequently expressed by MCC tumors, was recognized by CD8 TILs from a virus-negative MCC tumor and thus could be a target for immunotherapy in this setting.
Collapse
Affiliation(s)
- Mahtab Samimi
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France; Laboratoire "Biologie des infections à polyomavirus", ISP1282 INRA Université de Tours, France; Dermatology Department, University of Tours, CHU Tours, Tours, France
| | - Houssem Benlalam
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.
| | - Pascal Aumond
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Pauline Gaboriaud
- Laboratoire "Biologie des infections à polyomavirus", ISP1282 INRA Université de Tours, France
| | - Delphine Fradin
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Thibault Kervarrec
- Laboratoire "Biologie des infections à polyomavirus", ISP1282 INRA Université de Tours, France; Pathology Department, University of Tours, CHU Tours, Tours, France
| | - Laetitia Florenceau
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Virginie Vignard
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France; CHU Nantes, Nantes, France
| | - Astrid Blom
- Dermatology Department, Hôpital Ambroise Paré, Paris, France
| | - Antoine Touzé
- Laboratoire "Biologie des infections à polyomavirus", ISP1282 INRA Université de Tours, France
| | - Nadine Gervois
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Nathalie Labarriere
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| |
Collapse
|
19
|
Garcia-Garijo A, Fajardo CA, Gros A. Determinants for Neoantigen Identification. Front Immunol 2019; 10:1392. [PMID: 31293573 PMCID: PMC6601353 DOI: 10.3389/fimmu.2019.01392] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/03/2019] [Indexed: 12/22/2022] Open
Abstract
All tumors accumulate genetic alterations, some of which can give rise to mutated, non-self peptides presented by human leukocyte antigen (HLA) molecules and elicit T-cell responses. These immunogenic mutated peptides, or neoantigens, are foreign in nature and display exquisite tumor specificity. The correlative evidence suggesting they play an important role in the effectiveness of various cancer immunotherapies has triggered the development of vaccines and adoptive T-cell therapies targeting them. However, the systematic identification of personalized neoantigens in cancer patients, a critical requisite for the success of these therapies, remains challenging. A growing amount of evidence supports that only a small fraction of all tumor somatic non-synonymous mutations (NSM) identified represent bona fide neoantigens; mutated peptides that are processed, presented on the cell surface HLA molecules of cancer cells and are capable of triggering immune responses in patients. Here, we provide an overview of the existing strategies to identify candidate neoantigens and to evaluate their immunogenicity, two factors that impact on neoantigen identification. We will focus on their strengths and limitations to allow readers to rationally select and apply the most suitable method for their specific laboratory setting.
Collapse
Affiliation(s)
| | | | - Alena Gros
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
20
|
Laumont CM, Perreault C. Exploiting non-canonical translation to identify new targets for T cell-based cancer immunotherapy. Cell Mol Life Sci 2018; 75:607-621. [PMID: 28823056 PMCID: PMC11105255 DOI: 10.1007/s00018-017-2628-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/03/2017] [Accepted: 08/16/2017] [Indexed: 01/11/2023]
Abstract
Cryptic MHC I-associated peptides (MAPs) are produced via two mechanisms: translation of protein-coding genes in non-canonical reading frames and translation of allegedly non-coding sequences. In general, cryptic MAPs are coded by relatively short open reading frames whose translation can be regulated at the level of initiation, elongation or termination. In contrast to conventional MAPs, the processing of cryptic MAPs is frequently proteasome independent. The existence of cryptic MAPs derived from allegedly non-coding regions enlarges the scope of CD8 T cell immunosurveillance from a mere ~2% to as much as ~75% of the human genome. Considering that 99% of cancer-specific mutations are located in those allegedly non-coding regions, cryptic MAPs could furthermore represent a particularly rich source of tumor-specific antigens. However, extensive proteogenomic analyses will be required to determine the breath as well as the temporal and spatial plasticity of the cryptic MAP repertoire in normal and neoplastic cells.
Collapse
Affiliation(s)
- Céline M Laumont
- Institute for Research in Immunology and Cancer, Université de Montréal, Station Centre-Ville, PO Box 6128, Montreal, QC, H3C 3J7, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Station Centre-Ville, PO Box 6128, Montreal, QC, H3C 3J7, Canada
| | - Claude Perreault
- Institute for Research in Immunology and Cancer, Université de Montréal, Station Centre-Ville, PO Box 6128, Montreal, QC, H3C 3J7, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Station Centre-Ville, PO Box 6128, Montreal, QC, H3C 3J7, Canada.
- Division of Hematology, Hôpital Maisonneuve-Rosemont, 5415 de l'Assomption Boulevard, Montreal, QC, H1T 2M4, Canada.
| |
Collapse
|
21
|
Abstract
Harnessing the immune system to eradicate malignant cells is becoming a most powerful new approach to cancer therapy. FDA approval of the immunotherapy-based drugs, sipuleucel-T (Provenge), ipilimumab (Yervoy, anti-CTLA-4), and more recently, the programmed cell death (PD)-1 antibody (pembrolizumab, Keytruda), for the treatment of multiple types of cancer has greatly advanced research and clinical studies in the field of cancer immunotherapy. Furthermore, recent clinical trials, using NY-ESO-1-specific T cell receptor (TCR) or CD19-chimeric antigen receptor (CAR), have shown promising clinical results for patients with metastatic cancer. Current success of cancer immunotherapy is built upon the work of cancer antigens and co-inhibitory signaling molecules identified 20 years ago. Among the large numbers of target antigens, CD19 is the best target for CAR T cell therapy for blood cancer, but CAR-engineered T cell immunotherapy does not yet work in solid cancer. NY-ESO-1 is one of the best targets for TCR-based immunotherapy in solid cancer. Despite the great success of checkpoint blockade therapy, more than 50% of cancer patients fail to respond to blockade therapy. The advent of new technologies such as next-generation sequencing has enhanced our ability to search for new immune targets in onco-immunology and accelerated the development of immunotherapy with potentially broader coverage of cancer patients. In this review, we will discuss the recent progresses of cancer immunotherapy and novel strategies in the identification of new immune targets and mutation-derived antigens (neoantigens) for cancer immunotherapy and immunoprecision medicine.
Collapse
Affiliation(s)
- Rong-Fu Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
- Institute of Biosciences and Technology, College of Medicine, Texas A & M University, Houston, Texas 77030, USA
| | - Helen Y Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
22
|
Lopez M, Ghidouche A, Rochas C, Godelaine D, Carrasco J, Colau D, Hames G, Montero-Julian FA, Coulie PG, Olive D. Identification of a naturally processed HLA-A*02:01-restricted CTL epitope from the human tumor-associated antigen Nectin-4. Cancer Immunol Immunother 2016; 65:1177-88. [PMID: 27514672 PMCID: PMC11029526 DOI: 10.1007/s00262-016-1877-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 07/25/2016] [Indexed: 10/21/2022]
Abstract
Nectin-4 is a tumor antigen present on the surface of breast, ovarian and lung carcinoma cells. It is rarely present in normal adult tissues and is therefore a candidate target for cancer immunotherapy. Here, we identified a Nectin-4 antigenic peptide that is naturally presented to T cells by HLA-A2 molecules. We first screened the 502 nonamer peptides of Nectin-4 (510 amino acids) for binding to and off-rate from eight different HLA class I molecules. We then combined biochemical, cellular and algorithmic assays to select 5 Nectin-4 peptides that bound to HLA-A*02:01 molecules. Cytolytic T lymphocytes were obtained from healthy donors, that specifically lyzed HLA-A2(+) cells pulsed with 2 out of the 5 peptides, indicating the presence of anti-Nectin-4 CD8(+) T lymphocytes in the human T cell repertoire. Finally, an HLA-A2-restricted cytolytic T cell clone derived from a breast cancer patient recognized peptide Nectin-4145-153 (VLVPPLPSL) and lyzed HLA-A2(+) Nectin-4(+) breast carcinoma cells. These results indicate that peptide Nectin-4145-153 is naturally processed for recognition by T cells on HLA-A2 molecules. It could be used to monitor antitumor T cell responses or to immunize breast cancer patients.
Collapse
Affiliation(s)
- Marc Lopez
- Inserm U1068, Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Oncologie Moléculaire, UM 105, Institut Paoli-Calmettes, CNRS, UMR7258, Aix-Marseille University, Marseille, France
| | - Abderrezak Ghidouche
- Inserm U1068, Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Immunité et Cancer, UM 105, Institut Paoli-Calmettes, CNRS, UMR7258, Aix-Marseille University, 27 bd Lei Roure, 13009, Marseille, France
| | - Caroline Rochas
- Inserm U1068, Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Immunité et Cancer, UM 105, Institut Paoli-Calmettes, CNRS, UMR7258, Aix-Marseille University, 27 bd Lei Roure, 13009, Marseille, France
| | - Danièle Godelaine
- Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Javier Carrasco
- Department of Oncology and Hematology, Grand Hôpital de Charleroi, Charleroi, Belgium
| | - Didier Colau
- Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Gérald Hames
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | | | - Pierre G Coulie
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Daniel Olive
- Inserm U1068, Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Immunité et Cancer, UM 105, Institut Paoli-Calmettes, CNRS, UMR7258, Aix-Marseille University, 27 bd Lei Roure, 13009, Marseille, France.
| |
Collapse
|
23
|
Duvallet E, Boulpicante M, Yamazaki T, Daskalogianni C, Prado Martins R, Baconnais S, Manoury B, Fahraeus R, Apcher S. Exosome-driven transfer of tumor-associated Pioneer Translation Products (TA-PTPs) for the MHC class I cross-presentation pathway. Oncoimmunology 2016; 5:e1198865. [PMID: 27757298 PMCID: PMC5048765 DOI: 10.1080/2162402x.2016.1198865] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/28/2016] [Accepted: 05/31/2016] [Indexed: 12/18/2022] Open
Abstract
Cellular immune reactions against non-self-epitopes require activation of cytotoxic CD8+ T-cells via cross-presentation of MHC class I-restricted peptides by professional antigen presenting cells (pAPCs), with the consequent detection and elimination of cells expressing the same antigens via the endogenous (direct) pathway. The source of peptides for the endogenous pathway is constituted of alternative mRNA translation products; however, it is still unclear which source of peptides is used for cross-presentation. Furthermore, the presentation of non-canonical translation products, produced during a non-conventional translation event, on class I molecules of tumor cells has been reported but how these peptides are generated, presented to pAPCs, and their capacity to stimulate CD8+ T cells is still not known. Here, we report that pioneer translation peptides (PTPs) derived from intron or exon pre-mRNAs can serve as tumor-associated antigens (TA-PTPs) and are delivered from the producing tumor cells to pAPCs via exosomes where they are processed by the cytosolic pathway. Injection of TA-PTPs and tumor-derived exosomes efficiently induce CD8+ T-cell proliferation and prevent tumor growth in mice. Our results show that TA-PTPs represent an efficient source of antigenic peptides for CD8+ T cell activation and that full-length proteins are not required for cross-presentation. These findings can have interesting implications for generating tolerance and for designing vectors to generate vaccines.
Collapse
Affiliation(s)
- Emilie Duvallet
- Institut Gustave Roussy, Université Paris Sud, Université Paris Saclay, Unité 1015 département d'immunologie , Villejuif, France
| | - Mathilde Boulpicante
- Institut Gustave Roussy, Université Paris Sud, Université Paris Saclay, Unité 1015 département d'immunologie , Villejuif, France
| | - Takahiro Yamazaki
- Institut Gustave Roussy, Université Paris Sud, Université Paris Saclay, Unité 1015 département d'immunologie , Villejuif, France
| | - Chrysoula Daskalogianni
- Equipe Labellisée la Ligue Contre le Cancer, Inserm UMR1162, Université Paris 7, Institut de Génétique Moléculaire , Paris, France and RECAMO, Masaryk Memorial Cancer Institute , Brno, Czech Republic
| | - Rodrigo Prado Martins
- Equipe Labellisée la Ligue Contre le Cancer, Inserm UMR1162, Université Paris 7, Institut de Génétique Moléculaire , Paris, France and RECAMO, Masaryk Memorial Cancer Institute , Brno, Czech Republic
| | - Sonia Baconnais
- Signalisations, Noyaux et Innovations en Cancérologie, CNRS UMR8126, Université Paris Sud, Université Paris Saclay , Villejuif, France
| | - Bénédicte Manoury
- INEM, U1151-CNRS UMR8253 , Paris, France and Université Paris Descartes, Sorbonne Paris Cité, Faculté de medicine , Paris, France
| | - Robin Fahraeus
- Equipe Labellisée la Ligue Contre le Cancer, Inserm UMR1162, Université Paris 7, Institut de Génétique Moléculaire , Paris, France and RECAMO, Masaryk Memorial Cancer Institute , Brno, Czech Republic
| | - Sébastien Apcher
- Institut Gustave Roussy, Université Paris Sud, Université Paris Saclay, Unité 1015 département d'immunologie , Villejuif, France
| |
Collapse
|
24
|
Norbury CC. Defining cross presentation for a wider audience. Curr Opin Immunol 2016; 40:110-6. [DOI: 10.1016/j.coi.2016.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 04/03/2016] [Indexed: 01/10/2023]
|
25
|
Apcher S, Prado Martins R, Fåhraeus R. The source of MHC class I presented peptides and its implications. Curr Opin Immunol 2016; 40:117-22. [PMID: 27105144 DOI: 10.1016/j.coi.2016.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/04/2016] [Accepted: 04/03/2016] [Indexed: 10/21/2022]
Abstract
The source of peptides that enter the major histocompatibility class I (MHCI) pathway has been intensively debated over the last two decades. The initial assumption that peptides are derived from degradation of full length proteins was challenged by a model in which alternative translation products are a source of peptides. This model has been tested and supported by scientific data. We now need new hypotheses on the physiological implications of different sources of peptides for the MHCI pathway. The aim of this overview is to give an up-to-date account of the source of antigenic peptide material for the MHCI pathway and to incorporate the more recent observations of alternative mRNA translation products into existing models of the direct and cross-presentation pathways.
Collapse
Affiliation(s)
- Sébastien Apcher
- Institut Gustave Roussy, Université Paris Sud, Unité 1015 département d'immunologie, 114, rue Edouard Vaillant, 94805 Villejuif, France
| | - Rodrigo Prado Martins
- Equipe Labellisée la Ligue Contre le Cancer, Inserm UMR1162, Université Paris 7, Institut de Génétique Moléculaire, 27 rue Juliette Dodu, 75010 Paris, France
| | - Robin Fåhraeus
- Equipe Labellisée la Ligue Contre le Cancer, Inserm UMR1162, Université Paris 7, Institut de Génétique Moléculaire, 27 rue Juliette Dodu, 75010 Paris, France; RECAMO, Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53 Brno, Czech Republic; Department of Medical Biosciences, Umeå University, SE-90185 Umeå, Sweden.
| |
Collapse
|
26
|
Pfirschke C, Gebhardt C, Zörnig I, Pritsch M, Eichmüller SB, Jäger D, Enk A, Beckhove P. T cell responses in early-stage melanoma patients occur frequently and are not associated with humoral response. Cancer Immunol Immunother 2015; 64:1369-81. [PMID: 26160687 PMCID: PMC11028448 DOI: 10.1007/s00262-015-1739-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 06/20/2015] [Indexed: 11/26/2022]
Abstract
Endogenous tumor-specific T cells are detectable in patients with different tumor types including malignant melanoma (MM). They can control tumor growth, have impact on patient survival and correlate with improved clinical response to immune checkpoint therapy. Thus, they may represent a potent biomarker for respective treatment decisions. So far, major target antigens of endogenous MM-reactive T cells have not been determined systematically. Instead, autoantibodies are discussed as surrogate parameter for MM-specific T cells. Throughout a period of more than 60 days after tumor resection, we therefore determined in 38 non-metastasized primary MM patients and in healthy individuals by IFNγ ELISpot and bead-based fluorescent multiplex assay major target antigens of spontaneous T cell and humoral responses using a broad panel of MM antigens and assessed the presence and suppressive impact of MM-reactive regulatory T cells (Tregs). We show that MM-reactive T cells are frequent in MM patients, transiently increase after tumor removal and are mostly directed against Melan-A/MART-1, Tyrosinase, NA17-A and p53. MM-specific Tregs were only detected in few patients and inhibited MM-reactive T cells particularly early after tumor resection. Tumor-specific autoantibodies occurred in most patients, but did not correlate with T cell responses. Thus, endogenous antibodies may not be reliable surrogate parameters of MM-reactive T cells.
Collapse
Affiliation(s)
- Christina Pfirschke
- Division of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Christoffer Gebhardt
- Skin Cancer Unit, DKFZ, Heidelberg, Germany
- Department of Dermatology, Ruperto-Carola University of Heidelberg, Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruperto-Carola University of Heidelberg, Mannheim, Germany
| | - Inka Zörnig
- Department of Medical Oncology, NCT, Heidelberg University Hospital, Heidelberg, Germany
| | - Maria Pritsch
- Division of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Stefan B Eichmüller
- Division of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, NCT, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander Enk
- Department of Dermatology, Ruperto-Carola University of Heidelberg, Heidelberg, Germany
| | - Philipp Beckhove
- Division of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.
- Regensburg Center for Interventional Immunology (RCI), University of Regensburg, Regensburg, Germany.
- University Clinic of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
27
|
Abstract
DNA vaccination has emerged as an attractive immunotherapeutic approach against cancer
due to its simplicity, stability, and safety. Results from numerous clinical trials have
demonstrated that DNA vaccines are well tolerated by patients and do not trigger major
adverse effects. DNA vaccines are also very cost effective and can be administered
repeatedly for long-term protection. Despite all the practical advantages, DNA vaccines
face challenges in inducing potent antigen specific cellular immune responses as a result
of immune tolerance against endogenous self-antigens in tumors. Strategies to enhance
immunogenicity of DNA vaccines against self-antigens have been investigated including
encoding of xenogeneic versions of antigens, fusion of antigens to molecules that activate
T cells or trigger associative recognition, priming with DNA vectors followed by boosting
with viral vector, and utilization of immunomodulatory molecules. This review will focus
on discussing strategies that circumvent immune tolerance and provide updates on findings
from recent clinical trials.
Collapse
Key Words
- APCs, antigen presenting cells
- CEA, carcinoembryonic antigen
- CIN, cervical intraepithelial neoplasia
- CT antigens, cancer-testis antigens
- CTLs, cytotoxic lymphocytes
- DNA vaccines
- DOM, fragment c domain
- EP, electroporation
- GITR, glucocorticoid-induced tumor necrosis factor receptor family-related genes
- HER2, Her2/neu
- HSP70, heat shock protein 70
- IFNs, interferons
- IRF, interferon regulatory factor
- Id, idiotype
- MHC, major histocompatibility complex
- Mam-A, Mammaglobin-A
- NHP, non-human primate
- PAP, Prostatic acid phosphatase
- PMED, particle mediated epidermal delivery
- PSMA, prostate-specific membrane antigen
- SCT, single-chain trimer
- STING, stimulator of interferon genes
- TAAs, tumor-associated antigens
- TBK1, Tank-binding kinase 1
- TLRs, Toll-like receptors
- TT, tetanus toxin
- Trp2, tyrosinase related protein 2
- cellular immune response
- hTERT, human telomerase reverse transcriptase
- humoral immune response
- immune tolerance
- phTERT, optimized full-length hTERT
- tumor antigens
- vaccine delivery
Collapse
Affiliation(s)
- Benjamin Yang
- a Department of Pathology ; Johns Hopkins University ; Baltimore , MD USA
| | | | | | | | | |
Collapse
|
28
|
Frey AB. Suppression of T cell responses in the tumor microenvironment. Vaccine 2015; 33:7393-7400. [PMID: 26403368 DOI: 10.1016/j.vaccine.2015.08.096] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 07/06/2015] [Accepted: 08/05/2015] [Indexed: 12/29/2022]
Abstract
The immune system recognizes protein antigens expressed in transformed cells evidenced by accumulation of antigen-specific T cells in tumor and tumor draining lymph nodes. However, despite demonstrable immune response, cancers grow progressively suggesting that priming of antitumor immunity is insufficiently vigorous or that antitumor immunity is suppressed, or both. Compared to virus infection, antitumor T cells are low abundance that likely contributes to tumor escape and enhancement of priming is a long-sought goal of experimental vaccination therapy. Furthermore, patient treatment with antigen-specific T cells can in some cases overcome deficient priming and cause tumor regression supporting the notion that low numbers of T cells permits tumor outgrowth. However, tumor-induced suppression of antitumor immune response is now recognized as a significant factor contributing to cancer growth and reversal of the inhibitory influences within the tumor microenvironment is a major research objective. Multiple cell types and factors can inhibit T cell functions in tumors and may be grouped in two general classes: T cell intrinsic and T cell extrinsic. T cell intrinsic factors are exemplified by T cell expression of cell surface inhibitory signaling receptors that, after contact with cells expressing a cognate ligand, inactivate proximal T Cell Receptor-mediated signal transduction therein rendering T cells dysfunctional. T cell extrinsic factors are more diverse in nature and are produced by tumors and various non-tumor cells in the tumor microenvironment. These include proteins secreted by tumor or stromal cells, highly reactive soluble oxygen and nitrogen species, cytokines, chemokines, gangliosides, and toxic metabolites. These factors may restrict T cell entrance into the tumor parenchyma, cause inactivation of effector phase T cell functions, or induce T cell apoptosis ultimately causing diminished cancer elimination. Here, we review the contributions of inhibitory factors to tumor T cell dysfunction leading to tumor escape.
Collapse
Affiliation(s)
- Alan B Frey
- Department of Cell Biology, Perlmutter Cancer Center, New York University Langone School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
29
|
Galaine J, Borg C, Godet Y, Adotévi O. Interest of Tumor-Specific CD4 T Helper 1 Cells for Therapeutic Anticancer Vaccine. Vaccines (Basel) 2015; 3:490-502. [PMID: 26350591 PMCID: PMC4586463 DOI: 10.3390/vaccines3030490] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/23/2015] [Accepted: 06/24/2015] [Indexed: 12/16/2022] Open
Abstract
Nowadays, immunotherapy represents one promising approach for cancer treatment. Recently, spectacular results of cancer immunotherapy clinical trials have confirmed the crucial role of immune system in cancer regression. Therapeutic cancer vaccine represents one widely used immunotherapy strategy to stimulate tumor specific T cell responses but clinical impact remains disappointing in targeting CD8 T cells. Although CD8 T cells have been initially considered to be the main protagonists, it is now clear that CD4 T cells also play a critical role in antitumor response. In this article, we discuss the role of tumor antigen-specific CD4 T cell responses and how we can target these cells to improve cancer vaccines.
Collapse
Affiliation(s)
- Jeanne Galaine
- INSERM UMR1098, Besançon cedex F25020, France.
- Université de Franche-Comté, Besançon cedex F25020, France.
- EFS Bourgogne Franche-Comté, Besançon cedex F25020, France.
| | - Christophe Borg
- INSERM UMR1098, Besançon cedex F25020, France.
- Université de Franche-Comté, Besançon cedex F25020, France.
- EFS Bourgogne Franche-Comté, Besançon cedex F25020, France.
- Department of Medical Oncology, University Hospital of Besançon, Besançon cedex F25020, France.
| | - Yann Godet
- INSERM UMR1098, Besançon cedex F25020, France.
- Université de Franche-Comté, Besançon cedex F25020, France.
- EFS Bourgogne Franche-Comté, Besançon cedex F25020, France.
| | - Olivier Adotévi
- INSERM UMR1098, Besançon cedex F25020, France.
- Université de Franche-Comté, Besançon cedex F25020, France.
- EFS Bourgogne Franche-Comté, Besançon cedex F25020, France.
- Department of Medical Oncology, University Hospital of Besançon, Besançon cedex F25020, France.
| |
Collapse
|
30
|
Human Tumor Antigens and Cancer Immunotherapy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:948501. [PMID: 26161423 PMCID: PMC4487697 DOI: 10.1155/2015/948501] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/03/2015] [Indexed: 01/21/2023]
Abstract
With the recent developments of adoptive T cell therapies and the use of new monoclonal antibodies against the immune checkpoints, immunotherapy is at a turning point. Key players for the success of these therapies are the cytolytic T lymphocytes, which are a subset of T cells able to recognize and kill tumor cells. Here, I review the nature of the antigenic peptides recognized by these T cells and the processes involved in their presentation. I discuss the importance of understanding how each antigenic peptide is processed in the context of immunotherapy and vaccine delivery.
Collapse
|
31
|
Gruselle O, Coche T, Louahed J. Development of a Quantitative Real-Time RT-PCR Assay for the Detection of MAGE-A3-Positive Tumors. J Mol Diagn 2015; 17:382-91. [PMID: 25986000 DOI: 10.1016/j.jmoldx.2015.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 02/26/2015] [Accepted: 03/06/2015] [Indexed: 11/26/2022] Open
Abstract
Melanoma antigen A3 (MAGE-A3) is a member of the MAGE family of tumor antigens and a relevant candidate for use in cancer immunotherapy. However, not all tumors express MAGE-A3, and closely related members of the MAGE family can be co-expressed with MAGE-A3 in the same tumor. Therefore, in the frame of MAGE-A3 clinical trials, it appeared necessary to evaluate tumors for MAGE-A3 expression with a highly specific quantitative assay to select patients who are eligible for anti-MAGE-A3 immunotherapy treatment. Herein, we describe the development and validation of a quantitative real-time RT-PCR (RT-qPCR) assay for the determination of MAGEA3 gene expression in tumor tissues. In the early phases of development, the designed primers and probe were not able to distinguish between MAGE-A3 and MAGE-A6. To ensure the specificity for MAGE-A3 over MAGE-A6, our strategy was to use a 5'-nuclease probe (or hydrolysis probe). The final assay was shown to be specific and linear within the analytical range, with an acceptable CV for repeatability and intermediate precision. When compared with a reference semiquantitative RT-PCR assay, the two methods were in good agreement, with only 4.23% of the samples giving discordant results. In conclusion, we have developed a MAGE-A3-specific RT-qPCR assay, compatible with a high-throughput setting for the estimation of MAGEA3 gene expression in present and future clinical trials.
Collapse
Affiliation(s)
| | - Thierry Coche
- Immunotherapeutics, GSK Vaccines, Rixensart, Belgium.
| | | |
Collapse
|
32
|
Apcher S, Daskalogianni C, Fåhraeus R. Pioneer translation products as an alternative source for MHC-I antigenic peptides. Mol Immunol 2015; 68:68-71. [PMID: 25979818 DOI: 10.1016/j.molimm.2015.04.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/22/2015] [Indexed: 12/12/2022]
Abstract
The notion that alternative peptide substrates can be processed and presented to the MHC class I pathway has opened for new aspects on how the immune system detects infected or damaged cells. Recent works show that antigenic peptides are derived from intron sequences in pre-mRNAs target for the nonsense-mediated degradation pathway. Introns are spliced out co-transcriptionally suggesting that such pioneer translation products (PTPs) are synthesized on the nascent RNAs in the nuclear compartment to ensure that the first peptides to emerge from an mRNA are destined for the class I pathway. This illustrates an independent translation event during mRNA maturation that give rise to specific peptide products with a specific function in the immune system. The characterization of the translation apparatus responsible for PTP synthesis will pave the way for understanding how PTP production is regulated in different tissues under different conditions and will help designing new vaccine strategies.
Collapse
Affiliation(s)
- Sebastien Apcher
- Institut Gustave Roussy, Université Paris Sud, Unité 1015 département d'immunologie, 114, rue Edouard Vaillant, 94805 Villejuif, France.
| | - Chrysoula Daskalogianni
- Equipe Labellisée la Ligue Contre le Cancer, Inserm UMR1162, Université Paris 7, Institut de Génétique Moléculaire, 27 rue Juliette Dodu, 75010 Paris, France
| | - Robin Fåhraeus
- Equipe Labellisée la Ligue Contre le Cancer, Inserm UMR1162, Université Paris 7, Institut de Génétique Moléculaire, 27 rue Juliette Dodu, 75010 Paris, France; RECAMO, Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53 Brno, Czech Republic.
| |
Collapse
|
33
|
Parmiani G, Pilla L, Corti A, Doglioni C, Cimminiello C, Bellone M, Parolini D, Russo V, Capocefalo F, Maccalli C. A pilot Phase I study combining peptide-based vaccination and NGR-hTNF vessel targeting therapy in metastatic melanoma. Oncoimmunology 2014; 3:e963406. [PMID: 25941591 DOI: 10.4161/21624011.2014.963406] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/04/2014] [Indexed: 11/19/2022] Open
Abstract
Administration of NGR-TNF, a tumor vessel-targeting and tumor necrosis factor α TNFα) peptide conjugate, with immunotherapy has been shown to inhibit tumor growth in mice. Thus, we planned a Phase I pilot clinical trial to assess safety, immune and clinical response of this combination treatment for advanced melanoma. NA17.A2 and MAGE-3.A1 peptides were used as vaccine. HLA-A*0201 or HLA-A*01 metastatic melanoma patients received human NGR-hTNF i.v. alternating with s.c. weekly injections of either of the peptides emulsified in Montanide. The T-cell response was assessed ex-vivo using peripheral blood mononuclear cells (PBMCs) before, during and after therapy. The serum level of chromogranin A (CgA), soluble TNF receptors (sTNFR1/2), vascular endothelial growth factor (VEGF), and MIP-1β and MCP-1 chemokines, was determined. In 3 subjects, pre- and post-treatment tumor lesions were examined by immunohistochemistry. Clinically, chills were observed in 4 patients during NGR-hTNF infusion and erythema at vaccination site was seen in 7 patients. T-cell response against the vaccine or against other melanoma-associated antigens was detectable after treatment in 6 out of 7 tested patients. Low level or reduction of CgA and sTNFR and increase of MIP-1β and MCP-1 were found in patients sera. In the lesions examined the immune infiltrate was scanty but macrophage number increased in post-therapy lesions. From a clinical standpoint, a long term survival (>4 months) was found in 6 out of 8 evaluable patients (4, 4, 7, 11, 23+, 25+, 25+, 29+ months). The combination of NGR-hTNF and vaccine in metastatic melanoma patients was well tolerated, often associated with an ex-vivo T cell response and long-term overall survival. These findings warrant confirmation in a larger group of patients.
Collapse
Key Words
- APC, antigen presenting cell
- CT, cancer/testis
- CgA, chromogranin A
- DFS, disease-free survival
- MAA, melanoma-associated antigens
- MCP-1, macrophage chemoattractant protein 1
- MIP-1β, macrophage inflammatory protein 1β; OS, overall survival
- PBMC, peripheral blood mononuclear cell
- PD, progression of disease
- PFS, progression-free survival
- RR, response rate
- T cells
- TNFα, tumor necrosis factor α
- anti-vascular target therapy
- combination therapy
- inflammatory cytokines
- melanoma
- peptide-based vaccines
- sTNFR, soluble tumor necrosis factor receptor
Collapse
Affiliation(s)
- Giorgio Parmiani
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Lorenzo Pilla
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Angelo Corti
- Unit of Tumor Biology and Vascular Targeting; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Claudio Doglioni
- Unit of Pathology; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Carolina Cimminiello
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Matteo Bellone
- Unit of Cellular Immunology; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Danilo Parolini
- Unit of Gastrointestinal Surgery; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Vincenzo Russo
- Unit of Cancer Gene Therapy; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Filippo Capocefalo
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| | - Cristina Maccalli
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors; San Raffaele Foundation Research Institute ; Via Olgettina , Milan
| |
Collapse
|
34
|
Vauchy C, Gamonet C, Ferrand C, Daguindau E, Galaine J, Beziaud L, Chauchet A, Henry Dunand CJ, Deschamps M, Rohrlich PS, Borg C, Adotevi O, Godet Y. CD20 alternative splicing isoform generates immunogenic CD4 helper T epitopes. Int J Cancer 2014; 137:116-26. [DOI: 10.1002/ijc.29366] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/12/2014] [Accepted: 11/18/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Charline Vauchy
- INSERM UMR1098, F25020 Besançon cedex; France
- Université de Franche-Comté, F25020 Besançon cedex; France
- EFS Bourgogne Franche-Comté, F25020 Besançon cedex; France
| | - Clementine Gamonet
- INSERM UMR1098, F25020 Besançon cedex; France
- Université de Franche-Comté, F25020 Besançon cedex; France
- EFS Bourgogne Franche-Comté, F25020 Besançon cedex; France
| | - Christophe Ferrand
- INSERM UMR1098, F25020 Besançon cedex; France
- Université de Franche-Comté, F25020 Besançon cedex; France
- EFS Bourgogne Franche-Comté, F25020 Besançon cedex; France
| | - Etienne Daguindau
- Department of Hematology; University Hospital of Besançon, F25020 Besançon cedex; France
| | - Jeanne Galaine
- INSERM UMR1098, F25020 Besançon cedex; France
- Université de Franche-Comté, F25020 Besançon cedex; France
- EFS Bourgogne Franche-Comté, F25020 Besançon cedex; France
| | - Laurent Beziaud
- INSERM UMR1098, F25020 Besançon cedex; France
- Université de Franche-Comté, F25020 Besançon cedex; France
- EFS Bourgogne Franche-Comté, F25020 Besançon cedex; France
| | - Adrien Chauchet
- Department of Hematology; University Hospital of Besançon, F25020 Besançon cedex; France
| | - Carole J. Henry Dunand
- The Department of Medicine; Section of Rheumatology, The Knapp Center for Lupus and Immunology Research, The University of Chicago; Chicago IL
| | - Marina Deschamps
- INSERM UMR1098, F25020 Besançon cedex; France
- Université de Franche-Comté, F25020 Besançon cedex; France
- EFS Bourgogne Franche-Comté, F25020 Besançon cedex; France
| | - Pierre Simon Rohrlich
- INSERM UMR1098, F25020 Besançon cedex; France
- Université de Franche-Comté, F25020 Besançon cedex; France
- Department of Pediatrics; University Hospital of Besançon, F25020 Besançon cedex; France
| | - Christophe Borg
- INSERM UMR1098, F25020 Besançon cedex; France
- Université de Franche-Comté, F25020 Besançon cedex; France
- EFS Bourgogne Franche-Comté, F25020 Besançon cedex; France
- Department of Medical Oncology; University Hospital of Besançon, F25020 Besançon cedex; France
| | - Olivier Adotevi
- INSERM UMR1098, F25020 Besançon cedex; France
- Université de Franche-Comté, F25020 Besançon cedex; France
- EFS Bourgogne Franche-Comté, F25020 Besançon cedex; France
- Department of Medical Oncology; University Hospital of Besançon, F25020 Besançon cedex; France
| | - Yann Godet
- INSERM UMR1098, F25020 Besançon cedex; France
- Université de Franche-Comté, F25020 Besançon cedex; France
- EFS Bourgogne Franche-Comté, F25020 Besançon cedex; France
| |
Collapse
|
35
|
Andersen RS, Andersen SR, Hjortsø MD, Lyngaa R, Idorn M, Køllgård TM, Met O, Thor Straten P, Hadrup SR. High frequency of T cells specific for cryptic epitopes in melanoma patients. Oncoimmunology 2014; 2:e25374. [PMID: 24073381 PMCID: PMC3782131 DOI: 10.4161/onci.25374] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 06/11/2013] [Accepted: 06/12/2013] [Indexed: 11/24/2022] Open
Abstract
A number of cytotoxic T-cell epitopes are cryptic epitopes generated from non-conventional sources. These include epitopes that are encoded by alternative open reading frames or in generally non-coding genomic regions, such as introns. We have previously observed a frequent recognition of cryptic epitopes by tumor infiltrating lymphocytes isolated from melanoma patients. Here, we show that such cryptic epitopes are more frequently recognized than antigens of the same class encoded by canonical reading frames. Furthermore, we report the presence of T cells specific for three cryptic epitopes encoded in intronic sequences, as a result of incomplete splicing, in the circulation of melanoma patients. One of these epitopes derives from antigen isolated from immunoselected melanoma 2 (AIM2), while the two others are encoded in an alternative open reading frame of an incompletely spliced form of N-acetylglucosaminyl-transferase V (GNT-V) known as NA17-A. We have detected frequent T-cell responses against AIM2 and NA17-A epitopes in the blood of melanoma patients, both prior and after one round of in vitro peptide stimulation, but not in the circulation of healthy individuals and patients with breast or renal carcinoma. In summary, our findings indicate that the T-cell reactivity against AIM2 and NA17-A in the blood of melanoma patients is extensive, suggesting that—similar to melan A (also known as MART1)—these antigens might be used for immunomonitoring or as model antigens in several clinical and preclinical settings.
Collapse
Affiliation(s)
- Rikke Sick Andersen
- Center for Cancer Immune Therapy; University Hospital Herlev; Herlev, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Linnemann C, Mezzadra R, Schumacher TNM. TCR repertoires of intratumoral T-cell subsets. Immunol Rev 2014; 257:72-82. [PMID: 24329790 DOI: 10.1111/imr.12140] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The infiltration of human tumors by T cells is a common phenomenon, and over the past decades, it has become increasingly clear that the nature of such intratumoral T-cell populations can predict disease course. Furthermore, intratumoral T cells have been utilized therapeutically in clinical studies of adoptive T-cell therapy. In this review, we describe how novel methods that are either based on T-cell receptor (TCR) sequencing or on cancer exome analysis allow the analysis of the tumor reactivity and antigen-specificity of the intratumoral TCR repertoire with unprecedented detail. Furthermore, we discuss studies that have started to utilize these techniques to probe the link between cancer exomes and the intratumoral TCR pool. Based on the observation that both the cancer epitope repertoire and intratumoral TCR repertoire appear highly individual, we outline strategies, such as 'autologous TCR gene therapy', that exploit the tumor-resident TCR repertoire for the development of personalized immunotherapy.
Collapse
Affiliation(s)
- Carsten Linnemann
- Division of Immunology, The Netherlands Cancer Institute (NKI-AVL), Amsterdam, the Netherlands
| | | | | |
Collapse
|
37
|
Allard M, Oger R, Benlalam H, Florenceau L, Echasserieau K, Bernardeau K, Labarrière N, Lang F, Gervois N. Soluble HLA-I/peptide monomers mediate antigen-specific CD8 T cell activation through passive peptide exchange with cell-bound HLA-I molecules. THE JOURNAL OF IMMUNOLOGY 2014; 192:5090-7. [PMID: 24752447 DOI: 10.4049/jimmunol.1303226] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Accumulating evidence that serum levels of soluble class I HLA molecules (sHLA-I) can, under various pathological conditions, correlate with disease stage and/or patient survival, has stimulated interest in defining whether sHLA-I can exert immunological functions. However, despite a mounting number of publications suggesting the ability of sHLA-I to affect immune effectors in vitro, the precise underlying mechanism still remains controversial. In this article, we address potential functions of both classical and nonclassical sHLA-I, using soluble recombinant HLA-I/peptide monomers, and clearly demonstrate their ability to trigger Ag-specific activation of CD8 T cells in vitro. Furthermore, we provide strong evidence that this behavior results from the passive transfer of peptides from monomers to T cell-bound HLA-I molecules, allowing for fratricide representation and activation. Hence, we proposed a unifying model of T cell activation by HLA-I/peptide monomers, reappraising the potential involvement of sHLA-I molecules in the immune response.
Collapse
Affiliation(s)
- Mathilde Allard
- INSERM, U892, Nantes F-44007, France; Centre National de la Recherche Scientifique, U6299, Nantes F-44007, France; Université de Nantes, Nantes F-44007 France; and
| | - Romain Oger
- INSERM, U892, Nantes F-44007, France; Centre National de la Recherche Scientifique, U6299, Nantes F-44007, France; Université de Nantes, Nantes F-44007 France; and
| | - Houssem Benlalam
- INSERM, U892, Nantes F-44007, France; Centre National de la Recherche Scientifique, U6299, Nantes F-44007, France; Université de Nantes, Nantes F-44007 France; and
| | - Laetitia Florenceau
- INSERM, U892, Nantes F-44007, France; Centre National de la Recherche Scientifique, U6299, Nantes F-44007, France; Université de Nantes, Nantes F-44007 France; and
| | - Klara Echasserieau
- INSERM, U892, Nantes F-44007, France; Centre National de la Recherche Scientifique, U6299, Nantes F-44007, France; Université de Nantes, Nantes F-44007 France; and Recombinant Protein Production Facility, Federative Research Structure François Bonamy, Nantes, F-44007, France
| | - Karine Bernardeau
- INSERM, U892, Nantes F-44007, France; Recombinant Protein Production Facility, Federative Research Structure François Bonamy, Nantes, F-44007, France
| | - Nathalie Labarrière
- INSERM, U892, Nantes F-44007, France; Centre National de la Recherche Scientifique, U6299, Nantes F-44007, France; Université de Nantes, Nantes F-44007 France; and
| | - François Lang
- INSERM, U892, Nantes F-44007, France; Centre National de la Recherche Scientifique, U6299, Nantes F-44007, France; Université de Nantes, Nantes F-44007 France; and Recombinant Protein Production Facility, Federative Research Structure François Bonamy, Nantes, F-44007, France
| | - Nadine Gervois
- INSERM, U892, Nantes F-44007, France; Centre National de la Recherche Scientifique, U6299, Nantes F-44007, France; Université de Nantes, Nantes F-44007 France; and
| |
Collapse
|
38
|
Duvallet E, Boulpicante M, Apcher S. [Synthesis of MHC class I antigenic peptides in the nucleus: a role for the nuclear translation at last?]. Med Sci (Paris) 2014; 30:229-31. [PMID: 24685205 DOI: 10.1051/medsci/20143003002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Emilie Duvallet
- Institut Gustave Roussy, Université Paris Sud, département d'immunologie, Unité 1015, 114, rue Édouard Vaillant, 94805 Villejuif, France
| | - Mathilde Boulpicante
- Institut Gustave Roussy, Université Paris Sud, département d'immunologie, Unité 1015, 114, rue Édouard Vaillant, 94805 Villejuif, France
| | - Sébastien Apcher
- Institut Gustave Roussy, Université Paris Sud, département d'immunologie, Unité 1015, 114, rue Édouard Vaillant, 94805 Villejuif, France
| |
Collapse
|
39
|
Local immunostimulation leading to rejection of accepted male skin grafts by female mice as a model for cancer immunotherapy. Proc Natl Acad Sci U S A 2014; 111:3502-7. [PMID: 24550491 DOI: 10.1073/pnas.1401115111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Female mice of inbred strain CBA do not reject syngeneic male skin grafts even though they mount a T-cell response against the male-specific HY antigen. We show that local immunostimulation performed by injecting cytokines and Toll-like receptor ligands in close vicinity to the graft causes rejection. We feel that this approach should be tested in tumor-bearing human patients in combination with antitumor vaccination. Relief of intratumor immunosuppression may increase considerably the fraction of patients who respond to vaccination directed against tumor antigens recognized by T cells.
Collapse
|
40
|
Coulie PG, Van den Eynde BJ, van der Bruggen P, Boon T. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer 2014; 14:135-46. [PMID: 24457417 DOI: 10.1038/nrc3670] [Citation(s) in RCA: 839] [Impact Index Per Article: 76.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In this Timeline, we describe the characteristics of tumour antigens that are recognized by spontaneous T cell responses in cancer patients and the paths that led to their identification. We explain on what genetic basis most, but not all, of these antigens are tumour specific: that is, present on tumour cells but not on normal cells. We also discuss how strategies that target these tumour-specific antigens can lead either to tumour-specific or to crossreactive T cell responses, which is an issue that has important safety implications in immunotherapy. These safety issues are even more of a concern for strategies targeting antigens that are not known to induce spontaneous T cell responses in patients.
Collapse
Affiliation(s)
- Pierre G Coulie
- 1] de Duve Institute and the Université catholique de Louvain, B-1200 Brussels, Belgium. [2] WELBIO (Walloon Excellence in Lifesciences and Biotechnology), B-1200 Brussels, Belgium
| | - Benoît J Van den Eynde
- 1] de Duve Institute and the Université catholique de Louvain, B-1200 Brussels, Belgium. [2] Ludwig Institute for Cancer Research, B-1200 Brussels, Belgium. [3] WELBIO (Walloon Excellence in Lifesciences and Biotechnology), B-1200 Brussels, Belgium
| | - Pierre van der Bruggen
- 1] de Duve Institute and the Université catholique de Louvain, B-1200 Brussels, Belgium. [2] Ludwig Institute for Cancer Research, B-1200 Brussels, Belgium. [3] WELBIO (Walloon Excellence in Lifesciences and Biotechnology), B-1200 Brussels, Belgium
| | - Thierry Boon
- 1] de Duve Institute and the Université catholique de Louvain, B-1200 Brussels, Belgium. [2] Ludwig Institute for Cancer Research, B-1200 Brussels, Belgium
| |
Collapse
|
41
|
Michaux A, Larrieu P, Stroobant V, Fonteneau JF, Jotereau F, Van den Eynde BJ, Moreau-Aubry A, Vigneron N. A Spliced Antigenic Peptide Comprising a Single Spliced Amino Acid Is Produced in the Proteasome by Reverse Splicing of a Longer Peptide Fragment followed by Trimming. THE JOURNAL OF IMMUNOLOGY 2014; 192:1962-71. [DOI: 10.4049/jimmunol.1302032] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
42
|
van Buuren MM, Dijkgraaf FE, Linnemann C, Toebes M, Chang CXL, Mok JY, Nguyen M, van Esch WJE, Kvistborg P, Grotenbreg GM, Schumacher TNM. HLA micropolymorphisms strongly affect peptide-MHC multimer-based monitoring of antigen-specific CD8+ T cell responses. THE JOURNAL OF IMMUNOLOGY 2013; 192:641-8. [PMID: 24342804 DOI: 10.4049/jimmunol.1301770] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Peptide-MHC (pMHC) multimers have become one of the most widely used tools to measure Ag-specific T cell responses in humans. With the aim of understanding the requirements for pMHC-based personalized immunomonitoring, in which individuals expressing subtypes of the commonly studied HLA alleles are encountered, we assessed how the ability to detect Ag-specific T cells for a given peptide is affected by micropolymorphic differences between HLA subtypes. First, analysis of a set of 10 HLA-A*02:01-restricted T cell clones demonstrated that staining with pMHC multimers of seven distinct subtypes of the HLA-A*02 allele group was highly variable and not predicted by sequence homology. Second, to analyze the effect of minor sequence variation in a clinical setting, we screened tumor-infiltrating lymphocytes of an HLA-A*02:06 melanoma patient with either subtype-matched or HLA-A*02:01 multimers loaded with 145 different melanoma-associated Ags. This revealed that of the four HLA-A*02:06-restricted melanoma-associated T cell responses observed in this patient, two responses were underestimated and one was overlooked when using subtype-mismatched pMHC multimer collections. To our knowledge, these data provide the first demonstration of the strong effect of minor sequence variation on pMHC-based personalized immunomonitoring, and they provide tools to prevent this issue for common variants within the HLA-A*02 allele group.
Collapse
Affiliation(s)
- Marit M van Buuren
- Division of Immunology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Translation of pre-spliced RNAs in the nuclear compartment generates peptides for the MHC class I pathway. Proc Natl Acad Sci U S A 2013; 110:17951-6. [PMID: 24082107 DOI: 10.1073/pnas.1309956110] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The scanning of maturing mRNAs by ribosomes plays a key role in the mRNA quality control process. When ribosomes first engage with the newly synthesized mRNA, and if peptides are produced, is unclear, however. Here we show that ribosomal scanning of prespliced mRNAs occurs in the nuclear compartment, and that this event produces peptide substrates for the MHC class I pathway. Inserting antigenic peptide sequences in introns that are spliced out before the mRNAs exit the nuclear compartment results in an equal amount of antigenic peptide products as when the peptides are encoded from the main open reading frame (ORF). Taken together with the detection of intron-encoded nascent peptides and RPS6/RPL7-carrying complexes in the perinucleolar compartment, these results show that peptides are produced by a translation event occurring before mRNA splicing. This suggests that ribosomes occupy and scan mRNAs early in the mRNA maturation process, and suggests a physiological role for nuclear mRNA translation, and also helps explain how the immune system tolerates peptides derived from tissue-specific mRNA splice variants.
Collapse
|
44
|
Vigneron N, Stroobant V, Van den Eynde BJ, van der Bruggen P. Database of T cell-defined human tumor antigens: the 2013 update. CANCER IMMUNITY 2013; 13:15. [PMID: 23882160 PMCID: PMC3718731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The plethora of tumor antigens that have been--and are still being--defined required systematization to provide a comprehensive overview of those tumor antigens that are the most relevant targets for cancer immunotherapy approaches. Here, we provide a new update of a peptide database resource that we initiated many years ago. This database compiles all human antigenic peptides described in the literature that fulfill a set of strict criteria needed to ascertain their actual "tumor antigen" nature, as we aim at guiding scientists and clinicians searching for appropriate cancer vaccine candidates (www.cancerimmunity.org/peptide). In this review, we revisit those criteria in light of recent findings related to antigen processing. We also introduce the 29 new tumor antigens that were selected for this 2013 update. Two of the new peptides show unusual features, which will be briefly discussed. The database now comprises a total of 403 tumor antigenic peptides.
Collapse
Affiliation(s)
- Nathalie Vigneron
- Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium
- WELBIO and de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Vincent Stroobant
- Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium
- WELBIO and de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Benoît J. Van den Eynde
- Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium
- WELBIO and de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Pierre van der Bruggen
- Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium
- WELBIO and de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
45
|
Schweighoffer T. Molecular cancer vaccines: Tumor therapy using antigen-specific immunizations. Pathol Oncol Res 2012; 3:164-76. [PMID: 18470726 DOI: 10.1007/bf02899917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/1997] [Accepted: 08/24/1997] [Indexed: 10/21/2022]
Abstract
Vaccination against tumors promises selective destruction of malignant cells by the host's immune system. Molecular cancer vaccines rely on recently identified tumor antigens as immunogens. Tumor antigens can be applied in many forms, as genes in recombinant vectors, as proteins or peptides representing T cell epitopes.Analysis of various aspects indicates some advantage for peptide-based vaccines over the other modalities. Further refinements and extensively monitored clinical trials are necessary to advance molecular cancer vaccines from concepts into powerful therapy.
Collapse
Affiliation(s)
- T Schweighoffer
- Department Cell Biology, Boehringer Ingelheim Research and Development, Dr. Boehringer-Gasse 5, A-l 120, Wien, Austria,
| |
Collapse
|
46
|
An antigenic peptide produced by reverse splicing and double asparagine deamidation. Proc Natl Acad Sci U S A 2011; 108:E323-31. [PMID: 21670269 DOI: 10.1073/pnas.1101892108] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A variety of unconventional translational and posttranslational mechanisms contribute to the production of antigenic peptides, thereby increasing the diversity of the peptide repertoire presented by MHC class I molecules. Here, we describe a class I-restricted peptide that combines several posttranslational modifications. It is derived from tyrosinase and recognized by tumor-infiltrating lymphocytes isolated from a melanoma patient. This unusual antigenic peptide is made of two noncontiguous tyrosinase fragments that are spliced together in the reverse order. In addition, it contains two aspartate residues that replace the asparagines encoded in the tyrosinase sequence. We confirmed that this peptide is naturally presented at the surface of melanoma cells, and we showed that its processing sequentially requires translation of tyrosinase into the endoplasmic reticulum and its retrotranslocation into the cytosol, where deglycosylation of the two asparagines by peptide-N-glycanase turns them into aspartates by deamidation. This process is followed by cleavage and splicing of the appropriate fragments by the standard proteasome and additional transport of the resulting peptide into the endoplasmic reticulum through the transporter associated with antigen processing (TAP).
Collapse
|
47
|
Vigneron N, Van den Eynde BJ. Insights into the processing of MHC class I ligands gained from the study of human tumor epitopes. Cell Mol Life Sci 2011; 68:1503-20. [PMID: 21387143 PMCID: PMC11114561 DOI: 10.1007/s00018-011-0658-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 02/17/2011] [Accepted: 02/18/2011] [Indexed: 12/29/2022]
Abstract
The molecular definition of tumor antigens recognized by cytolytic T lymphocytes (CTL) started in the late 1980s, at a time when the MHC class I antigen processing field was in its infancy. Born together, these two fields of science evolved together and provided each other with critical insights. Over the years, stimulated by the potential interest of tumor antigens for cancer immunotherapy, scientists have identified and characterized numerous antigens recognized by CTL on human tumors. These studies have provided a wealth of information relevant to the mode of production of antigenic peptides presented by MHC class I molecules. A number of tumor antigenic peptides were found to result from unusual mechanisms occurring at the level of transcription, translation or processing. Although many of these mechanisms occur in the cell at very low level, they are relevant to the immune system as they determine the killing of tumor cells by CTL, which are sensitive to low levels of peptide/MHC complexes. Moreover, these unusual mechanisms were found to occur not only in tumor cells but also in normal cells. Thereby, the study of tumor antigens has illuminated many aspects of MHC class I processing. We review here those insights into the MHC I antigen processing pathway that result from the characterization of human tumor antigens recognized by CTL.
Collapse
Affiliation(s)
- Nathalie Vigneron
- Ludwig Institute for Cancer Research, Brussels Branch and de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 74, UCL 7459, 1200 Brussels, Belgium
| | - Benoît J. Van den Eynde
- Ludwig Institute for Cancer Research, Brussels Branch and de Duve Institute, Université Catholique de Louvain, Avenue Hippocrate 74, UCL 7459, 1200 Brussels, Belgium
| |
Collapse
|
48
|
Starck SR, Shastri N. Non-conventional sources of peptides presented by MHC class I. Cell Mol Life Sci 2011; 68:1471-9. [PMID: 21390547 PMCID: PMC3071930 DOI: 10.1007/s00018-011-0655-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 02/17/2011] [Accepted: 02/18/2011] [Indexed: 12/14/2022]
Abstract
Effectiveness of immune surveillance of intracellular viruses and bacteria depends upon a functioning antigen presentation pathway that allows infected cells to reveal the presence of an intracellular pathogen. The antigen presentation pathway uses virtually all endogenous polypeptides as a source to produce antigenic peptides that are eventually chaperoned to the cell surface by MHC class I molecules. Intriguingly, MHC I molecules present peptides encoded not only in the primary open reading frames but also those encoded in alternate reading frames. Here, we review recent studies on the generation of cryptic pMHC I. We focus on the immunological significance of cryptic pMHC I, and the novel translational mechanisms that allow production of these antigenic peptides from unconventional sources.
Collapse
Affiliation(s)
- Shelley R. Starck
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, LSA 421, Berkeley, CA 94720-3200 USA
| | - Nilabh Shastri
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, LSA 421, Berkeley, CA 94720-3200 USA
| |
Collapse
|
49
|
Cuffel C, Rivals JP, Zaugg Y, Salvi S, Seelentag W, Speiser DE, Liénard D, Monnier P, Romero P, Bron L, Rimoldi D. Pattern and clinical significance of cancer-testis gene expression in head and neck squamous cell carcinoma. Int J Cancer 2010; 128:2625-34. [DOI: 10.1002/ijc.25607] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 06/22/2010] [Indexed: 12/17/2022]
|
50
|
Renaud V, Godefroy E, Larrieu P, Fleury F, Jotereau F, Guilloux Y. Folding of matrix metalloproteinase-2 prevents endogenous generation of MHC class-I restricted epitope. PLoS One 2010; 5:e11894. [PMID: 20689590 PMCID: PMC2912773 DOI: 10.1371/journal.pone.0011894] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 07/05/2010] [Indexed: 11/18/2022] Open
Abstract
Background We previously demonstrated that the matrix metalloproteinase-2 (MMP-2) contained an antigenic peptide recognized by a CD8 T cell clone in the HLA-A*0201 context. The presentation of this peptide on class I molecules by human melanoma cells required a cross-presentation mechanism. Surprisingly, the classical endogenous processing pathway did not process this MMP-2 epitope. Methodology/Principal Findings By PCR directed mutagenesis we showed that disruption of a single disulfide bond induced MMP-2 epitope presentation. By Pulse-Chase experiment, we demonstrated that disulfide bonds stabilized MMP-2 and impeded its degradation. Finally, using drugs, we documented that mutated MMP-2 epitope presentation used the proteasome and retrotranslocation complex. Conclusions/Significance These data appear crucial to us since they established the existence of a new inhibitory mechanism for the generation of a T cell epitope. In spite of MMP-2 classified as a self-antigen, the fact that cross-presentation is the only way to present this MMP-2 epitope underlines the importance to target this type of antigen in immunotherapy protocols.
Collapse
Affiliation(s)
- Virginie Renaud
- Institut National de la Santé et de la Recherche Médicale, UMR 892, Nantes, France
| | | | | | | | | | | |
Collapse
|