1
|
Arshad S, Cameron B, Joglekar AV. Immunopeptidomics for autoimmunity: unlocking the chamber of immune secrets. NPJ Syst Biol Appl 2025; 11:10. [PMID: 39833247 PMCID: PMC11747513 DOI: 10.1038/s41540-024-00482-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
T cells mediate pathogenesis of several autoimmune disorders by recognizing self-epitopes presented on Major Histocompatibility Complex (MHC) or Human Leukocyte Antigen (HLA) complex. The majority of autoantigens presented to T cells in various autoimmune disorders are not known, which has impeded autoantigen identification. Recent advances in immunopeptidomics have started to unravel the repertoire of antigenic epitopes presented on MHC. In several autoimmune diseases, immunopeptidomics has led to the identification of novel autoantigens and has enhanced our understanding of the mechanisms behind autoimmunity. Especially, immunopeptidomics has provided key evidence to explain the genetic risk posed by HLA alleles. In this review, we shed light on how immunopeptidomics can be leveraged to discover potential autoantigens. We highlight the application of immunopeptidomics in Type 1 Diabetes (T1D), Systemic Lupus Erythematosus (SLE), and Rheumatoid Arthritis (RA). Finally, we highlight the practical considerations of implementing immunopeptidomics successfully and the technical challenges that need to be addressed. Overall, this review will provide an important context for using immunopeptidomics for understanding autoimmunity.
Collapse
Affiliation(s)
- Sanya Arshad
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Benjamin Cameron
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alok V Joglekar
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Mishto M, Takala I, Bonfanti P, Liepe J. Proteasome isoforms in human thymi and mouse models. Immunol Lett 2024; 269:106899. [PMID: 39019403 DOI: 10.1016/j.imlet.2024.106899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/02/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024]
Abstract
The thymus is the organ where functional and self-tolerant T cells are selected through processes of positive and negative selection before migrating to the periphery. The antigenic peptides presented on MHC class I molecules of thymic epithelial cells (TECs) in the cortex and medulla of the thymus are key players in these processes. It has been theorized that these cells express different proteasome isoforms, which generate MHC class I immunopeptidomes with features that differentiate cortex and medulla, and hence positive and negative CD8+ T cell selection. This theory is largely based on mouse models and does not consider the large variety of noncanonical antigenic peptides that could be produced by proteasomes and presented on MHC class I molecules. Here, we review the multi-omics, biochemical and cellular studies carried out on mouse models and human thymi to investigate their content of proteasome isoforms, briefly summarize the implication that noncanonical antigenic peptide presentation in the thymus could have on CD8+ T cell repertoire and put these aspects in the larger framework of anatomical and immunological differences between these two species.
Collapse
Affiliation(s)
- Michele Mishto
- Molecular Immunology laboratory, the Francis Crick Institute, NW1 1AT London, United Kingdom; Centre for Inflammation Biology and Cancer Immunology & Peter Gorer Department of Immunobiology, King's College London, SE1 1UL London, United Kingdom.
| | - Iina Takala
- Research group of Quantitative System Biology, Max-Planck-Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Paola Bonfanti
- Epithelial Stem Cell Biology & Regenerative Medicine laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Institute of Immunity & Transplantation, Division of Infection & Immunity, UCL, Pears Building, London NW3 2PP, United Kingdom
| | - Juliane Liepe
- Research group of Quantitative System Biology, Max-Planck-Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| |
Collapse
|
3
|
Gul A, Pewe LL, Willems P, Mayer R, Thery F, Asselman C, Aernout I, Verbeke R, Eggermont D, Van Moortel L, Upton E, Zhang Y, Boucher K, Miret-Casals L, Demol H, De Smedt SC, Lentacker I, Radoshevich L, Harty JT, Impens F. Immunopeptidomics Mapping of Listeria monocytogenes T Cell Epitopes in Mice. Mol Cell Proteomics 2024; 23:100829. [PMID: 39147027 PMCID: PMC11414675 DOI: 10.1016/j.mcpro.2024.100829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/21/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024] Open
Abstract
Listeria monocytogenes is a foodborne intracellular bacterial model pathogen. Protective immunity against Listeria depends on an effective CD8+ T cell response, but very few T cell epitopes are known in mice as a common animal infection model for listeriosis. To identify epitopes, we screened for Listeria immunopeptides presented in the spleen of infected mice by mass spectrometry-based immunopeptidomics. We mapped more than 6000 mouse self-peptides presented on MHC class I molecules, including 12 high confident Listeria peptides from 12 different bacterial proteins. Bacterial immunopeptides with confirmed fragmentation spectra were further tested for their potential to activate CD8+ T cells, revealing VTYNYINI from the putative cell wall surface anchor family protein LMON_0576 as a novel bona fide peptide epitope. The epitope showed high biological potency in a prime boost model and can be used as a research tool to probe CD8+ T cell responses in the mouse models of Listeria infection. Together, our results demonstrate the power of immunopeptidomics for bacterial antigen identification.
Collapse
Affiliation(s)
- Adillah Gul
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Lecia L Pewe
- Department of Pathology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA
| | - Patrick Willems
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB-UGent Center for Plant Systems Biology, VIB, Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Rupert Mayer
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Fabien Thery
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Caroline Asselman
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Ilke Aernout
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Rein Verbeke
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Denzel Eggermont
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Laura Van Moortel
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Ellen Upton
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Yifeng Zhang
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA
| | - Katie Boucher
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Laia Miret-Casals
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Hans Demol
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lilliana Radoshevich
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA; Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA.
| | - John T Harty
- Department of Pathology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium.
| |
Collapse
|
4
|
de Greef PC, Njeru SN, Benz C, Fillatreau S, Malissen B, Agenès F, de Boer RJ, Kirberg J. The TCR assigns naive T cells to a preferred lymph node. SCIENCE ADVANCES 2024; 10:eadl0796. [PMID: 39047099 PMCID: PMC11268406 DOI: 10.1126/sciadv.adl0796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 06/21/2024] [Indexed: 07/27/2024]
Abstract
Naive T cells recirculate between the spleen and lymph nodes where they mount immune responses when meeting dendritic cells presenting foreign antigen. As this may happen anywhere, naive T cells ought to visit all lymph nodes. Here, deep sequencing almost-complete TCR repertoires led to a comparison of different lymph nodes within and between individual mice. We find strong evidence for a deterministic CD4/CD8 lineage choice and a consistent spatial structure. Specifically, some T cells show a preference for one or multiple lymph nodes, suggesting that their TCR interacts with locally presented (self-)peptides. These findings are mirrored in TCR-transgenic mice showing localized CD69 expression, retention, and cell division. Thus, naive T cells intermittently sense antigenically dissimilar niches, which is expected to affect their homeostatic competition.
Collapse
MESH Headings
- Animals
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Mice
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Mice, Transgenic
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Lectins, C-Type/metabolism
- Lectins, C-Type/genetics
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Peter C. de Greef
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, Netherlands
| | | | - Claudia Benz
- Division of Immunology, Paul-Ehrlich-Institut, IMG53, Langen, Germany
| | - Simon Fillatreau
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Université Paris Cité, Faculté de Médecine, Paris, France
- AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Bernard Malissen
- Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - Fabien Agenès
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
- Inserm, Délégation Régionale Auvergne Rhône Alpes, 69500 Bron, France
| | - Rob J. de Boer
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, Netherlands
| | - Jörg Kirberg
- Division of Immunology, Paul-Ehrlich-Institut, IMG53, Langen, Germany
| |
Collapse
|
5
|
Burton OT, Bricard O, Tareen S, Gergelits V, Andrews S, Biggins L, Roca CP, Whyte C, Junius S, Brajic A, Pasciuto E, Ali M, Lemaitre P, Schlenner SM, Ishigame H, Brown BD, Dooley J, Liston A. The tissue-resident regulatory T cell pool is shaped by transient multi-tissue migration and a conserved residency program. Immunity 2024; 57:1586-1602.e10. [PMID: 38897202 DOI: 10.1016/j.immuni.2024.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/27/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024]
Abstract
The tissues are the site of many important immunological reactions, yet how the immune system is controlled at these sites remains opaque. Recent studies have identified Foxp3+ regulatory T (Treg) cells in non-lymphoid tissues with unique characteristics compared with lymphoid Treg cells. However, tissue Treg cells have not been considered holistically across tissues. Here, we performed a systematic analysis of the Treg cell population residing in non-lymphoid organs throughout the body, revealing shared phenotypes, transient residency, and common molecular dependencies. Tissue Treg cells from different non-lymphoid organs shared T cell receptor (TCR) sequences, with functional capacity to drive multi-tissue Treg cell entry and were tissue-agnostic on tissue homing. Together, these results demonstrate that the tissue-resident Treg cell pool in most non-lymphoid organs, other than the gut, is largely constituted by broadly self-reactive Treg cells, characterized by transient multi-tissue migration. This work suggests common regulatory mechanisms may allow pan-tissue Treg cells to safeguard homeostasis across the body.
Collapse
Affiliation(s)
- Oliver T Burton
- Department of Pathology, University of Cambridge, Cambridge, UK; VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium; Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Orian Bricard
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Samar Tareen
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Vaclav Gergelits
- Department of Pathology, University of Cambridge, Cambridge, UK; Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Simon Andrews
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Laura Biggins
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Carlos P Roca
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Carly Whyte
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Steffie Junius
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium
| | - Aleksandra Brajic
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium
| | - Emanuela Pasciuto
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium; University of Antwerp, Center of Molecular Neurology, Antwerp, Belgium
| | - Magda Ali
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Pierre Lemaitre
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium
| | - Susan M Schlenner
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium
| | - Harumichi Ishigame
- Laboratory for Tissue Dynamics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan; Near-InfraRed Photo-Immunotherapy Research Institute, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Brian D Brown
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James Dooley
- Department of Pathology, University of Cambridge, Cambridge, UK; VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium; Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Adrian Liston
- Department of Pathology, University of Cambridge, Cambridge, UK; VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium; Babraham Institute, Babraham Research Campus, Cambridge, UK.
| |
Collapse
|
6
|
Wang L, Yang S, Zhu G, Li J, Meng G, Chen X, Zhang M, Wang S, Li X, Pan Y, Huang Y, Wang L, Wu Y. Immunopeptidome mining reveals a novel ERS-induced target in T1D. Cell Mol Immunol 2024; 21:604-619. [PMID: 38689020 PMCID: PMC11143349 DOI: 10.1038/s41423-024-01150-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 03/03/2024] [Indexed: 05/02/2024] Open
Abstract
Autoreactive CD8+ T cells play a key role in type 1 diabetes (T1D), but the antigen spectrum that activates autoreactive CD8+ T cells remains unclear. Endoplasmic reticulum stress (ERS) has been implicated in β-cell autoantigen generation. Here, we analyzed the major histocompatibility complex class I (MHC-I)-associated immunopeptidome (MIP) of islet β-cells under steady and ERS conditions and found that ERS reshaped the MIP of β-cells and promoted the MHC-I presentation of a panel of conventional self-peptides. Among them, OTUB258-66 showed immunodominance, and the corresponding autoreactive CD8+ T cells were diabetogenic in nonobese diabetic (NOD) mice. High glucose intake upregulated pancreatic OTUB2 expression and amplified the OTUB258-66-specific CD8+ T-cell response in NOD mice. Repeated OTUB258-66 administration significantly reduced the incidence of T1D in NOD mice. Interestingly, peripheral blood mononuclear cells (PBMCs) from patients with T1D, but not from healthy controls, showed a positive IFN-γ response to human OTUB2 peptides. This study provides not only a new explanation for the role of ERS in promoting β-cell-targeted autoimmunity but also a potential target for the prevention and treatment of T1D. The data are available via ProteomeXchange with the identifier PXD041227.
Collapse
Affiliation(s)
- Lina Wang
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, 261053, China
- Department of Immunology, Medical College of Qingdao University, Qingdao, 266071, China
| | - Shushu Yang
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Gaohui Zhu
- Department of Endocrinology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Jie Li
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Gang Meng
- Department of Pathology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiaoling Chen
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Mengjun Zhang
- Department of Pharmaceutical Analysis, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Shufeng Wang
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiangqian Li
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yu Pan
- Department of Endocrinology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Yi Huang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Li Wang
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Yuzhang Wu
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
7
|
Finnigan JP, Newman JH, Patskovsky Y, Patskovska L, Ishizuka AS, Lynn GM, Seder RA, Krogsgaard M, Bhardwaj N. Structural basis for self-discrimination by neoantigen-specific TCRs. Nat Commun 2024; 15:2140. [PMID: 38459027 PMCID: PMC10924104 DOI: 10.1038/s41467-024-46367-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/26/2024] [Indexed: 03/10/2024] Open
Abstract
T cell receptors (TCR) are pivotal in mediating tumour cell cytolysis via recognition of mutation-derived tumour neoantigens (neoAgs) presented by major histocompatibility class-I (MHC-I). Understanding the factors governing the emergence of neoAg from somatic mutations is a major focus of current research. However, the structural and cellular determinants controlling TCR recognition of neoAgs remain poorly understood. This study describes the multi-level analysis of a model neoAg from the B16F10 murine melanoma, H2-Db/Hsf2 p.K72N68-76, as well as its cognate TCR 47BE7. Through cellular, molecular and structural studies we demonstrate that the p.K72N mutation enhances H2-Db binding, thereby improving cell surface presentation and stabilizing the TCR 47BE7 epitope. Furthermore, TCR 47BE7 exhibited high functional avidity and selectivity, attributable to a broad, stringent, binding interface enabling recognition of native B16F10 despite low antigen density. Our findings provide insight into the generation of anchor-residue modified neoAg, and emphasize the value of molecular and structural investigations of neoAg in diverse MHC-I contexts for advancing the understanding of neoAg immunogenicity.
Collapse
Affiliation(s)
- John P Finnigan
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Pl., New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave., New York, NY, USA
- Department of Medicine, Division of Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY, USA
- Department of Surgery, Division of Thoracic and Cardiac Surgery, Brigham and Women's Hospital, 75 Francis St., Boston, MA, USA
| | - Jenna H Newman
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Pl., New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave., New York, NY, USA
- Department of Medicine, Division of Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY, USA
| | - Yury Patskovsky
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York, NY, USA
| | - Larysa Patskovska
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York, NY, USA
| | - Andrew S Ishizuka
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Barinthus Biotherapeutics, Germantown, MD, USA
| | - Geoffrey M Lynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Barinthus Biotherapeutics, Germantown, MD, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michelle Krogsgaard
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA.
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York, NY, USA.
| | - Nina Bhardwaj
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Pl., New York, NY, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave., New York, NY, USA.
- Department of Medicine, Division of Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, Francisco, CA, USA.
| |
Collapse
|
8
|
Balasco N, Tagliamonte M, Buonaguro L, Vitagliano L, Paladino A. Structural and Dynamic-Based Characterization of the Recognition Patterns of E7 and TRP-2 Epitopes by MHC Class I Receptors through Computational Approaches. Int J Mol Sci 2024; 25:1384. [PMID: 38338663 PMCID: PMC10855917 DOI: 10.3390/ijms25031384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
A detailed comprehension of MHC-epitope recognition is essential for the design and development of new antigens that could be effectively used in immunotherapy. Yet, the high variability of the peptide together with the large abundance of MHC variants binding makes the process highly specific and large-scale characterizations extremely challenging by standard experimental techniques. Taking advantage of the striking predictive accuracy of AlphaFold, we report a structural and dynamic-based strategy to gain insights into the molecular basis that drives the recognition and interaction of MHC class I in the immune response triggered by pathogens and/or tumor-derived peptides. Here, we investigated at the atomic level the recognition of E7 and TRP-2 epitopes to their known receptors, thus offering a structural explanation for the different binding preferences of the studied receptors for specific residues in certain positions of the antigen sequences. Moreover, our analysis provides clues on the determinants that dictate the affinity of the same epitope with different receptors. Collectively, the data here presented indicate the reliability of the approach that can be straightforwardly extended to a large number of related systems.
Collapse
Affiliation(s)
- Nicole Balasco
- Institute of Molecular Biology and Pathology IBPM-CNR c/o Department Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| | - Maria Tagliamonte
- Immunological Models Lab, Istituto Nazionale Tumori—Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)—“Fond. G. Pascale”, Via Mariano Semmola 53, 80131 Napoli, Italy; (M.T.); (L.B.)
| | - Luigi Buonaguro
- Immunological Models Lab, Istituto Nazionale Tumori—Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)—“Fond. G. Pascale”, Via Mariano Semmola 53, 80131 Napoli, Italy; (M.T.); (L.B.)
| | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging IBB-CNR, Via Pietro Castellino 111, 80131 Napoli, Italy;
| | - Antonella Paladino
- Institute of Biostructures and Bioimaging IBB-CNR, Via Pietro Castellino 111, 80131 Napoli, Italy;
| |
Collapse
|
9
|
Huang X, Gan Z, Cui H, Lan T, Liu Y, Caron E, Shao W. The SysteMHC Atlas v2.0, an updated resource for mass spectrometry-based immunopeptidomics. Nucleic Acids Res 2024; 52:D1062-D1071. [PMID: 38000392 PMCID: PMC10767952 DOI: 10.1093/nar/gkad1068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/16/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
The SysteMHC Atlas v1.0 was the first public repository dedicated to mass spectrometry-based immunopeptidomics. Here we introduce a newly released version of the SysteMHC Atlas v2.0 (https://systemhc.sjtu.edu.cn), a comprehensive collection of 7190 MS files from 303 allotypes. We extended and optimized a computational pipeline that allows the identification of MHC-bound peptides carrying on unexpected post-translational modifications (PTMs), thereby resulting in 471K modified peptides identified over 60 distinct PTM types. In total, we identified approximately 1.0 million and 1.1 million unique peptides for MHC class I and class II immunopeptidomes, respectively, indicating a 6.8-fold increase and a 28-fold increase to those in v1.0. The SysteMHC Atlas v2.0 introduces several new features, including the inclusion of non-UniProt peptides, and the incorporation of several novel computational tools for FDR estimation, binding affinity prediction and motif deconvolution. Additionally, we enhanced the user interface, upgraded website framework, and provided external links to other resources related. Finally, we built and provided various spectral libraries as community resources for data mining and future immunopeptidomic and proteomic analysis. We believe that the SysteMHC Atlas v2.0 is a unique resource to provide key insights to the immunology and proteomics community and will accelerate the development of vaccines and immunotherapies.
Collapse
Affiliation(s)
- Xiaoxiang Huang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Science & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ziao Gan
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Science & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haowei Cui
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Science & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tian Lan
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Science & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yansheng Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Etienne Caron
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Wenguang Shao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Science & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
10
|
Stutzmann C, Peng J, Wu Z, Savoie C, Sirois I, Thibault P, Wheeler AR, Caron E. Unlocking the potential of microfluidics in mass spectrometry-based immunopeptidomics for tumor antigen discovery. CELL REPORTS METHODS 2023; 3:100511. [PMID: 37426761 PMCID: PMC10326451 DOI: 10.1016/j.crmeth.2023.100511] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
The identification of tumor-specific antigens (TSAs) is critical for developing effective cancer immunotherapies. Mass spectrometry (MS)-based immunopeptidomics has emerged as a powerful tool for identifying TSAs as physical molecules. However, current immunopeptidomics platforms face challenges in measuring low-abundance TSAs in a precise, sensitive, and reproducible manner from small needle-tissue biopsies (<1 mg). Inspired by recent advances in single-cell proteomics, microfluidics technology offers a promising solution to these limitations by providing improved isolation of human leukocyte antigen (HLA)-associated peptides with higher sensitivity. In this context, we highlight the challenges in sample preparation and the rationale for developing microfluidics technology in immunopeptidomics. Additionally, we provide an overview of promising microfluidic methods, including microchip pillar arrays, valved-based systems, droplet microfluidics, and digital microfluidics, and discuss the latest research on their application in MS-based immunopeptidomics and single-cell proteomics.
Collapse
Affiliation(s)
| | - Jiaxi Peng
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - Zhaoguan Wu
- CHU Sainte Justine Research Center, Montreal, QC, Canada
| | | | | | - Pierre Thibault
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
- Department of Chemistry, University of Montreal, Montreal, QC, Canada
| | - Aaron R. Wheeler
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Etienne Caron
- CHU Sainte Justine Research Center, Montreal, QC, Canada
- Department of Pathology and Cellular Biology, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
11
|
Admon A. The biogenesis of the immunopeptidome. Semin Immunol 2023; 67:101766. [PMID: 37141766 DOI: 10.1016/j.smim.2023.101766] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
The immunopeptidome is the repertoire of peptides bound and presented by the MHC class I, class II, and non-classical molecules. The peptides are produced by the degradation of most cellular proteins, and in some cases, peptides are produced from extracellular proteins taken up by the cells. This review attempts to first describe some of its known and well-accepted concepts, and next, raise some questions about a few of the established dogmas in this field: The production of novel peptides by splicing is questioned, suggesting here that spliced peptides are extremely rare, if existent at all. The degree of the contribution to the immunopeptidome by degradation of cellular protein by the proteasome is doubted, therefore this review attempts to explain why it is likely that this contribution to the immunopeptidome is possibly overstated. The contribution of defective ribosome products (DRiPs) and non-canonical peptides to the immunopeptidome is noted and methods are suggested to quantify them. In addition, the common misconception that the MHC class II peptidome is mostly derived from extracellular proteins is noted, and corrected. It is stressed that the confirmation of sequence assignments of non-canonical and spliced peptides should rely on targeted mass spectrometry using spiking-in of heavy isotope-labeled peptides. Finally, the new methodologies and modern instrumentation currently available for high throughput kinetics and quantitative immunopeptidomics are described. These advanced methods open up new possibilities for utilizing the big data generated and taking a fresh look at the established dogmas and reevaluating them critically.
Collapse
Affiliation(s)
- Arie Admon
- Faculty of Biology, Technion-Israel Institute of Technology, Israel.
| |
Collapse
|
12
|
Son ET, Paul-Heng M, Leong M, Wang C, Hill AE, Denkova M, Purcell AW, Mifsud NA, Sharland AF. Screening self-peptides for recognition by mouse alloreactive CD8 + T cells using direct ex vivo multimer staining. STAR Protoc 2023; 4:101943. [PMID: 36525346 PMCID: PMC9792548 DOI: 10.1016/j.xpro.2022.101943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/31/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022] Open
Abstract
Here, we present a protocol to identify immunogenic self-peptide/allogeneic major histocompatibility complex (MHC) epitopes. We describe the generation of enriched alloreactive CD8+ T cells by priming mice with a skin graft expressing the allogeneic MHC class I molecule of interest, followed by boosting with a liver-specific AAV vector encoding the heavy chain of that donor MHC allomorph. We then use a peptide-exchange approach to assemble a range of peptide-MHC (pMHC) multimers for measuring recognition of the various epitopes by these alloreactive T cells. For complete details on the use and execution of this protocol, please refer to Son et al. (2021).1.
Collapse
Affiliation(s)
- Eric T Son
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia
| | - Moumita Paul-Heng
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia
| | - Mario Leong
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia
| | - Chuanmin Wang
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia
| | - Alexandra E Hill
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia
| | - Martina Denkova
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia
| | - Anthony W Purcell
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Nicole A Mifsud
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Alexandra F Sharland
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia.
| |
Collapse
|
13
|
Shapiro IE, Bassani-Sternberg M. The impact of immunopeptidomics: From basic research to clinical implementation. Semin Immunol 2023; 66:101727. [PMID: 36764021 DOI: 10.1016/j.smim.2023.101727] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023]
Abstract
The immunopeptidome is the set of peptides presented by the major histocompatibility complex (MHC) molecules, in humans also known as the human leukocyte antigen (HLA), on the surface of cells that mediate T-cell immunosurveillance. The immunopeptidome is a sampling of the cellular proteome and hence it contains information about the health state of cells. The peptide repertoire is influenced by intra- and extra-cellular perturbations - such as in the case of drug exposure, infection, or oncogenic transformation. Immunopeptidomics is the bioanalytical method by which the presented peptides are extracted from biological samples and analyzed by high-performance liquid chromatography coupled to tandem mass spectrometry (MS), resulting in a deep qualitative and quantitative snapshot of the immunopeptidome. In this review, we discuss published immunopeptidomics studies from recent years, grouped into three main domains: i) basic, ii) pre-clinical and iii) clinical research and applications. We review selected fundamental immunopeptidomics studies on the antigen processing and presentation machinery, on HLA restriction and studies that advanced our understanding of various diseases, and how exploration of the antigenic landscape allowed immune targeting at the pre-clinical stage, paving the way to pioneering exploratory clinical trials where immunopeptidomics is directly implemented in the conception of innovative treatments for cancer patients.
Collapse
Affiliation(s)
- Ilja E Shapiro
- Ludwig Institute for Cancer Research, University of Lausanne, 1005 Lausanne, Switzerland; Department of Oncology, Centre hospitalier universitaire vaudois (CHUV), 1005 Lausanne, Switzerland; Agora Cancer Research Centre, 1011 Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Ludwig Institute for Cancer Research, University of Lausanne, 1005 Lausanne, Switzerland; Department of Oncology, Centre hospitalier universitaire vaudois (CHUV), 1005 Lausanne, Switzerland; Agora Cancer Research Centre, 1011 Lausanne, Switzerland; Center of Experimental Therapeutics, Department of Oncology, Centre hospitalier universitaire vaudois (CHUV), 1005 Lausanne, Switzerland.
| |
Collapse
|
14
|
Contemplating immunopeptidomes to better predict them. Semin Immunol 2023; 66:101708. [PMID: 36621290 DOI: 10.1016/j.smim.2022.101708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 01/09/2023]
Abstract
The identification of T-cell epitopes is key for a complete molecular understanding of immune recognition mechanisms in infectious diseases, autoimmunity and cancer. T-cell epitopes further provide targets for personalized vaccines and T-cell therapy, with several therapeutic applications in cancer immunotherapy and elsewhere. T-cell epitopes consist of short peptides displayed on Major Histocompatibility Complex (MHC) molecules. The recent advances in mass spectrometry (MS) based technologies to profile the ensemble of peptides displayed on MHC molecules - the so-called immunopeptidome - had a major impact on our understanding of antigen presentation and MHC ligands. On the one hand, these techniques enabled researchers to directly identify hundreds of thousands of peptides presented on MHC molecules, including some that elicited T-cell recognition. On the other hand, the data collected in these experiments revealed fundamental properties of antigen presentation pathways and significantly improved our ability to predict naturally presented MHC ligands and T-cell epitopes across the wide spectrum of MHC alleles found in human and other organisms. Here we review recent computational developments to analyze experimentally determined immunopeptidomes and harness these data to improve our understanding of antigen presentation and MHC binding specificities, as well as our ability to predict MHC ligands. We further discuss the strengths and limitations of the latest approaches to move beyond predictions of antigen presentation and tackle the challenges of predicting TCR recognition and immunogenicity.
Collapse
|
15
|
Shahbazy M, Ramarathinam SH, Illing PT, Jappe EC, Faridi P, Croft NP, Purcell AW. Benchmarking bioinformatics pipelines in data-independent acquisition mass spectrometry for immunopeptidomics. Mol Cell Proteomics 2023; 22:100515. [PMID: 36796644 PMCID: PMC10060114 DOI: 10.1016/j.mcpro.2023.100515] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 01/26/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
Immunopeptidomes are the peptide repertoires bound by the molecules encoded by the major histocompatibility complex (MHC) (human leukocyte antigen (HLA) in humans). These HLA-peptide complexes are presented on the cell surface for immune T-cell recognition. Immunopeptidomics denotes the utilization of tandem mass spectrometry (MS/MS) to identify and quantify peptides bound to HLA molecules. Data-independent acquisition (DIA) has emerged as a powerful strategy for quantitative proteomics and deep proteome-wide identification; however, DIA application to immunopeptidomics analyses has so far seen limited use. Further, of the many DIA data processing tools currently available, there is no consensus in the immunopeptidomics community on the most appropriate pipeline(s) for in-depth and accurate HLA peptide identification. Herein, we benchmarked four commonly used spectral library-based DIA pipelines developed for proteomics applications (Skyline, Spectronaut, DIA-NN, and PEAKS) for their ability to perform immunopeptidome quantification. We validated and assessed the capability of each tool to identify and quantify HLA-bound peptides. Generally, DIA-NN and PEAKS provided higher immunopeptidome coverage with more reproducible results. Skyline and Spectronaut conferred more accurate peptide identification with lower experimental false-positive rates. All tools demonstrated reasonable correlations in quantifying precursors of HLA-bound peptides. Our benchmarking study suggests a combined strategy of applying at least two complementary DIA software tools to achieve the greatest degree of confidence and in-depth coverage of immunopeptidome data.
Collapse
Affiliation(s)
- Mohammad Shahbazy
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Sri H Ramarathinam
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Patricia T Illing
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Emma C Jappe
- Evaxion Biotech, Bredgade 34E, DK-1260 Copenhagen, Denmark
| | - Pouya Faridi
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC 3800, Australia.
| | - Nathan P Croft
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia.
| | - Anthony W Purcell
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
16
|
Marin I, Boix O, Garcia-Garijo A, Sirois I, Caballe A, Zarzuela E, Ruano I, Attolini CSO, Prats N, López-Domínguez JA, Kovatcheva M, Garralda E, Muñoz J, Caron E, Abad M, Gros A, Pietrocola F, Serrano M. Cellular Senescence Is Immunogenic and Promotes Antitumor Immunity. Cancer Discov 2023. [PMID: 36302218 DOI: 10.1101/2022.06.05.494912] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
UNLABELLED Cellular senescence is a stress response that activates innate immune cells, but little is known about its interplay with the adaptive immune system. Here, we show that senescent cells combine several features that render them highly efficient in activating dendritic cells (DC) and antigen-specific CD8 T cells. This includes the release of alarmins, activation of IFN signaling, enhanced MHC class I machinery, and presentation of senescence-associated self-peptides that can activate CD8 T cells. In the context of cancer, immunization with senescent cancer cells elicits strong antitumor protection mediated by DCs and CD8 T cells. Interestingly, this protection is superior to immunization with cancer cells undergoing immunogenic cell death. Finally, the induction of senescence in human primary cancer cells also augments their ability to activate autologous antigen-specific tumor-infiltrating CD8 lymphocytes. Our study indicates that senescent cancer cells can be exploited to develop efficient and protective CD8-dependent antitumor immune responses. SIGNIFICANCE Our study shows that senescent cells are endowed with a high immunogenic potential-superior to the gold standard of immunogenic cell death. We harness these properties of senescent cells to trigger efficient and protective CD8-dependent antitumor immune responses. See related article by Chen et al., p. 432. This article is highlighted in the In This Issue feature, p. 247.
Collapse
Affiliation(s)
- Ines Marin
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Olga Boix
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Isabelle Sirois
- CHU Sainte-Justine Research Center, Montréal, Québec, Canada
| | - Adrià Caballe
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Irene Ruano
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Neus Prats
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - José A López-Domínguez
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marta Kovatcheva
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Javier Muñoz
- Spanish National Cancer Research Center, Madrid, Spain
| | - Etienne Caron
- CHU Sainte-Justine Research Center, Montréal, Québec, Canada
- Department of Pathology and Cellular Biology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - María Abad
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Alena Gros
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Federico Pietrocola
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| |
Collapse
|
17
|
Marin I, Boix O, Garcia-Garijo A, Sirois I, Caballe A, Zarzuela E, Ruano I, Attolini CSO, Prats N, López-Domínguez JA, Kovatcheva M, Garralda E, Muñoz J, Caron E, Abad M, Gros A, Pietrocola F, Serrano M. Cellular Senescence Is Immunogenic and Promotes Antitumor Immunity. Cancer Discov 2023; 13:410-431. [PMID: 36302218 PMCID: PMC7614152 DOI: 10.1158/2159-8290.cd-22-0523] [Citation(s) in RCA: 171] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/31/2022] [Accepted: 10/24/2022] [Indexed: 02/07/2023]
Abstract
Cellular senescence is a stress response that activates innate immune cells, but little is known about its interplay with the adaptive immune system. Here, we show that senescent cells combine several features that render them highly efficient in activating dendritic cells (DC) and antigen-specific CD8 T cells. This includes the release of alarmins, activation of IFN signaling, enhanced MHC class I machinery, and presentation of senescence-associated self-peptides that can activate CD8 T cells. In the context of cancer, immunization with senescent cancer cells elicits strong antitumor protection mediated by DCs and CD8 T cells. Interestingly, this protection is superior to immunization with cancer cells undergoing immunogenic cell death. Finally, the induction of senescence in human primary cancer cells also augments their ability to activate autologous antigen-specific tumor-infiltrating CD8 lymphocytes. Our study indicates that senescent cancer cells can be exploited to develop efficient and protective CD8-dependent antitumor immune responses. SIGNIFICANCE Our study shows that senescent cells are endowed with a high immunogenic potential-superior to the gold standard of immunogenic cell death. We harness these properties of senescent cells to trigger efficient and protective CD8-dependent antitumor immune responses. See related article by Chen et al., p. 432. This article is highlighted in the In This Issue feature, p. 247.
Collapse
Affiliation(s)
- Ines Marin
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Olga Boix
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Isabelle Sirois
- CHU Sainte-Justine Research Center, Montréal, Québec, Canada
| | - Adrià Caballe
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Irene Ruano
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Neus Prats
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - José A López-Domínguez
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marta Kovatcheva
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Javier Muñoz
- Spanish National Cancer Research Center, Madrid, Spain
| | - Etienne Caron
- CHU Sainte-Justine Research Center, Montréal, Québec, Canada
- Department of Pathology and Cellular Biology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - María Abad
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Alena Gros
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Federico Pietrocola
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| |
Collapse
|
18
|
Kacen A, Javitt A, Kramer MP, Morgenstern D, Tsaban T, Shmueli MD, Teo GC, da Veiga Leprevost F, Barnea E, Yu F, Admon A, Eisenbach L, Samuels Y, Schueler-Furman O, Levin Y, Nesvizhskii AI, Merbl Y. Post-translational modifications reshape the antigenic landscape of the MHC I immunopeptidome in tumors. Nat Biotechnol 2023; 41:239-251. [PMID: 36203013 PMCID: PMC11197725 DOI: 10.1038/s41587-022-01464-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 08/09/2022] [Indexed: 11/08/2022]
Abstract
Post-translational modification (PTM) of antigens provides an additional source of specificities targeted by immune responses to tumors or pathogens, but identifying antigen PTMs and assessing their role in shaping the immunopeptidome is challenging. Here we describe the Protein Modification Integrated Search Engine (PROMISE), an antigen discovery pipeline that enables the analysis of 29 different PTM combinations from multiple clinical cohorts and cell lines. We expanded the antigen landscape, uncovering human leukocyte antigen class I binding motifs defined by specific PTMs with haplotype-specific binding preferences and revealing disease-specific modified targets, including thousands of new cancer-specific antigens that can be shared between patients and across cancer types. Furthermore, we uncovered a subset of modified peptides that are specific to cancer tissue and driven by post-translational changes that occurred in the tumor proteome. Our findings highlight principles of PTM-driven antigenicity, which may have broad implications for T cell-mediated therapies in cancer and beyond.
Collapse
Affiliation(s)
- Assaf Kacen
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Aaron Javitt
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Matthias P Kramer
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - David Morgenstern
- De Botton Institute for Protein Profiling, Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Tomer Tsaban
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Merav D Shmueli
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Guo Ci Teo
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Eilon Barnea
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Arie Admon
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Lea Eisenbach
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Yishai Levin
- De Botton Institute for Protein Profiling, Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Yifat Merbl
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
19
|
Wang L, Li X, Yang S, Chen X, Li J, Wang S, Zhang M, Zheng Z, Zhou J, Wang L, Wu Y. Proteomic identification of MHC class I-associated peptidome derived from non-obese diabetic mouse thymus and pancreas. J Proteomics 2023; 270:104746. [PMID: 36210013 DOI: 10.1016/j.jprot.2022.104746] [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: 05/18/2022] [Revised: 09/17/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
Abstract
The peptides repertoire presented to CD8+ T cells by major histocompatibility complex (MHC) class I molecules is referred to as the MHC I-associated peptidome (MIP), which regulates thymus development, peripheral survival and function during lifetime of CD8+ T cells. Type 1 diabetes (T1D) is an organ-specific autoimmune disease caused by pancreatic β cells destruction mediated primarily by autoreactive CD8+ T cells. Non-obese diabetic (NOD) mouse is an important animal model of T1D. Here, we deeply analyzed the MIP derived from NOD mice thymus and pancreas, and demonstrated that the thymus MIP source proteins partially shared with the MIP source proteins derived from NOD mice pancreas and β cell line. One H-2Kd restricted peptide SLC35B126-34 which was shared by MIP derived from both NOD mice pancreatic tissues and islet β-cell line, but absent in MIP from NOD thymus tissues, showed ability to stimulate IFN-γ secretion and proliferation of NOD mice splenic CD8+ T cells. The global view of the MHC I-associated self-peptides repertoire in the thymus and pancreas of NOD mice may serve as a biological reference to identify potential autoantigens targeted by autoreactive CD8+ T cells in T1D. Data are available via ProteomeXchange with identifier PXD031966. SIGNIFICANCE: The peptides repertoire presented to CD8+ T cells by major histocompatibility complex (MHC) class I molecules is referred to as the MHC I-associated peptidome (MIP). The MIP presented by thymic antigen presenting cells (APCs) is crucial for shaping CD8+ T cell repertoire and self-tolerance, while the MIP presented by peripheral tissues and organs is not only involved in maintaining periphery CD8+ T cell survival and homeostasis, but also mediates immune surveillance and autoimmune responses of CD8+ T cells under pathological conditions. Type 1 diabetes (T1D) is an organ-specific autoimmune disease caused by the destruction of pancreatic β cells, mediated primarily by autoreactive CD8+ T cells. Non-obese diabetic (NOD) mouse is one of important animal models of spontaneous autoimmune diabetes that shares several key features with human T1D. The global view of the MHC I-associated self-peptides repertoire in the thymus and pancreas of NOD mice may serve as a good biological reference to identify potential autoantigens targeted by autoreactive CD8+ T cells in T1D. It has great significance for further clarifying the immune recognition and effect mechanism of autoreactive CD8+ T cells in the pathogenesis of T1D, and then developing antigen-specific immune intervention strategies.
Collapse
Affiliation(s)
- Lina Wang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, China; Institute of Immunology PLA & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China; Department of Immunology, College of Basic Medicine, Weifang Medical University, Weifang 261053, China
| | - Xiangqian Li
- Institute of Immunology PLA & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Shushu Yang
- Institute of Immunology PLA & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiaoling Chen
- Institute of Immunology PLA & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jie Li
- Institute of Immunology PLA & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Shufeng Wang
- Institute of Immunology PLA & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Mengjun Zhang
- Department of Pharmaceutical Analysis, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhengni Zheng
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jie Zhou
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Li Wang
- Institute of Immunology PLA & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Yuzhang Wu
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, China; Institute of Immunology PLA & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
20
|
Tcyganov EN, Sanseviero E, Marvel D, Beer T, Tang HY, Hembach P, Speicher DW, Zhang Q, Donthireddy LR, Mostafa A, Tsyganova S, Pisarev V, Laufer T, Ignatov D, Ferrone S, Meyer C, Maby-El Hajjami H, Speiser DE, Altiok S, Antonia S, Xu X, Xu W, Zheng C, Schuchter LM, Amaravadi RK, Mitchell TC, Karakousis GC, Yuan Z, Montaner LJ, Celis E, Gabrilovich DI. Peroxynitrite in the tumor microenvironment changes the profile of antigens allowing escape from cancer immunotherapy. Cancer Cell 2022; 40:1173-1189.e6. [PMID: 36220073 PMCID: PMC9566605 DOI: 10.1016/j.ccell.2022.09.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 06/12/2022] [Accepted: 08/31/2022] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy often depends on recognition of peptide epitopes by cytotoxic T lymphocytes (CTLs). The tumor microenvironment (TME) is enriched for peroxynitrite (PNT), a potent oxidant produced by infiltrating myeloid cells and some tumor cells. We demonstrate that PNT alters the profile of MHC class I bound peptides presented on tumor cells. Only CTLs specific for PNT-resistant peptides have a strong antitumor effect in vivo, whereas CTLs specific for PNT-sensitive peptides are not effective. Therapeutic targeting of PNT in mice reduces resistance of tumor cells to CTLs. Melanoma patients with low PNT activity in their tumors demonstrate a better clinical response to immunotherapy than patients with high PNT activity. Our data suggest that intratumoral PNT activity should be considered for the design of neoantigen-based therapy and also may be an important immunotherapeutic target.
Collapse
Affiliation(s)
- Evgenii N Tcyganov
- Immunology, Microenvironment, and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | | | - Douglas Marvel
- Immunology, Microenvironment, and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Thomas Beer
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Hsin-Yao Tang
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Peter Hembach
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - David W Speicher
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Qianfei Zhang
- AstraZeneca, ICC, Early Oncology, Gaithersburg, MD 20878, USA
| | | | - Ali Mostafa
- AstraZeneca, ICC, Early Oncology, Gaithersburg, MD 20878, USA
| | - Sabina Tsyganova
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vladimir Pisarev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia; Central Institute of Epidemiology, 111123 Moscow, Russia
| | - Terri Laufer
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dmitriy Ignatov
- Max Planck Unit for the Science of Pathogens, Charitéplatz 1, 10117 Berlin, Germany
| | - Soldano Ferrone
- Department of Surgery, Harvard University, Boston, MA 02114, USA
| | - Christiane Meyer
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | | | - Daniel E Speiser
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | | | | | - Xiaowei Xu
- Abramson Cancer Center, Department of Pathology and Molecular Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Wei Xu
- Abramson Cancer Center, Department of Pathology and Molecular Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Cathy Zheng
- Abramson Cancer Center, Department of Pathology and Molecular Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Lynn M Schuchter
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Ravi K Amaravadi
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Tara C Mitchell
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Giorgos C Karakousis
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Zhe Yuan
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Luis J Montaner
- Immunology, Microenvironment, and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Esteban Celis
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | | |
Collapse
|
21
|
Sandalova T, Sala BM, Achour A. Structural aspects of chemical modifications in the MHC-restricted immunopeptidome; Implications for immune recognition. Front Chem 2022; 10:861609. [PMID: 36017166 PMCID: PMC9395651 DOI: 10.3389/fchem.2022.861609] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/12/2022] [Indexed: 11/26/2022] Open
Abstract
Significant advances in mass-spectroscopy (MS) have made it possible to investigate the cellular immunopeptidome, a large collection of MHC-associated epitopes presented on the surface of healthy, stressed and infected cells. These approaches have hitherto allowed the unambiguous identification of large cohorts of epitope sequences that are restricted to specific MHC class I and II molecules, enhancing our understanding of the quantities, qualities and origins of these peptide populations. Most importantly these analyses provide essential information about the immunopeptidome in responses to pathogens, autoimmunity and cancer, and will hopefully allow for future tailored individual therapies. Protein post-translational modifications (PTM) play a key role in cellular functions, and are essential for both maintaining cellular homeostasis and increasing the diversity of the proteome. A significant proportion of proteins is post-translationally modified, and thus a deeper understanding of the importance of PTM epitopes in immunopeptidomes is essential for a thorough and stringent understanding of these peptide populations. The aim of the present review is to provide a structural insight into the impact of PTM peptides on stability of MHC/peptide complexes, and how these may alter/modulate immune responses.
Collapse
Affiliation(s)
- Tatyana Sandalova
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Section for Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Benedetta Maria Sala
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Section for Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Section for Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- *Correspondence: Adnane Achour,
| |
Collapse
|
22
|
Jaeger AM, Stopfer LE, Ahn R, Sanders EA, Sandel DA, Freed-Pastor WA, Rideout WM, Naranjo S, Fessenden T, Nguyen KB, Winter PS, Kohn RE, Westcott PMK, Schenkel JM, Shanahan SL, Shalek AK, Spranger S, White FM, Jacks T. Deciphering the immunopeptidome in vivo reveals new tumour antigens. Nature 2022; 607:149-155. [PMID: 35705813 PMCID: PMC9945857 DOI: 10.1038/s41586-022-04839-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 05/06/2022] [Indexed: 11/09/2022]
Abstract
Immunosurveillance of cancer requires the presentation of peptide antigens on major histocompatibility complex class I (MHC-I) molecules1-5. Current approaches to profiling of MHC-I-associated peptides, collectively known as the immunopeptidome, are limited to in vitro investigation or bulk tumour lysates, which limits our understanding of cancer-specific patterns of antigen presentation in vivo6. To overcome these limitations, we engineered an inducible affinity tag into the mouse MHC-I gene (H2-K1) and targeted this allele to the KrasLSL-G12D/+Trp53fl/fl mouse model (KP/KbStrep)7. This approach enabled us to precisely isolate MHC-I peptides from autochthonous pancreatic ductal adenocarcinoma and from lung adenocarcinoma (LUAD) in vivo. In addition, we profiled the LUAD immunopeptidome from the alveolar type 2 cell of origin up to late-stage disease. Differential peptide presentation in LUAD was not predictable by mRNA expression or translation efficiency and is probably driven by post-translational mechanisms. Vaccination with peptides presented by LUAD in vivo induced CD8+ T cell responses in naive mice and tumour-bearing mice. Many peptides specific to LUAD, including immunogenic peptides, exhibited minimal expression of the cognate mRNA, which prompts the reconsideration of antigen prediction pipelines that triage peptides according to transcript abundance8. Beyond cancer, the KbStrep allele is compatible with other Cre-driver lines to explore antigen presentation in vivo in the pursuit of understanding basic immunology, infectious disease and autoimmunity.
Collapse
Affiliation(s)
- Alex M Jaeger
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Lauren E Stopfer
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ryuhjin Ahn
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emma A Sanders
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Demi A Sandel
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - William A Freed-Pastor
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - William M Rideout
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Santiago Naranjo
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Tim Fessenden
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Kim B Nguyen
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Peter S Winter
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ryan E Kohn
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Peter M K Westcott
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Jason M Schenkel
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Sean-Luc Shanahan
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Alex K Shalek
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Stefani Spranger
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Forest M White
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tyler Jacks
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
23
|
Fessenden TB, Stopfer LE, Chatterjee F, Zulueta J, Mesfin J, Cordero Dumit T, Reijers I, Hoefsmit EP, Blank C, White F, Spranger S. Dendritic cell-mediated cross presentation of tumor-derived peptides is biased against plasma membrane proteins. J Immunother Cancer 2022; 10:jitc-2021-004159. [PMID: 35820727 PMCID: PMC9277371 DOI: 10.1136/jitc-2021-004159] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2022] [Indexed: 11/06/2022] Open
Abstract
Background For effective tumor elimination, cytotoxic CD8+ T cells must recognize tumor-derived antigens presented on class I major histocompatibility complex (MHC-I). Despite a general association between the expression of immunogenic antigens, typically neoantigens, and response to immunotherapy, the majority of patients lack strong endogenous responses to most putative neoantigens due to mechanisms that are not well understood. Cytotoxic CD8+ T-cell responses are induced by dendritic cells (DCs) cross-presenting tumor-derived peptides on MHC-I. We hypothesized that cross presentation may form an unappreciated source of bias in the induction of cytotoxic T-cell responses. Methods We used stable isotope labeling of amino acids combined with immunopeptidomics to distinguish cross-presented from endogenous MHC-I peptides on DCs. To test impacts on T-cell activation, we targeted the model antigen SIINFEKL to specific subcellular compartments in tumor cells, which were used as sources for cross presentation to T cells. In vitro observations were validated using DNA and RNA sequencing data from two cohorts of patients with melanoma undergoing checkpoint blockade therapy. We used a novel quantitative mass spectrometry approach to measure the levels of model antigen on cross-presenting DCs following various means of tumor cell death. Results DCs exhibited a strong bias for cross-presenting peptides derived from cytoplasmic proteins and against those from plasma membrane proteins, which was confirmed using the model antigen SIINFEKL. In patients with melanoma, the proportion of membrane-derived neoantigens was correlated with reduced survival and failure to respond to therapy. Quantification of cross-presented SIINFEKL revealed that the mode of cell death could overcome DCs’ bias against plasma membrane proteins. Conclusions Cross presentation of cellular antigens by DCs may impose constraints on the range of peptides available to activate CD8+ T cells that have previously gone unappreciated. The share of neoantigens arising from membrane-derived sources may render some tumors less immunogenic due to inefficient cross presentation. These observations carry important implications for the encounter and intracellular processing of cellular antigens by DCs and merit further clinical studies for their therapeutic potential in stratifying patient populations and design of vaccine-based therapies.
Collapse
Affiliation(s)
- Tim B Fessenden
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Lauren E Stopfer
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Fiona Chatterjee
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Julian Zulueta
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Josh Mesfin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Therese Cordero Dumit
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Irene Reijers
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Esmee P Hoefsmit
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Christian Blank
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Forest White
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA .,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
24
|
Feola S, Chiaro J, Martins B, Russo S, Fusciello M, Ylösmäki E, Bonini C, Ruggiero E, Hamdan F, Feodoroff M, Antignani G, Viitala T, Pesonen S, Grönholm M, Branca RMM, Lehtiö J, Cerullo V. A novel immunopeptidomic-based pipeline for the generation of personalized oncolytic cancer vaccines. eLife 2022; 11:71156. [PMID: 35314027 PMCID: PMC8989416 DOI: 10.7554/elife.71156] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 03/01/2022] [Indexed: 12/16/2022] Open
Abstract
Besides the isolation and identification of major histocompatibility complex I-restricted peptides from the surface of cancer cells, one of the challenges is eliciting an effective antitumor CD8+ T-cell-mediated response as part of therapeutic cancer vaccine. Therefore, the establishment of a solid pipeline for the downstream selection of clinically relevant peptides and the subsequent creation of therapeutic cancer vaccines are of utmost importance. Indeed, the use of peptides for eliciting specific antitumor adaptive immunity is hindered by two main limitations: the efficient selection of the most optimal candidate peptides and the use of a highly immunogenic platform to combine with the peptides to induce effective tumor-specific adaptive immune responses. Here, we describe for the first time a streamlined pipeline for the generation of personalized cancer vaccines starting from the isolation and selection of the most immunogenic peptide candidates expressed on the tumor cells and ending in the generation of efficient therapeutic oncolytic cancer vaccines. This immunopeptidomics-based pipeline was carefully validated in a murine colon tumor model CT26. Specifically, we used state-of-the-art immunoprecipitation and mass spectrometric methodologies to isolate >8000 peptide targets from the CT26 tumor cell line. The selection of the target candidates was then based on two separate approaches: RNAseq analysis and HEX software. The latter is a tool previously developed by Jacopo, 2020, able to identify tumor antigens similar to pathogen antigens in order to exploit molecular mimicry and tumor pathogen cross-reactive T cells in cancer vaccine development. The generated list of candidates (26 in total) was further tested in a functional characterization assay using interferon-γ enzyme-linked immunospot (ELISpot), reducing the number of candidates to six. These peptides were then tested in our previously described oncolytic cancer vaccine platform PeptiCRAd, a vaccine platform that combines an immunogenic oncolytic adenovirus (OAd) coated with tumor antigen peptides. In our work, PeptiCRAd was successfully used for the treatment of mice bearing CT26, controlling the primary malignant lesion and most importantly a secondary, nontreated, cancer lesion. These results confirmed the feasibility of applying the described pipeline for the selection of peptide candidates and generation of therapeutic oncolytic cancer vaccine, filling a gap in the field of cancer immunotherapy, and paving the way to translate our pipeline into human therapeutic approach.
Collapse
Affiliation(s)
- Sara Feola
- Drug Research Program (DRP) ImmunoViroTherapy Lab, University of Helsinki, Helsinki, Finland
| | - Jacopo Chiaro
- Drug Research Program (DRP) ImmunoViroTherapy Lab, University of Helsinki, Helsinki, Finland
| | - Beatriz Martins
- Drug Research Program (DRP) ImmunoViroTherapy Lab, University of Helsinki, Helsinki, Finland
| | - Salvatore Russo
- Drug Research Program (DRP) ImmunoViroTherapy Lab, University of Helsinki, Helsinki, Finland
| | - Manlio Fusciello
- Drug Research Program (DRP) ImmunoViroTherapy Lab, University of Helsinki, Helsinki, Finland
| | - Erkko Ylösmäki
- Drug Research Program (DRP) ImmunoViroTherapy Lab, University of Helsinki, Helsinki, Finland
| | - Chiara Bonini
- Experimental Hematology Unit, University Vita e Salute San Raffaele, Milan, Italy
| | - Eliana Ruggiero
- Experimental Hematology Unit, University Vita e Salute San Raffaele, Milan, Italy
| | - Firas Hamdan
- Drug Research Program (DRP) ImmunoViroTherapy Lab, University of Helsinki, Helsinki, Finland
| | - Michaela Feodoroff
- Drug Research Program (DRP) ImmunoViroTherapy Lab, University of Helsinki, Helsinki, Finland
| | - Gabriella Antignani
- Drug Research Program (DRP) ImmunoViroTherapy Lab, University of Helsinki, Helsinki, Finland
| | - Tapani Viitala
- Pharmaceutical Biophysics Research Group, University of Helsinki, Helsinki, Finland
| | | | - Mikaela Grönholm
- Drug Research Program (DRP) ImmunoViroTherapy Lab, University of Helsinki, Helsinki, Finland
| | - Rui M M Branca
- Department of Oncology-Pathology, Karolinska Institutet, stockholm, Sweden
| | - Janne Lehtiö
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Vincenzo Cerullo
- ImmunoVirothearpy Lab, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
25
|
Kubiniok P, Marcu A, Bichmann L, Kuchenbecker L, Schuster H, Hamelin DJ, Duquette JD, Kovalchik KA, Wessling L, Kohlbacher O, Rammensee HG, Neidert MC, Sirois I, Caron E. Understanding the constitutive presentation of MHC class I immunopeptidomes in primary tissues. iScience 2022; 25:103768. [PMID: 35141507 PMCID: PMC8810409 DOI: 10.1016/j.isci.2022.103768] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 06/15/2021] [Accepted: 01/11/2022] [Indexed: 12/20/2022] Open
Abstract
Understanding the molecular principles that govern the composition of the MHC-I immunopeptidome across different primary tissues is fundamentally important to predict how T cells respond in different contexts in vivo. Here, we performed a global analysis of the MHC-I immunopeptidome from 29 to 19 primary human and mouse tissues, respectively. First, we observed that different HLA-A, HLA-B, and HLA-C allotypes do not contribute evenly to the global composition of the MHC-I immunopeptidome across multiple human tissues. Second, we found that tissue-specific and housekeeping MHC-I peptides share very distinct properties. Third, we discovered that proteins that are evolutionarily hyperconserved represent the primary source of the MHC-I immunopeptidome at the organism-wide scale. Fourth, we uncovered new components of the antigen processing and presentation network, including the carboxypeptidases CPE, CNDP1/2, and CPVL. Together, this study opens up new avenues toward a system-wide understanding of antigen presentation in vivo across mammalian species. Tissue-specific and housekeeping MHC class I peptides share distinct properties HLA-A, HLA-B, and HLA-C allotypes contribute very unevenly to the pool of class I peptides MHC-I immunopeptidomes are represented by evolutionarily conserved proteins An extended antigen processing and presentation pathway is uncovered
Collapse
Affiliation(s)
- Peter Kubiniok
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - Ana Marcu
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Baden-Württemberg, Germany
- Cluster of Excellence iFIT (EXC 2180), “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Baden-Württemberg, Germany
| | - Leon Bichmann
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Baden-Württemberg, Germany
- Applied Bioinformatics, Department of Computer Science, University of Tübingen, 72074 Tübingen, Baden-Württemberg, Germany
| | - Leon Kuchenbecker
- Applied Bioinformatics, Department of Computer Science, University of Tübingen, 72074 Tübingen, Baden-Württemberg, Germany
| | - Heiko Schuster
- Immatics Biotechnologies GmbH, 72076 Tübingen, Baden-Württemberg, Germany
| | - David J. Hamelin
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | | | | | - Laura Wessling
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - Oliver Kohlbacher
- Applied Bioinformatics, Department of Computer Science, University of Tübingen, 72074 Tübingen, Baden-Württemberg, Germany
- Institute for Bioinformatics and Medical Informatics, University of Tübingen, 72076 Tübingen, Baden-Württemberg, Germany
- Biomolecular Interactions, Max Planck Institute for Developmental Biology, 72076 Tübingen, Baden-Württemberg, Germany
- Cluster of Excellence Machine Learning in the Sciences (EXC 2064), University of Tübingen, 72074 Tübingen, Baden-Württemberg, Germany
- Translational Bioinformatics, University Hospital Tübingen, 72076 Tübingen, Baden-Württemberg, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Baden-Württemberg, Germany
- Cluster of Excellence iFIT (EXC 2180), “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Baden-Württemberg, Germany
- DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), 72076 Tübingen, Baden-Württemberg, Germany
| | - Marian C. Neidert
- Clinical Neuroscience Center and Department of Neurosurgery, University Hospital and University of Zürich, 8057&8091 Zürich, Switzerland
| | - Isabelle Sirois
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - Etienne Caron
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
- Department of Pathology and Cellular Biology, Faculty of Medicine, Université de Montréal, QC H3T 1J4, Canada
- Corresponding author
| |
Collapse
|
26
|
Barbosa CRR, Barton J, Shepherd AJ, Mishto M. Mechanistic diversity in MHC class I antigen recognition. Biochem J 2021; 478:4187-4202. [PMID: 34940832 PMCID: PMC8786304 DOI: 10.1042/bcj20200910] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 12/20/2022]
Abstract
Throughout its evolution, the human immune system has developed a plethora of strategies to diversify the antigenic peptide sequences that can be targeted by the CD8+ T cell response against pathogens and aberrations of self. Here we provide a general overview of the mechanisms that lead to the diversity of antigens presented by MHC class I complexes and their recognition by CD8+ T cells, together with a more detailed analysis of recent progress in two important areas that are highly controversial: the prevalence and immunological relevance of unconventional antigen peptides; and cross-recognition of antigenic peptides by the T cell receptors of CD8+ T cells.
Collapse
Affiliation(s)
- Camila R. R. Barbosa
- Centre for Inflammation Biology and Cancer Immunology (CIBCI) & Peter Gorer Department of Immunobiology, King's College London, SE1 1UL London, U.K
- Francis Crick Institute, NW1 1AT London, U.K
| | - Justin Barton
- Department of Biological Sciences and Institute of Structural and Molecular Biology, Birkbeck, University of London, WC1E 7HX London, U.K
| | - Adrian J. Shepherd
- Department of Biological Sciences and Institute of Structural and Molecular Biology, Birkbeck, University of London, WC1E 7HX London, U.K
| | - Michele Mishto
- Centre for Inflammation Biology and Cancer Immunology (CIBCI) & Peter Gorer Department of Immunobiology, King's College London, SE1 1UL London, U.K
- Francis Crick Institute, NW1 1AT London, U.K
| |
Collapse
|
27
|
Kovalchik KA, Ma Q, Wessling L, Saab F, Despault J, Kubiniok P, Hamelin DJ, Faridi P, Li C, Purcell AW, Jang A, Paramithiotis E, Tognetti M, Reiter L, Bruderer R, Lanoix J, Bonneil É, Courcelles M, Thibault P, Caron E, Sirois I. MhcVizPipe: A Quality Control Software for Rapid Assessment of Small- to Large-Scale Immunopeptidome Data Sets. Mol Cell Proteomics 2021; 21:100178. [PMID: 34798331 PMCID: PMC8717601 DOI: 10.1016/j.mcpro.2021.100178] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
Mass spectrometry (MS)-based immunopeptidomics is maturing into an automatized, high-throughput technology, producing small- to large-scale datasets of clinically relevant MHC class I- and II-associated peptides. Consequently, the development of quality control (QC) and quality assurance (QA) systems capable of detecting sample and/or measurement issues is important for instrument operators and scientists in charge of downstream data interpretation. Here, we created MhcVizPipe (MVP), a semi-automated QC software tool that enables rapid and simultaneous assessment of multiple MHC class I and II immunopeptidomic datasets generated by MS, including datasets generated from large sample cohorts. In essence, MVP provides a rapid and consolidated view of sample quality, composition and MHC-specificity to greatly accelerate the 'pass-fail' QC decision-making process toward data interpretation. MVP parallelizes the use of well-established immunopeptidomic algorithms (NetMHCpan, NetMHCIIpan and GibbsCluster) and rapidly generates organized and easy-to-understand reports in HTML format. The reports are fully portable and can be viewed on any computer with a modern web browser. MVP is intuitive to use and will find utility in any specialized immunopeptidomic laboratory and proteomics core facility that provides immunopeptidomic services to the community.
Collapse
Affiliation(s)
| | - Qing Ma
- School of Electrical Engineering and Computer Science, Faculty of Engineering, University of Ottawa, ON K1N 6N5, Canada
| | - Laura Wessling
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - Frederic Saab
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - Jérôme Despault
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - Peter Kubiniok
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - David J Hamelin
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - Pouya Faridi
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Chen Li
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Anthony W Purcell
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Anne Jang
- CellCarta, Montreal, QC H2X 3Y7, Canada
| | | | | | - Lukas Reiter
- Biognosys, Wagistrasse 21, 8952 Schlieren, Switzerland
| | | | - Joël Lanoix
- Institute of Research in Immunology and Cancer, Montreal, QC H3T 1J4, Canada
| | - Éric Bonneil
- Institute of Research in Immunology and Cancer, Montreal, QC H3T 1J4, Canada
| | - Mathieu Courcelles
- Institute of Research in Immunology and Cancer, Montreal, QC H3T 1J4, Canada
| | - Pierre Thibault
- Institute of Research in Immunology and Cancer, Montreal, QC H3T 1J4, Canada; Department of Chemistry, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Etienne Caron
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada; Department of Pathology and Cellular Biology, Faculty of Medicine, Université de Montréal, QC H3T 1J4, Canada.
| | - Isabelle Sirois
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada.
| |
Collapse
|
28
|
Stopfer L, D'Souza A, White F. 1,2,3, MHC: a review of mass-spectrometry-based immunopeptidomics methods for relative and absolute quantification of pMHCs. IMMUNO-ONCOLOGY TECHNOLOGY 2021; 11:100042. [PMID: 35756972 PMCID: PMC9216433 DOI: 10.1016/j.iotech.2021.100042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Quantitative mass-spectrometry-based methods to perform relative and absolute quantification of peptides in the immunopeptidome are growing in popularity as researchers aim to measure the dynamic nature of the peptide major histocompatibility complex repertoire and make copies-per-cell estimations of target antigens of interest. Multiple methods to carry out these experiments have been reported, each with unique advantages and limitations. This article describes existing methods and recent applications, offering guidance for improving quantitative accuracy and selecting an appropriate experimental set-up to maximize data quality and quantity.
Collapse
Affiliation(s)
- L.E. Stopfer
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA,Koch Institute for Integrative Cancer Research, Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, USA
| | - A.D. D'Souza
- Koch Institute for Integrative Cancer Research, Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, USA,Program in Health Sciences and Technology, Harvard Medical School and Massachusetts Institute of Technology, Boston, USA
| | - F.M. White
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA,Koch Institute for Integrative Cancer Research, Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, USA,Correspondence to: Prof. Forest M. White, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Tel: 617-258-8949
| |
Collapse
|
29
|
Lu SX, De Neef E, Thomas JD, Sabio E, Rousseau B, Gigoux M, Knorr DA, Greenbaum B, Elhanati Y, Hogg SJ, Chow A, Ghosh A, Xie A, Zamarin D, Cui D, Erickson C, Singer M, Cho H, Wang E, Lu B, Durham BH, Shah H, Chowell D, Gabel AM, Shen Y, Liu J, Jin J, Rhodes MC, Taylor RE, Molina H, Wolchok JD, Merghoub T, Diaz LA, Abdel-Wahab O, Bradley RK. Pharmacologic modulation of RNA splicing enhances anti-tumor immunity. Cell 2021; 184:4032-4047.e31. [PMID: 34171309 PMCID: PMC8684350 DOI: 10.1016/j.cell.2021.05.038] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/26/2021] [Accepted: 05/24/2021] [Indexed: 01/26/2023]
Abstract
Although mutations in DNA are the best-studied source of neoantigens that determine response to immune checkpoint blockade, alterations in RNA splicing within cancer cells could similarly result in neoepitope production. However, the endogenous antigenicity and clinical potential of such splicing-derived epitopes have not been tested. Here, we demonstrate that pharmacologic modulation of splicing via specific drug classes generates bona fide neoantigens and elicits anti-tumor immunity, augmenting checkpoint immunotherapy. Splicing modulation inhibited tumor growth and enhanced checkpoint blockade in a manner dependent on host T cells and peptides presented on tumor MHC class I. Splicing modulation induced stereotyped splicing changes across tumor types, altering the MHC I-bound immunopeptidome to yield splicing-derived neoepitopes that trigger an anti-tumor T cell response in vivo. These data definitively identify splicing modulation as an untapped source of immunogenic peptides and provide a means to enhance response to checkpoint blockade that is readily translatable to the clinic.
Collapse
Affiliation(s)
- Sydney X Lu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Emma De Neef
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - James D Thomas
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Erich Sabio
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Center for Immunotherapy and Precision-Oncology, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Benoit Rousseau
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Mathieu Gigoux
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - David A Knorr
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Benjamin Greenbaum
- Department of Epidemiology and Biostatistics, Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Yuval Elhanati
- Department of Epidemiology and Biostatistics, Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Simon J Hogg
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Andrew Chow
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Arnab Ghosh
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Abigail Xie
- Medical Scientist Training Program, Weill Cornell Medical School, New York, NY 10065, USA
| | - Dmitriy Zamarin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Daniel Cui
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Caroline Erickson
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Michael Singer
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Hana Cho
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Eric Wang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Bin Lu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Benjamin H Durham
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Harshal Shah
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Diego Chowell
- Center for Immunotherapy and Precision-Oncology, Cleveland Clinic, Cleveland, OH 44195, USA; The Precision Immunology Institute, The Tisch Cancer Institute, Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Austin M Gabel
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Yudao Shen
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jing Liu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Matthew C Rhodes
- The Warren Family Research Center for Drug Discovery and Development and the Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Richard E Taylor
- The Warren Family Research Center for Drug Discovery and Development and the Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Henrik Molina
- Proteomics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Jedd D Wolchok
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Taha Merghoub
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Luis A Diaz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA.
| | - Robert K Bradley
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
30
|
Peltonen K, Feola S, Umer HM, Chiaro J, Mermelekas G, Ylösmäki E, Pesonen S, Branca RMM, Lehtiö J, Cerullo V. Therapeutic Cancer Vaccination with Immunopeptidomics-Discovered Antigens Confers Protective Antitumor Efficacy. Cancers (Basel) 2021; 13:cancers13143408. [PMID: 34298622 PMCID: PMC8306067 DOI: 10.3390/cancers13143408] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022] Open
Abstract
Knowledge of clinically targetable tumor antigens is becoming vital for broader design and utility of therapeutic cancer vaccines. This information is obtained reliably by directly interrogating the MHC-I presented peptide ligands, the immunopeptidome, with state-of-the-art mass spectrometry. Our manuscript describes direct identification of novel tumor antigens for an aggressive triple-negative breast cancer model. Immunopeptidome profiling revealed 2481 unique antigens, among them a novel ERV antigen originating from an endogenous retrovirus element. The clinical benefit and tumor control potential of the identified tumor antigens and ERV antigen were studied in a preclinical model using two vaccine platforms and therapeutic settings. Prominent control of established tumors was achieved using an oncolytic adenovirus platform designed for flexible and specific tumor targeting, namely PeptiCRAd. Our study presents a pipeline integrating immunopeptidome analysis-driven antigen discovery with a therapeutic cancer vaccine platform for improved personalized oncolytic immunotherapy.
Collapse
Affiliation(s)
- Karita Peltonen
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland; (K.P.); (S.F.); (J.C.); (E.Y.)
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790 Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00790 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00790 Helsinki, Finland
| | - Sara Feola
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland; (K.P.); (S.F.); (J.C.); (E.Y.)
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790 Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00790 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00790 Helsinki, Finland
| | - Husen M. Umer
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, 171 65 Solna, Sweden; (H.M.U.); (G.M.); (R.M.M.B.)
| | - Jacopo Chiaro
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland; (K.P.); (S.F.); (J.C.); (E.Y.)
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790 Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00790 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00790 Helsinki, Finland
| | - Georgios Mermelekas
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, 171 65 Solna, Sweden; (H.M.U.); (G.M.); (R.M.M.B.)
| | - Erkko Ylösmäki
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland; (K.P.); (S.F.); (J.C.); (E.Y.)
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790 Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00790 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00790 Helsinki, Finland
| | | | - Rui M. M. Branca
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, 171 65 Solna, Sweden; (H.M.U.); (G.M.); (R.M.M.B.)
| | - Janne Lehtiö
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, 171 65 Solna, Sweden; (H.M.U.); (G.M.); (R.M.M.B.)
- Correspondence: (J.L.); (V.C.); Tel.: +46-8-5248-1416 (J.L.); +358-50-31-85754 (V.C.)
| | - Vincenzo Cerullo
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland; (K.P.); (S.F.); (J.C.); (E.Y.)
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00790 Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00790 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00790 Helsinki, Finland
- Correspondence: (J.L.); (V.C.); Tel.: +46-8-5248-1416 (J.L.); +358-50-31-85754 (V.C.)
| |
Collapse
|
31
|
Bauzá-Martinez J, Heck AJR, Wu W. HLA-B and cysteinylated ligands distinguish the antigen presentation landscape of extracellular vesicles. Commun Biol 2021; 4:825. [PMID: 34211107 PMCID: PMC8249458 DOI: 10.1038/s42003-021-02364-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles can modulate diverse processes ranging from proliferation and tissue repair, to chemo-resistance and cellular differentiation. With the advent of tissue and immunological targeting, extracellular vesicles are also increasingly viewed as promising vectors to deliver peptide-based cancer antigens to the human immune system. Despite the clinical relevance and therapeutic potential of such 'cell-free' approaches, the natural antigen presentation landscape exported in extracellular vesicles is still largely uncharted, due to the challenging nature of such preparations and analyses. In the context of therapeutic vesicle production, a critical evaluation of the similarity in vesicular antigen presentation is also urgently needed. In this work, we compared the HLA-I peptide ligandomes of extracellular vesicles against that of whole-cells of the same cell line. We found that extracellular vesicles not only over-represent HLA-B complexes and peptide ligands, but also cysteinylated peptides that may modulate immune responses. Collectively, these findings describe the pre-existing provision of vesicular HLA complexes that may be utilized to carry peptide vaccines, as well as the propensity for different peptide and post-translationally modified ligands to be presented, and will outline critical considerations in devising novel EV vaccination strategies.
Collapse
Affiliation(s)
- Julia Bauzá-Martinez
- grid.5477.10000000120346234Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands ,grid.4818.50000 0001 0791 5666Netherlands Proteomics Centre, Utrecht, The Netherlands
| | - Albert J. R. Heck
- grid.5477.10000000120346234Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands ,grid.4818.50000 0001 0791 5666Netherlands Proteomics Centre, Utrecht, The Netherlands
| | - Wei Wu
- grid.5477.10000000120346234Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands ,grid.4818.50000 0001 0791 5666Netherlands Proteomics Centre, Utrecht, The Netherlands
| |
Collapse
|
32
|
Borg DJ, Faridi P, Giam KL, Reeves P, Fotheringham AK, McCarthy DA, Leung S, Ward MS, Harcourt BE, Ayala R, Scheijen JL, Briskey D, Dudek NL, Schalkwijk CG, Steptoe R, Purcell AW, Forbes JM. Short Duration Alagebrium Chloride Therapy Prediabetes Does Not Inhibit Progression to Autoimmune Diabetes in an Experimental Model. Metabolites 2021; 11:426. [PMID: 34203471 PMCID: PMC8305727 DOI: 10.3390/metabo11070426] [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: 05/24/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/17/2022] Open
Abstract
Mechanisms by which advanced glycation end products (AGEs) contribute to type 1 diabetes (T1D) pathogenesis are poorly understood. Since life-long pharmacotherapy with alagebrium chloride (ALT) slows progression to experimental T1D, we hypothesized that acute ALT therapy delivered prediabetes, may be effective. However, in female, non-obese diabetic (NODShiLt) mice, ALT administered prediabetes (day 50-100) did not protect against experimental T1D. ALT did not decrease circulating AGEs or their precursors. Despite this, pancreatic β-cell function was improved, and insulitis and pancreatic CD45.1+ cell infiltration was reduced. Lymphoid tissues were unaffected. ALT pre-treatment, prior to transfer of primed GC98 CD8+ T cell receptor transgenic T cells, reduced blood glucose concentrations and delayed diabetes, suggesting islet effects rather than immune modulation by ALT. Indeed, ALT did not reduce interferon-γ production by leukocytes from ovalbumin-pre-immunised NODShiLt mice and NODscid recipients given diabetogenic ALT treated NOD splenocytes were not protected against T1D. To elucidate β-cell effects, NOD-derived MIN6N8 β-cell major histocompatibility complex (MHC) Class Ia surface antigens were examined using immunopeptidomics. Overall, no major changes in the immunopeptidome were observed during the various treatments with all peptides exhibiting allele specific consensus binding motifs. As expected, longer MHC Class Ia peptides were captured bound to H-2Db than H-2Kb under all conditions. Moreover, more 10-12 mer peptides were isolated from H-2Db after AGE modified bovine serum albumin (AGE-BSA) treatment, compared with bovine serum albumin (BSA) or AGE-BSA+ALT treatment. Proteomics of MIN6N8 cells showed enrichment of processes associated with catabolism, the immune system, cell cycling and presynaptic endocytosis with AGE-BSA compared with BSA treatments. These data show that short-term ALT intervention, given prediabetes, does not arrest experimental T1D but transiently impacts β-cell function.
Collapse
Affiliation(s)
- Danielle J. Borg
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia; (D.J.B.); (A.K.F.); (D.A.M.); (S.L.); (M.S.W.); (B.E.H.)
- Pregnancy and Development, Mater Research Institute, The University of Queensland, South Brisbane, QLD 4101, Australia
| | - Pouya Faridi
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (P.F.); (K.L.G.); (R.A.); (N.L.D.); (A.W.P.)
| | - Kai Lin Giam
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (P.F.); (K.L.G.); (R.A.); (N.L.D.); (A.W.P.)
| | - Peta Reeves
- Tolerance and Autoimmunity Group, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD 4102, Australia; (P.R.); (R.S.)
| | - Amelia K. Fotheringham
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia; (D.J.B.); (A.K.F.); (D.A.M.); (S.L.); (M.S.W.); (B.E.H.)
| | - Domenica A. McCarthy
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia; (D.J.B.); (A.K.F.); (D.A.M.); (S.L.); (M.S.W.); (B.E.H.)
| | - Sherman Leung
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia; (D.J.B.); (A.K.F.); (D.A.M.); (S.L.); (M.S.W.); (B.E.H.)
| | - Micheal S. Ward
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia; (D.J.B.); (A.K.F.); (D.A.M.); (S.L.); (M.S.W.); (B.E.H.)
| | - Brooke E. Harcourt
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC 3052, Australia
| | - Rochelle Ayala
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (P.F.); (K.L.G.); (R.A.); (N.L.D.); (A.W.P.)
| | - Jean L. Scheijen
- Laboratory for Metabolism and Vascular Medicine, Department of Internal Medicine, Maastricht University, 6211 Maastricht, The Netherlands; (J.L.S.); (C.G.S.)
- Cardiovascular Research Institute Maastricht, 6211 Maastricht, The Netherlands
| | - David Briskey
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD 4067, Australia;
| | - Nadine L. Dudek
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (P.F.); (K.L.G.); (R.A.); (N.L.D.); (A.W.P.)
| | - Casper G. Schalkwijk
- Laboratory for Metabolism and Vascular Medicine, Department of Internal Medicine, Maastricht University, 6211 Maastricht, The Netherlands; (J.L.S.); (C.G.S.)
- Cardiovascular Research Institute Maastricht, 6211 Maastricht, The Netherlands
| | - Raymond Steptoe
- Tolerance and Autoimmunity Group, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD 4102, Australia; (P.R.); (R.S.)
| | - Anthony W. Purcell
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (P.F.); (K.L.G.); (R.A.); (N.L.D.); (A.W.P.)
| | - Josephine M. Forbes
- Glycation and Diabetes Complications, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia; (D.J.B.); (A.K.F.); (D.A.M.); (S.L.); (M.S.W.); (B.E.H.)
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC 3052, Australia
- Mater Clinical School, The University of Queensland, Brisbane, QLD 4101, Australia
| |
Collapse
|
33
|
Nanaware PP, Jurewicz MM, Clement CC, Lu L, Santambrogio L, Stern LJ. Distinguishing Signal From Noise in Immunopeptidome Studies of Limiting-Abundance Biological Samples: Peptides Presented by I-A b in C57BL/6 Mouse Thymus. Front Immunol 2021; 12:658601. [PMID: 33995376 PMCID: PMC8116589 DOI: 10.3389/fimmu.2021.658601] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
Antigen presentation by MHC-II proteins in the thymus is central to selection of CD4 T cells, but analysis of the full repertoire of presented peptides responsible for positive and negative selection is complicated by the low abundance of antigen presenting cells. A key challenge in analysis of limiting abundance immunopeptidomes by mass spectrometry is distinguishing true MHC-binding peptides from co-eluting non-specifically bound peptides present in the mixture eluted from immunoaffinity-purified MHC molecules. Herein we tested several approaches to minimize the impact of non-specific background peptides, including analyzing eluates from isotype-control antibody-conjugated beads, considering only peptides present in nested sets, and using predicted binding motif analysis to identify core epitopes. We evaluated these methods using well-understood human cell line samples, and then applied them to analysis of the I-Ab presented immunopeptidome of the thymus of C57BL/6 mice, comparing this to the more easily characterized splenic B cell and dendritic cell populations. We identified a total of 3473 unique peptides eluted from the various tissues, using a data dependent acquisition strategy with a false-discovery rate of <1%. The immunopeptidomes presented in thymus as compared to splenic B cells and DCs identified shared and tissue-specific epitopes. A broader length distribution was observed for peptides presented in the thymus as compared to splenic B cells or DCs. Detailed analysis of 61 differentially presented peptides indicated a wider distribution of I-Ab binding affinities in thymus as compared to splenic B cells. These results suggest different constraints on antigen processing and presentation pathways in central versus peripheral tissues.
Collapse
Affiliation(s)
- Padma P. Nanaware
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Mollie M. Jurewicz
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Cristina C. Clement
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Liying Lu
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Lawrence J. Stern
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
34
|
Hammond S, Thomson P, Meng X, Naisbitt D. In-Vitro Approaches to Predict and Study T-Cell Mediated Hypersensitivity to Drugs. Front Immunol 2021; 12:630530. [PMID: 33927714 PMCID: PMC8076677 DOI: 10.3389/fimmu.2021.630530] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/17/2021] [Indexed: 01/11/2023] Open
Abstract
Mitigating the risk of drug hypersensitivity reactions is an important facet of a given pharmaceutical, with poor performance in this area of safety often leading to warnings, restrictions and withdrawals. In the last 50 years, efforts to diagnose, manage, and circumvent these obscure, iatrogenic diseases have resulted in the development of assays at all stages of a drugs lifespan. Indeed, this begins with intelligent lead compound selection/design to minimize the existence of deleterious chemical reactivity through exclusion of ominous structural moieties. Preclinical studies then investigate how compounds interact with biological systems, with emphasis placed on modeling immunological/toxicological liabilities. During clinical use, competent and accurate diagnoses are sought to effectively manage patients with such ailments, and pharmacovigilance datasets can be used for stratification of patient populations in order to optimise safety profiles. Herein, an overview of some of the in-vitro approaches to predict intrinsic immunogenicity of drugs and diagnose culprit drugs in allergic patients after exposure is detailed, with current perspectives and opportunities provided.
Collapse
Affiliation(s)
- Sean Hammond
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
- ApconiX, Alderley Park, Alderley Edge, United Kingdom
| | - Paul Thomson
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | - Xiaoli Meng
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | - Dean Naisbitt
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
35
|
Darrigrand R, Pierson A, Rouillon M, Renko D, Boulpicante M, Bouyssié D, Mouton-Barbosa E, Marcoux J, Garcia C, Ghosh M, Alami M, Apcher S. Isoginkgetin derivative IP2 enhances the adaptive immune response against tumor antigens. Commun Biol 2021; 4:269. [PMID: 33649389 PMCID: PMC7921396 DOI: 10.1038/s42003-021-01801-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 02/05/2021] [Indexed: 11/25/2022] Open
Abstract
The success of cancer immunotherapy relies on the induction of an immunoprotective response targeting tumor antigens (TAs) presented on MHC-I molecules. We demonstrated that the splicing inhibitor isoginkgetin and its water-soluble and non-toxic derivative IP2 act at the production stage of the pioneer translation products (PTPs). We showed that IP2 increases PTP-derived antigen presentation in cancer cells in vitro and impairs tumor growth in vivo. IP2 action is long-lasting and dependent on the CD8+ T cell response against TAs. We observed that the antigen repertoire displayed on MHC-I molecules at the surface of MCA205 fibrosarcoma is modified upon treatment with IP2. In particular, IP2 enhances the presentation of an exon-derived epitope from the tumor suppressor nischarin. The combination of IP2 with a peptide vaccine targeting the nischarin-derived epitope showed a synergistic antitumor effect in vivo. These findings identify the spliceosome as a druggable target for the development of epitope-based immunotherapies.
Collapse
Affiliation(s)
- Romain Darrigrand
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie des tumeurs et Immunothérapie, Villejuif, France
| | - Alison Pierson
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie des tumeurs et Immunothérapie, Villejuif, France
| | - Marine Rouillon
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie des tumeurs et Immunothérapie, Villejuif, France
- SATT Paris Saclay, Orsay, France
| | - Dolor Renko
- Université Paris-Saclay, CNRS, BioCIS, Châtenay-Malabry, France
| | - Mathilde Boulpicante
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie des tumeurs et Immunothérapie, Villejuif, France
| | - David Bouyssié
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Emmanuelle Mouton-Barbosa
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Julien Marcoux
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Camille Garcia
- Institut Jacques Monod, CNRS U7592 Université Paris Diderot, Paris, France
- Institut Pasteur, Unité de Spectrométrie de Masse pour la Biologie (MSBio), Centre de Ressources et Recherches Technologiques (C2RT), USR 2000 CNRS, Paris, France
| | - Michael Ghosh
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Mouad Alami
- Université Paris-Saclay, CNRS, BioCIS, Châtenay-Malabry, France
| | - Sébastien Apcher
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie des tumeurs et Immunothérapie, Villejuif, France.
| |
Collapse
|
36
|
Chen R, Fulton KM, Twine SM, Li J. IDENTIFICATION OF MHC PEPTIDES USING MASS SPECTROMETRY FOR NEOANTIGEN DISCOVERY AND CANCER VACCINE DEVELOPMENT. MASS SPECTROMETRY REVIEWS 2021; 40:110-125. [PMID: 31875992 DOI: 10.1002/mas.21616] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Immunotherapy with neoantigens presented by major histocompatibility complex (MHC) is one of the most promising approaches in cancer treatment. Using this approach, cancer vaccines can be designed to target tumor-specific mutations that are not found in normal tissues. Clinical trials have demonstrated an increased immune response and eradication of tumors after injecting synthetic peptides selected from the immunopeptidome. Although the sequence of MHC binding peptides can be predicted from genome sequencing and prediction algorithms, this approach results in large numbers of predicted peptides, requiring the confirmation by mass spectrometry (MS) analysis. Identification of MHC peptides by direct MS analysis of immunopeptidome is accurate and sensitive, with tens of thousands of unique peptides potentially identified from either cancer cell line or tumor tissue. Peptides with mutations can also be identified with patient-specific protein databases constructed from genome or transcriptome sequencing data. MS analysis also enables the characterization of the post-translational modifications (PTMs) of those antigens that cannot be predicted. Moreover, PTMs were found to be more efficient in triggering an immune response. In addition to reviewing recent advances in the identification of neoantigens using MS, the techniques for cancer vaccine candidate selection and formulation, vaccine delivery systems, and the potential for use in combination with other therapeutics are also discussed. It is anticipated that MS-based techniques will play an important role in future cancer vaccine development. © 2019 John Wiley & Sons Ltd. Mass Spec Rev 40:110-125, 2021.
Collapse
Affiliation(s)
- Rui Chen
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, K1A 0R6, Canada
| | - Kelly M Fulton
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, K1A 0R6, Canada
| | - Susan M Twine
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, K1A 0R6, Canada
| | - Jianjun Li
- Human Health Therapeutics Research Centre, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, K1A 0R6, Canada
| |
Collapse
|
37
|
Weerakoon H, Potriquet J, Shah AK, Reed S, Jayakody B, Kapil C, Midha MK, Moritz RL, Lepletier A, Mulvenna J, Miles JJ, Hill MM. A primary human T-cell spectral library to facilitate large scale quantitative T-cell proteomics. Sci Data 2020; 7:412. [PMID: 33230158 PMCID: PMC7683684 DOI: 10.1038/s41597-020-00744-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/15/2020] [Indexed: 12/23/2022] Open
Abstract
Data independent analysis (DIA) exemplified by sequential window acquisition of all theoretical mass spectra (SWATH-MS) provides robust quantitative proteomics data, but the lack of a public primary human T-cell spectral library is a current resource gap. Here, we report the generation of a high-quality spectral library containing data for 4,833 distinct proteins from human T-cells across genetically unrelated donors, covering ~24% proteins of the UniProt/SwissProt reviewed human proteome. SWATH-MS analysis of 18 primary T-cell samples using the new human T-cell spectral library reliably identified and quantified 2,850 proteins at 1% false discovery rate (FDR). In comparison, the larger Pan-human spectral library identified and quantified 2,794 T-cell proteins in the same dataset. As the libraries identified an overlapping set of proteins, combining the two libraries resulted in quantification of 4,078 human T-cell proteins. Collectively, this large data archive will be a useful public resource for human T-cell proteomic studies. The human T-cell library is available at SWATHAtlas and the data are available via ProteomeXchange (PXD019446 and PXD019542) and PeptideAtlas (PASS01587).
Collapse
Affiliation(s)
- Harshi Weerakoon
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, 4072, Australia
- Faculty of Medicine and Allied Sciences, Rajarata University of Sri Lanka, Saliyapura, 50000, Sri Lanka
| | - Jeremy Potriquet
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD, 4006, Australia
- SCIEX Australia Pty Ltd, Mt Waverley, VIC, 3149, Australia
| | - Alok K Shah
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD, 4006, Australia
- CSL Limited, 45 Poplar Rd, Parkville, VIC, 3052, Australia
| | - Sarah Reed
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4006, Australia
| | - Buddhika Jayakody
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4006, Australia
| | - Charu Kapil
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Mukul K Midha
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | | | - Ailin Lepletier
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD, 4006, Australia
- Institute for Glycomics, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Jason Mulvenna
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD, 4006, Australia
| | - John J Miles
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4878, Australia.
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, 4878, Australia.
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, 4878, Australia.
| | - Michelle M Hill
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD, 4006, Australia.
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4006, Australia.
| |
Collapse
|
38
|
Klatt MG, Mack KN, Bai Y, Aretz ZEH, Nathan LI, Mun SS, Dao T, Scheinberg DA. Solving an MHC allele-specific bias in the reported immunopeptidome. JCI Insight 2020; 5:141264. [PMID: 32897882 PMCID: PMC7566711 DOI: 10.1172/jci.insight.141264] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/31/2020] [Indexed: 12/31/2022] Open
Abstract
Identification of MHC class I–bound peptides by immunopurification of MHC complexes and subsequent analysis by mass spectrometry is crucial for understanding T cell immunology and immunotherapy. Investigation of the steps for the MHC ligand isolation process revealed biases in widely used isolation techniques toward peptides of lower hydrophobicity. As MHC ligand hydrophobicity correlates positively with immunogenicity, identification of more hydrophobic MHC ligands could potentially lead to more effective isolation of immunogenic peptides as targets for immunotherapies. We solved this problem by use of higher concentrations of acetonitrile for the separation of MHC ligands and their respective complexes. This increased overall MHC ligand identifications by 2-fold, increased detection of cancer germline antigen–derived peptides by 50%, and resulted in profound variations in isolation efficacy between different MHC alleles correlating with the hydrophobicity of their anchor residues. Overall, these insights enabled a more complete view of the immunopeptidome and overcame a systematic underrepresentation of these critical MHC ligands of high hydrophobicity. An approach is identified to prevent bias in the immunopeptidome towards MHC ligands of lower hydrophobicity and therefore immunogenicity.
Collapse
Affiliation(s)
- Martin G Klatt
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kyeara N Mack
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Pharmacology Department and
| | - Yang Bai
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Pharmacology Department and
| | - Zita E H Aretz
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Physiology Biophysics and Systems Biology Program, Weill Cornell Medicine, New York, New York, USA
| | - Levy I Nathan
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sung Soo Mun
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Tao Dao
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - David A Scheinberg
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Pharmacology Department and
| |
Collapse
|
39
|
In-depth mining of the immunopeptidome of an acute myeloid leukemia cell line using complementary ligand enrichment and data acquisition strategies. Mol Immunol 2020; 123:7-17. [PMID: 32387766 DOI: 10.1016/j.molimm.2020.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/07/2020] [Accepted: 04/12/2020] [Indexed: 12/15/2022]
Abstract
The identification of T cell epitopes derived from tumour specific antigens remains a significant challenge for the development of peptide-based vaccines and immunotherapies. The use of mass spectrometry-based approaches (immunopeptidomics) can provide powerful new avenues for the identification of such epitopes. In this study we report the use of complementary peptide antigen enrichment methods and a comprehensive mass spectrometric acquisition strategy to provide in-depth immunopeptidome data for the THP-1 cell line, a cell line used widely as a model of human leukaemia. To accomplish this, we combined robust experimental workflows that incorporated ultrafiltration or off-line reversed phase chromatography to enrich peptide ligand as well as a multifaceted data acquisition strategy using an Orbitrap Fusion LC-MS instrument. Using the combined datasets from the two ligand enrichment methods we gained significant depth in immunopeptidome coverage by identifying a total of 41,816 HLA class I peptides from THP-1 cells, including a significant number of peptides derived from different oncogenes or over expressed proteins associated with cancer. The physicochemical properties of the HLA-bound peptides dictated their recovery using the two ligand enrichment approaches and their distribution across the different precursor charge states considered in the data acquisition strategy. The data highlight the complementarity of the two enrichment procedures, and in cases where sample is not limiting, suggest that the combination of both approaches will yield the most comprehensive immunopeptidome information.
Collapse
|
40
|
Chen X, Wang S, Huang Y, Zhao X, Jia X, Meng G, Zheng Q, Zhang M, Wu Y, Wang L. Obesity Reshapes Visceral Fat-Derived MHC I Associated-Immunopeptidomes and Generates Antigenic Peptides to Drive CD8 + T Cell Responses. iScience 2020; 23:100977. [PMID: 32217358 PMCID: PMC7109454 DOI: 10.1016/j.isci.2020.100977] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/21/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022] Open
Abstract
Adaptive CD8+ T cells were observed to contribute to the initiation and progression of obesity-induced visceral adipose tissue (VAT) chronic inflammation that is critically linked to metabolic disorders. Numerous peptides presented by the major histocompatibility complex (MHC) class I molecules at the cell surface are collectively termed as MHC I-associated immunopeptidome (MIP) for the interaction with CD8+ T cells. We conducted the in-depth mapping of MIP of VAT from lean and obese mice using large-scale high-resolution mass spectrometry and observed that obesity significantly alters the landscape of VAT MIPs. Additionally, the obese VAT-exclusive MIP source proteome reflected a distinct obesity-associated signature. A peptide derived from lactate dehydrogenase A (LDHA) or B chain, named LDHA237-244, was identified as an obese VAT-exclusive immunogenic peptide that was capable of eliciting pro-inflammatory CD8+ T cells responses. Our findings suggest that certain immunogenic peptides generated by obesity may trigger CD8+ T cell-mediated VAT inflammation. Obesity reshapes the landscape of VAT-derived MIP The obese VAT-exclusive MIP reflects an obesity-associated signature An obese VAT-exclusive peptide LDHA237-244 can stimulate CD8+ T cell responses LDHA237-244-reactive CD8+ T cells were present in obese mice but not lean mice
Collapse
Affiliation(s)
- Xiaoling Chen
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Shufeng Wang
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yi Huang
- Biomedical Analysis Center, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xia Zhao
- Bioinformatics Center, Department of Microbiology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xu Jia
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing 400038, China; Department of Physiology, North Sichuan Medical College, Nanchong 637007, China
| | - Gang Meng
- Department of Pathology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Qian Zheng
- Department of Physiology, North Sichuan Medical College, Nanchong 637007, China
| | - Mengjun Zhang
- Department of Pharmaceutical Analysis, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yuzhang Wu
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Li Wang
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
41
|
Is T Cell Negative Selection a Learning Algorithm? Cells 2020; 9:cells9030690. [PMID: 32168897 PMCID: PMC7140671 DOI: 10.3390/cells9030690] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 11/28/2022] Open
Abstract
Our immune system can destroy most cells in our body, an ability that needs to be tightly controlled. To prevent autoimmunity, the thymic medulla exposes developing T cells to normal “self” peptides and prevents any responders from entering the bloodstream. However, a substantial number of self-reactive T cells nevertheless reaches the periphery, implying that T cells do not encounter all self peptides during this negative selection process. It is unclear if T cells can still discriminate foreign peptides from self peptides they haven’t encountered during negative selection. We use an “artificial immune system”—a machine learning model of the T cell repertoire—to investigate how negative selection could alter the recognition of self peptides that are absent from the thymus. Our model reveals a surprising new role for T cell cross-reactivity in this context: moderate T cell cross-reactivity should skew the post-selection repertoire towards peptides that differ systematically from self. Moreover, even some self-like foreign peptides can be distinguished provided that the peptides presented in the thymus are not too similar to each other. Thus, our model predicts that negative selection on a well-chosen subset of self peptides would generate a repertoire that tolerates even “unseen” self peptides better than foreign peptides. This effect would resemble a “generalization” process as it is found in learning systems. We discuss potential experimental approaches to test our theory.
Collapse
|
42
|
Vizcaíno JA, Kubiniok P, Kovalchik KA, Ma Q, Duquette JD, Mongrain I, Deutsch EW, Peters B, Sette A, Sirois I, Caron E. The Human Immunopeptidome Project: A Roadmap to Predict and Treat Immune Diseases. Mol Cell Proteomics 2020; 19:31-49. [PMID: 31744855 PMCID: PMC6944237 DOI: 10.1074/mcp.r119.001743] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/18/2019] [Indexed: 12/11/2022] Open
Abstract
The science that investigates the ensembles of all peptides associated to human leukocyte antigen (HLA) molecules is termed "immunopeptidomics" and is typically driven by mass spectrometry (MS) technologies. Recent advances in MS technologies, neoantigen discovery and cancer immunotherapy have catalyzed the launch of the Human Immunopeptidome Project (HIPP) with the goal of providing a complete map of the human immunopeptidome and making the technology so robust that it will be available in every clinic. Here, we provide a long-term perspective of the field and we use this framework to explore how we think the completion of the HIPP will truly impact the society in the future. In this context, we introduce the concept of immunopeptidome-wide association studies (IWAS). We highlight the importance of large cohort studies for the future and how applying quantitative immunopeptidomics at population scale may provide a new look at individual predisposition to common immune diseases as well as responsiveness to vaccines and immunotherapies. Through this vision, we aim to provide a fresh view of the field to stimulate new discussions within the community, and present what we see as the key challenges for the future for unlocking the full potential of immunopeptidomics in this era of precision medicine.
Collapse
Affiliation(s)
- Juan Antonio Vizcaíno
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Peter Kubiniok
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | | | - Qing Ma
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada; School of Electrical Engineering and Computer Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | | | - Ian Mongrain
- Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, QC, Canada; Montreal Heart Institute, Montreal, QC, Canada
| | - Eric W Deutsch
- Institute for Systems Biology, Seattle, Washington, 98109
| | - Bjoern Peters
- La Jolla Institute for Allergy and Immunology, La Jolla, California, 92037
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, La Jolla, California, 92037
| | - Isabelle Sirois
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - Etienne Caron
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada; Department of Pathology and Cellular Biology, Faculty of Medicine, Université de Montréal, QC H3T 1J4, Canada.
| |
Collapse
|
43
|
Shao W, Caron E, Pedrioli P, Aebersold R. The SysteMHC Atlas: a Computational Pipeline, a Website, and a Data Repository for Immunopeptidomic Analyses. Methods Mol Biol 2020; 2120:173-181. [PMID: 32124319 DOI: 10.1007/978-1-0716-0327-7_12] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mass spectrometry has emerged as the method of choice for the exploration of the immunopeptidome. Insights from the immunopeptidome promise novel cancer therapeutic approaches and a better understanding of the basic mechanisms of our immune system. To meet the computational demands from the steady gain in popularity and reach of mass spectrometry-based immunopeptidomics analysis, we created the SysteMHC Atlas project, a first-of-its-kind computational pipeline and resource repository dedicated to standardizing data analysis and public dissemination of immunopeptidomic datasets.
Collapse
Affiliation(s)
- Wenguang Shao
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
| | - Etienne Caron
- CHU Sainte-Justine Research Center, Montreal, QC, Canada. .,Department of Pathology and Cellular Biology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
| | - Patrick Pedrioli
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.,Faculty of Science, University of Zurich, Zurich, Switzerland
| |
Collapse
|
44
|
Heather JM, Myers PT, Shi F, Aziz-Zanjani MO, Mahoney KE, Perez M, Morin B, Brittsan C, Shabanowitz J, Hunt DF, Cobbold M. Murine xenograft bioreactors for human immunopeptidome discovery. Sci Rep 2019; 9:18558. [PMID: 31811195 PMCID: PMC6898210 DOI: 10.1038/s41598-019-54700-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/05/2019] [Indexed: 11/17/2022] Open
Abstract
The study of peptides presented by MHC class I and class II molecules is limited by the need for relatively large cell numbers, especially when studying post-translationally modified or otherwise rare peptide species. To overcome this problem, we pose the hypothesis that human cells grown as xenografts in immunodeficient mice should produce equivalent immunopeptidomes as cultured cells. Comparing human cell lines grown either in vitro or as murine xenografts, we show that the immunopeptidome is substantially preserved. Numerous features are shared across both sample types, including peptides and proteins featured, length distributions, and HLA-binding motifs. Peptides well-represented in both groups were from more abundant proteins, or those with stronger predicted HLA binding affinities. Samples grown in vivo also recapitulated a similar phospho-immunopeptidome, with common sequences being those found at high copy number on the cell surface. These data indicate that xenografts are indeed a viable methodology for the production of cells for immunopeptidomic discovery.
Collapse
Affiliation(s)
- James M Heather
- Center for Cancer Immunology, Massachusetts General Hospital, Boston, Massachusetts, USA.
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.
| | | | - Feng Shi
- Center for Cancer Immunology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Keira E Mahoney
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | | | | | | | - Jeffrey Shabanowitz
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - Donald F Hunt
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Mark Cobbold
- Center for Cancer Immunology, Massachusetts General Hospital, Boston, Massachusetts, USA.
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
45
|
Lorente E, Palomo C, Barnea E, Mir C, del Val M, Admon A, López D. Natural Spleen Cell Ligandome in Transporter Antigen Processing-Deficient Mice. J Proteome Res 2019; 18:3512-3520. [DOI: 10.1021/acs.jproteome.9b00416] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Elena Lorente
- Unidad de Presentación y Regulación Inmunes, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain
| | - Concepción Palomo
- Unidad de Presentación y Regulación Inmunes, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain
| | - Eilon Barnea
- Department of Biology, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Carmen Mir
- Unidad de Presentación y Regulación Inmunes, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain
| | - Margarita del Val
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain
| | - Arie Admon
- Department of Biology, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Daniel López
- Unidad de Presentación y Regulación Inmunes, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain
| |
Collapse
|
46
|
Murphy JP, Kim Y, Clements DR, Konda P, Schuster H, Kowalewski DJ, Paulo JA, Cohen AM, Stevanovic S, Gygi SP, Gujar S. Therapy-Induced MHC I Ligands Shape Neo-Antitumor CD8 T Cell Responses during Oncolytic Virus-Based Cancer Immunotherapy. J Proteome Res 2019; 18:2666-2675. [PMID: 31095916 DOI: 10.1021/acs.jproteome.9b00173] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Oncolytic viruses (OVs), known for their cancer-killing characteristics, also overturn tumor-associated defects in antigen presentation through the MHC class I pathway and induce protective neo-antitumor CD8 T cell responses. Nonetheless, whether OVs shape the tumor MHC-I ligandome remains unknown. Here, we investigated if an OV induces the presentation of novel MHC I-bound tumor antigens (termed tumor MHC-I ligands). Using comparative mass spectrometry (MS)-based MHC-I ligandomics, we determined differential tumor MHC-I ligand expression following treatment with oncolytic reovirus in a murine ovarian cancer model. In vitro, we found that reovirus changes the tumor ligandome of cancer cells. Concurrent multiplexed quantitative proteomics revealed that the reovirus-induced changes in tumor MHC-I ligand presentation were mostly independent of their source proteins. In an in vivo model, tumor MHC-I ligands induced by reovirus were detectable not only in tumor tissues but also the spleens (a source of antigen-presenting cells) of tumor-bearing mice. Most importantly, therapy-induced MHC-I ligands stimulated antigen-specific IFNγ responses in antitumor CD8 T cells from mice treated with reovirus. These data show that therapy-induced MHC-I ligands may shape underlying neo-antitumor CD8 T cell responses. As such, they should be considered in strategies promoting the efficacy of OV-based cancer immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | - Heiko Schuster
- Department of Immunology, Interfaculty Institute for Cell Biology , University of Tübingen , 72074 Tübingen , Germany.,Immatics Biotechnologies GmbH , 72076 Tübingen , Germany
| | - Daniel J Kowalewski
- Department of Immunology, Interfaculty Institute for Cell Biology , University of Tübingen , 72074 Tübingen , Germany.,Immatics Biotechnologies GmbH , 72076 Tübingen , Germany
| | - Joao A Paulo
- Department of Cell Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | | | - Stefan Stevanovic
- Department of Immunology, Interfaculty Institute for Cell Biology , University of Tübingen , 72074 Tübingen , Germany
| | - Steven P Gygi
- Department of Cell Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | | |
Collapse
|