1
|
Mauriello A, Cavalluzzo B, Ragone C, Tagliamonte M, Buonaguro L. Shared neoantigens' atlas for off-the-shelf cancer vaccine development. J Transl Med 2025; 23:558. [PMID: 40390041 PMCID: PMC12087128 DOI: 10.1186/s12967-025-06478-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 04/10/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND We have recently described that the most prevalent 100 mutations identified in human cancers, both single nucleotide variations (SNVs) and InDels, generate a handful number of shared mutated neoantigens (SNV and InDel-NeoAgs) in association with 5 HLA-A and 7 B haplotypes. METHODS In the present study, we expanded such analysis to 50 haplotypes in the three MHC class I loci (10 HLA-A, 27 HLA-B and 13 HLA-C), including all the mutated proteins identified in at least 5% of cancer patients. RESULTS Overall, the extended analysis identified 15 SNV-NeoAgs and 55 InDel-NeoAgs with a significant affinity improvement over the corresponding wt (DAI > 10). These targetable shared NeoAgs are prevalently derived from PIK3CAH1047R (6/15 SNV-NeoAgs) and LARP4BT163Hfs (30/55 InDel-NeoAgs). From the HLA perspective, the HLA-A*33:03 is associated with the largest number of SNV-NeoAgs (4/15 NeoAgs) and the HLA-B*58:01 is associated with the largest number of InDel-NeoAgs (16/55 NeoAgs). According to the distribution of each HLA haplotype in at least 10% of the regional populations, therapeutic cancer vaccines based on mutated shared SNV and InDel-NeoAgs, might be developed for COAD, STAD and UCEC cancers, with a global coverage, and for PAAD and UVM, with a regional coverage. CONCLUSIONS This represents the first in-depth analysis for the identification of a specific repertoire of shared mutated NeoAgs, most of which never reported before. Such shared SNV and InDel-NeoAgs are indispensable for the development of "off-the-shelf" cancer vaccines targeting a relevant percentage of cancers in a significant percentage of cancer patients worldwide.
Collapse
Affiliation(s)
- Angela Mauriello
- Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS - "Fond G. Pascale", Via Mariano Semmola, 52, Naples, Italy
| | - Beatrice Cavalluzzo
- Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS - "Fond G. Pascale", Via Mariano Semmola, 52, Naples, Italy
| | - Concetta Ragone
- Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS - "Fond G. Pascale", Via Mariano Semmola, 52, Naples, Italy
| | - Maria Tagliamonte
- Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS - "Fond G. Pascale", Via Mariano Semmola, 52, Naples, Italy.
| | - Luigi Buonaguro
- Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS - "Fond G. Pascale", Via Mariano Semmola, 52, Naples, Italy.
| |
Collapse
|
2
|
Shah BA, Holden JA, Lenzo JC, Hadjigol S, O'Brien-Simpson NM. Multi-disciplinary approaches paving the way for clinically effective peptide vaccines for cancer. NPJ Vaccines 2025; 10:68. [PMID: 40204832 PMCID: PMC11982186 DOI: 10.1038/s41541-025-01118-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
Cytotoxic CD8+ T lymphocyte (CTL) cells are central in mediating antitumor immunity. Induction of a robust CTL response requires, CTL interaction with professional antigen-presenting cells, such as dendritic cells, displaying onco-antigenic peptide, often derived from tumor-associated antigens (TAAs) or neoantigens, and costimulation via CD4+ T helper cells which then elicits an effector and memory immune response that targets and kills cancer cells. Despite the tumoricidal capacity of CTLs, cancer cells can escape immune surveillance and killing due to their immunosuppressive tumor microenvironment (TME). Therefore, to harness the CTL immune response and combat the effect of the TME, peptide-based T cell vaccines targeting specific onco-antigens, conjugated with adjuvants are a subject of ongoing research for cancer immunotherapy; particularly, multi-peptide vaccines, containing both CTL and CD4+ T helper cell epitopes along with an immunostimulant. Historically, peptide-based T cell vaccines have been investigated as a potential strategy for cancer immunotherapy. Despite initial enthusiasm, these peptide vaccines have not demonstrated success in clinical outcomes. However, recent advancements in our understanding of cancer immunology and the design of peptide vaccines targeting specific tumor antigens have paved the way for novel strategies in peptide-based immunotherapy. These advancements have reignited optimism surrounding the potential of peptide-based vaccines as a viable cancer therapeutic. This review explores the new strategies and discusses the exciting possibilities they offer. Specifically, this review develops an understanding of vaccine design and clinical outcomes, by discussing mechanisms of CTL effector and memory responses, and how peptide-based vaccines can induce and enhance these responses. It addresses the challenge of Major Histocompatibility Complex (MHC) restriction, which limits the effectiveness of traditional peptide vaccines in individuals with diverse MHC types. It also delves into the immunosuppressive tumor microenvironment and overcoming its inhibitory effects using peptide-based vaccines for efficient cancer cell elimination. The review aims to provide an understanding of the complexities faced by each field in vaccine design, enhancing dialogue and understanding among researchers by bringing together the chemistry of vaccine synthesis, cancer immunology, and clinical studies to support the development of a peptide-based vaccine.
Collapse
Affiliation(s)
- Bansari A Shah
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital, and The Graeme Clark Institute, The University of Melbourne, Carlton, VIC, Australia
| | - James A Holden
- Centre for Oral Health Research, Melbourne Dental School, Royal Dental Hospital, The University of Melbourn, Carlton, VIC, Australia
| | - Jason C Lenzo
- Western Australian Health Translation Network, Harry Perkins Institute of Medical Research, Level 6, Nedlands, Perth, WA, Australia
| | - Sara Hadjigol
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital, and The Graeme Clark Institute, The University of Melbourne, Carlton, VIC, Australia.
| | - Neil M O'Brien-Simpson
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital, and The Graeme Clark Institute, The University of Melbourne, Carlton, VIC, Australia.
| |
Collapse
|
3
|
Xu X, Guo S, Gu H, Cha Z, Shi X, Yin X, Wang H, Gao S, Li B, Zhu L, Jing W, Zheng K, Shao Z, Cheng P, Zheng C, Shih YP, Li Y, Qian B, Gao D, Tran E, Jin G. Identification and validation of a T cell receptor targeting KRAS G12V in HLA-A*11:01 pancreatic cancer patients. JCI Insight 2025; 10:e181873. [PMID: 39846249 PMCID: PMC11790028 DOI: 10.1172/jci.insight.181873] [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] [Indexed: 01/24/2025] Open
Abstract
T cells targeting a KRAS mutation can induce durable tumor regression in some patients with metastatic epithelial cancer. It is unknown whether T cells targeting mutant KRAS that are capable of killing tumor cells can be identified from peripheral blood of patients with pancreatic cancer. We developed an in vitro stimulation approach and identified HLA-A*11:01-restricted KRAS G12V-reactive CD8+ T cells and HLA-DRB1*15:01-restricted KRAS G12V-reactive CD4+ T cells from peripheral blood of 2 out of 6 HLA-A*11:01-positive patients with pancreatic cancer whose tumors expressed KRAS G12V. The HLA-A*11:01-restricted KRAS G12V-reactive T cell receptor (TCR) was isolated and validated to specifically recognize the KRAS G12V8-16 neoepitope. While T cells engineered to express this TCR specifically recognized all 5 tested human HLA-A*11:01+ and KRAS G12V+ pancreatic cancer organoids, the recognition was often modest, and tumor cell killing was observed in only 2 out of 5 organoids. IFN-γ priming of the organoids enhanced the recognition and killing by the TCR-engineered T cells. The TCR-engineered T cells could significantly slow the growth of an established organoid-derived xenograft in immunodeficient mice. Our data suggest that this TCR has potential for use in TCR-gene therapy, but additional strategies that enhance tumor recognition by the TCR-engineered T cells likely will be required to increase clinical activity.
Collapse
Affiliation(s)
- Xiongfei Xu
- Department of Hepatobiliary Pancreatic Surgery
- Shanghai Institute of Pancreatic Diseases, and
| | - Shiwei Guo
- Department of Hepatobiliary Pancreatic Surgery
- Shanghai Institute of Pancreatic Diseases, and
| | - Haihui Gu
- Department of Transfusion Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhanshan Cha
- Department of Transfusion Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery
| | - Xiaoyi Yin
- Department of Hepatobiliary Pancreatic Surgery
| | - Huan Wang
- Department of Hepatobiliary Pancreatic Surgery
| | - Suizhi Gao
- Department of Hepatobiliary Pancreatic Surgery
| | - Bo Li
- Department of Hepatobiliary Pancreatic Surgery
| | - Lingyu Zhu
- Department of Hepatobiliary Pancreatic Surgery
| | - Wei Jing
- Department of Hepatobiliary Pancreatic Surgery
| | | | - Zhuo Shao
- Department of Hepatobiliary Pancreatic Surgery
| | - Peng Cheng
- Department of Hepatobiliary Pancreatic Surgery
| | - Chunhong Zheng
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
- International Cancer Institute, Peking University, Beijing, China
| | - Yi-Ping Shih
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Yunguang Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Baohua Qian
- Department of Transfusion Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dong Gao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Eric Tran
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery
- Shanghai Institute of Pancreatic Diseases, and
| |
Collapse
|
4
|
Han X, Han X, Hao Y, Wang B, Li L, Chen S, Zou L, Huang J, Chen T, Wang W, Liu S, Jin A, Shen M. Identification of novel KRAS G12D neoantigen specific TCRs and a strategy to eliminate off-target recognition. J Transl Med 2025; 23:78. [PMID: 39819441 PMCID: PMC11740425 DOI: 10.1186/s12967-025-06094-1] [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: 10/08/2024] [Accepted: 01/08/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND T cell receptor (TCR)-engineered T cells targeting neoantigens originated from mutations in KRAS gene have demonstrated promising outcomes in clinical trials against solid tumors. However, the challenge lies in developing tumor-specific TCRs that avoid cross-reactivity with self-antigens to minimize the possibility of severe clinical toxicities. Current research efforts have been put towards strategies to eliminate TCR off-target recognition. METHODS Naive T cell repertoire was used for screening KRASG12D-reactive TCRs. Specific TCRs were subsequently identified and their functionality was assessed using TCR Jurkat cells and TCR T cells. Peptide specificity was evaluated using the X-scan assay. To enhance TCR specificity for KRASG12D and reduce their reactivity to self-peptide SMC1A29-38, mammalian TCR display libraries were employed for the design of modification in the complementarity-determining region (CDR). RESULTS HLA-A*11:01-restricted TCRs targeting the KRASG12D epitope were isolated, and TCR1 was characterized with superior functional avidity and specificity. Alongside a robust recognition of endogenous KRASG12D epitope, this TCR displayed cross-reactivity with the SMC1A29-38 epitope. With an approach utilizing structural-guided mutations in the CDR-1A region of TCR1, we obtained an engineered TCR variant (TCR1a7). Functional characterization of TCR1a7 showed that this TCR not only exhibited enhanced specificity towards KRASG12D, but also demonstrated successful elimination of the off-target recognition of SMC1A29-38. CONCLUSIONS TCRs targeting the KRASG12D peptide could be isolated from naive T cell repertoires. Integrating the TCR-peptide-HLA complex structure with a mammalian TCR library system could serve as a functional strategy to reduce potential TCR cross-reactivity with self-antigens, such as SMC1A29-38. Our findings evidenced an operable method to enhance TCR peptide specificity, while maintaining advanced functional avidity and potent anti-tumor activity.
Collapse
Affiliation(s)
- Xiaojian Han
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Xiaxia Han
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Yanan Hao
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Bozhi Wang
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Luo Li
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Siyin Chen
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Lin Zou
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Jingjing Huang
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Tong Chen
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Wang Wang
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Shengchun Liu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Aishun Jin
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China.
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China.
| | - Meiying Shen
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
5
|
Tran E. More T cell receptors to the RAScue in cancer? J Clin Invest 2024; 134:e184782. [PMID: 39484723 PMCID: PMC11527437 DOI: 10.1172/jci184782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
Abstract
Treatment with T cells genetically engineered to express tumor-reactive T cell receptors (TCRs), known as TCR-gene therapy (TCR-T), is a promising immunotherapeutic approach for patients with cancer. The identification of optimal TCRs to use and tumor antigens to target are key considerations for TCR-T. In this issue of the JCI, Bear and colleagues report on their use of in vitro assays to characterize four HLA-A*03:01- or HLA-A*11:01-restricted TCRs targeting the oncogenic KRAS G12V mutation. The TCRs were derived from healthy donors or patients with pancreatic cancer who had received a vaccine against mutant KRAS. The most promising TCRs warrant testing in patients with KRAS G12V-positive cancers.
Collapse
|
6
|
Lee JW, Chen EY, Hu T, Perret R, Chaffee ME, Martinov T, Mureli S, McCurdy CL, Jones LA, Gafken PR, Chanana P, Su Y, Chapuis AG, Bradley P, Schmitt TM, Greenberg PD. Overcoming immune evasion from post-translational modification of a mutant KRAS epitope to achieve TCR-T cell-mediated antitumor activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.612965. [PMID: 39345486 PMCID: PMC11429761 DOI: 10.1101/2024.09.18.612965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
T cell receptor (TCR)-T cell immunotherapy, in which T cells are engineered to express a TCR targeting a tumor epitope, is a form of adoptive cell therapy (ACT) that has exhibited promise against various tumor types. Mutants of oncoprotein KRAS, particularly at glycine-12 (G12), are frequent drivers of tumorigenicity, making them attractive targets for TCR-T cell therapy. However, class I-restricted TCRs specifically targeting G12-mutant KRAS epitopes in the context of tumors expressing HLA-A2, the most common human HLA-A allele, have remained elusive despite evidence an epitope encompassing such mutations can bind HLA-A2 and induce T cell responses. We report post-translational modifications (PTMs) on this epitope may allow tumor cells to evade immunologic pressure from TCR-T cells. A lysine side chain-methylated KRAS G12V peptide, rather than the unmodified epitope, may be presented in HLA-A2 by tumor cells and impact TCR recognition. Using a novel computationally guided approach, we developed by mutagenesis TCRs that recognize this methylated peptide, enhancing tumor recognition and destruction. Additionally, we identified TCRs with similar functional activity in normal repertoires from primary T cells by stimulation with modified peptide, clonal expansion, and selection. Mechanistically, a gene knockout screen to identify mechanism(s) by which tumor cells methylate/demethylate this epitope unveiled SPT6 as a demethylating protein that could be targeted to improve effectiveness of these new TCRs. Our findings highlight the role of PTMs in immune evasion and suggest identifying and targeting such modifications should make effective ACTs available for a substantially greater range of tumors than the current therapeutic landscape. One-sentence summary Tumor cell methylation of KRAS G12V epitope in HLA-A2 permits immune evasion, and new TCRs were generated to overcome this with engineered cell therapy.
Collapse
|
7
|
Bear AS, Nadler RB, O'Hara MH, Stanton KL, Xu C, Saporito RJ, Rech AJ, Baroja ML, Blanchard T, Elliott MH, Ford MJ, Jones R, Patel S, Brennan A, O'Neil Z, Powell DJ, Vonderheide RH, Linette GP, Carreno BM. Natural TCRs targeting KRASG12V display fine specificity and sensitivity to human solid tumors. J Clin Invest 2024; 134:e175790. [PMID: 39287991 PMCID: PMC11529987 DOI: 10.1172/jci175790] [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: 09/14/2023] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUNDNeoantigens derived from KRASMUT have been described, but the fine antigen specificity of T cell responses directed against these epitopes is poorly understood. Here, we explore KRASMUT immunogenicity and the properties of 4 T cell receptors (TCRs) specific for KRASG12V restricted to the HLA-A3 superfamily of class I alleles.METHODSA phase 1 clinical vaccine trial targeting KRASMUT was conducted. TCRs targeting KRASG12V restricted to HLA-A*03:01 or HLA-A*11:01 were isolated from vaccinated patients or healthy individuals. A comprehensive analysis of TCR antigen specificity, affinity, crossreactivity, and CD8 coreceptor dependence was performed. TCR lytic activity was evaluated, and target antigen density was determined by quantitative immunopeptidomics.RESULTSVaccination against KRASMUT resulted in the priming of CD8+ and CD4+ T cell responses. KRASG12V -specific natural (not affinity enhanced) TCRs exhibited exquisite specificity to mutated protein with no discernible reactivity against KRASWT. TCR-recognition motifs were determined and used to identify and exclude crossreactivity to noncognate peptides derived from the human proteome. Both HLA-A*03:01 and HLA-A*11:01-restricted TCR-redirected CD8+ T cells exhibited potent lytic activity against KRASG12V cancers, while only HLA-A*11:01-restricted TCR-T CD4+ T cells exhibited antitumor effector functions consistent with partial coreceptor dependence. All KRASG12V-specific TCRs displayed high sensitivity for antigen as demonstrated by their ability to eliminate tumor cell lines expressing low levels of peptide/HLA (4.4 to 242) complexes per cell.CONCLUSIONThis study identifies KRASG12V-specific TCRs with high therapeutic potential for the development of TCR-T cell therapies.TRIAL REGISTRATIONClinicalTrials.gov NCT03592888.FUNDINGAACR SU2C/Lustgarten Foundation, Parker Institute for Cancer Immunotherapy, and NIH.
Collapse
Affiliation(s)
- Adham S Bear
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine
| | | | - Mark H O'Hara
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine
- Abramson Cancer Center, and
| | - Kelsey L Stanton
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Chong Xu
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert J Saporito
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrew J Rech
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Miren L Baroja
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tatiana Blanchard
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maxwell H Elliott
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Shivang Patel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrea Brennan
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zachary O'Neil
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel J Powell
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine and
| | - Robert H Vonderheide
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine
- Abramson Cancer Center, and
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gerald P Linette
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine
- Abramson Cancer Center, and
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Beatriz M Carreno
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine and
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Wang X, Wang W, Zou S, Xu Z, Cao D, Zhang S, Wei M, Zhan Q, Wen C, Li F, Chen H, Fu D, Jiang L, Zhao M, Shen B. Combination therapy of KRAS G12V mRNA vaccine and pembrolizumab: clinical benefit in patients with advanced solid tumors. Cell Res 2024; 34:661-664. [PMID: 38914844 PMCID: PMC11369195 DOI: 10.1038/s41422-024-00990-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 06/03/2024] [Indexed: 06/26/2024] Open
Affiliation(s)
- Xinjing Wang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Wang
- Shanghai Xinpu BioTechnology Company Limited, Shanghai, China
| | - Siyi Zou
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiwei Xu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dan Cao
- Hongene Biotech Corporation, Shanghai, China
| | - Shuai Zhang
- Shanghai Xinpu BioTechnology Company Limited, Shanghai, China
| | - Minzhi Wei
- Hongene Biotech Corporation, Shanghai, China
| | - Qian Zhan
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chenlei Wen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fanlu Li
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Chen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Da Fu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Lingxi Jiang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Ming Zhao
- Shanghai Xinpu BioTechnology Company Limited, Shanghai, China.
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
9
|
Meng W, Takeuchi Y, Ward JP, Sultan H, Arthur CD, Mardis ER, Artyomov MN, Lichti CF, Schreiber RD. Improvement of Tumor Neoantigen Detection by High-Field Asymmetric Waveform Ion Mobility Mass Spectrometry. Cancer Immunol Res 2024; 12:988-1006. [PMID: 38768391 PMCID: PMC11456315 DOI: 10.1158/2326-6066.cir-23-0900] [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: 10/26/2023] [Revised: 03/05/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
Cancer neoantigens have been shown to elicit cancer-specific T-cell responses and have garnered much attention for their roles in both spontaneous and therapeutically induced antitumor responses. Mass spectrometry (MS) profiling of tumor immunopeptidomes has been used, in part, to identify MHC-bound mutant neoantigen ligands. However, under standard conditions, MS-based detection of such rare but clinically relevant neoantigens is relatively insensitive, requiring 300 million cells or more. Here, to quantitatively define the minimum detectable amounts of therapeutically relevant MHC-I and MHC-II neoantigen peptides, we analyzed different dilutions of immunopeptidomes isolated from the well-characterized T3 mouse methylcholanthrene (MCA)-induced cell line by MS. Using either data-dependent acquisition or parallel reaction monitoring (PRM), we established the minimum amount of material required to detect the major T3 neoantigens in the presence or absence of high field asymmetric waveform ion mobility spectrometry (FAIMS). This analysis yielded a 14-fold enhancement of sensitivity in detecting the major T3 MHC-I neoantigen (mLama4) with FAIMS-PRM compared with PRM without FAIMS, allowing ex vivo detection of this neoantigen from an individual 100 mg T3 tumor. These findings were then extended to two other independent MCA-sarcoma lines (1956 and F244). This study demonstrates that FAIMS substantially increases the sensitivity of MS-based characterization of validated neoantigens from tumors.
Collapse
Affiliation(s)
- Wei Meng
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Yoshiko Takeuchi
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Jeffrey P. Ward
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Hussein Sultan
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Cora D. Arthur
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Elaine R. Mardis
- The Steve and Cindy Rasmussen Institute for Genomic Medicine at Nationwide Children’s Hospital, Columbus, OH 43215, U.S.A
| | - Maxim N. Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Cheryl F. Lichti
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Robert D. Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| |
Collapse
|
10
|
Gurung HR, Heidersbach AJ, Darwish M, Chan PPF, Li J, Beresini M, Zill OA, Wallace A, Tong AJ, Hascall D, Torres E, Chang A, Lou K'HW, Abdolazimi Y, Hammer C, Xavier-Magalhães A, Marcu A, Vaidya S, Le DD, Akhmetzyanova I, Oh SA, Moore AJ, Uche UN, Laur MB, Notturno RJ, Ebert PJR, Blanchette C, Haley B, Rose CM. Systematic discovery of neoepitope-HLA pairs for neoantigens shared among patients and tumor types. Nat Biotechnol 2024; 42:1107-1117. [PMID: 37857725 PMCID: PMC11251992 DOI: 10.1038/s41587-023-01945-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 08/14/2023] [Indexed: 10/21/2023]
Abstract
The broad application of precision cancer immunotherapies is limited by the number of validated neoepitopes that are common among patients or tumor types. To expand the known repertoire of shared neoantigen-human leukocyte antigen (HLA) complexes, we developed a high-throughput platform that coupled an in vitro peptide-HLA binding assay with engineered cellular models expressing individual HLA alleles in combination with a concatenated transgene harboring 47 common cancer neoantigens. From more than 24,000 possible neoepitope-HLA combinations, biochemical and computational assessment yielded 844 unique candidates, of which 86 were verified after immunoprecipitation mass spectrometry analyses of engineered, monoallelic cell lines. To evaluate the potential for immunogenicity, we identified T cell receptors that recognized select neoepitope-HLA pairs and elicited a response after introduction into human T cells. These cellular systems and our data on therapeutically relevant neoepitopes in their HLA contexts will aid researchers studying antigen processing as well as neoepitope targeting therapies.
Collapse
Affiliation(s)
| | | | | | | | - Jenny Li
- Genentech, South San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Ana Marcu
- Genentech, South San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Shen M, Chen S, Han X, Hao Y, Wang J, Li L, Chen T, Wang B, Zou L, Zhang T, Zhang W, Han X, Wang W, Yu H, Li K, Liu S, Jin A. Identification of an HLA-A*11:01-restricted neoepitope of mutant PIK3CA and its specific T cell receptors for cancer immunotherapy targeting hotspot driver mutations. Cancer Immunol Immunother 2024; 73:150. [PMID: 38832948 PMCID: PMC11150344 DOI: 10.1007/s00262-024-03729-y] [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: 04/10/2024] [Accepted: 05/10/2024] [Indexed: 06/06/2024]
Abstract
Hotspot driver mutations presented by human leukocyte antigens might be recognized by anti-tumor T cells. Based on their advantages of tumor-specificity and immunogenicity, neoantigens derived from hotspot mutations, such as PIK3CAH1047L, may serve as emerging targets for cancer immunotherapies. NetMHCpan V4.1 was utilized for predicting neoepitopes of PIK3CA hotspot mutation. Using in vitro stimulation, antigen-specific T cells targeting the HLA-A*11:01-restricted PIK3CA mutation were isolated from healthy donor-derived peripheral blood mononuclear cells. T cell receptors (TCRs) were cloned using single-cell PCR and sequencing. Their functionality was assessed through T cell activation markers, cytokine production and cytotoxic response to cancer cell lines pulsed with peptides or transduced genes of mutant PIK3CA. Immunogenic mutant antigens from PIK3CA and their corresponding CD8+ T cells were identified. These PIK3CA mutation-specific CD8+ T cells were subsequently enriched, and their TCRs were isolated. The TCR clones exhibited mutation-specific and HLA-restricted reactivity, demonstrating varying degrees of functional avidity. Identified TCR genes were transferred into CD8+ Jurkat cells and primary T cells deficient of endogenous TCRs. TCR-expressing cells demonstrated specific recognition and reactivity against the PIK3CAH1047L peptide presented by HLA-A*11:01-expressing K562 cells. Furthermore, mutation-specific TCR-T cells demonstrated an elevation in cytokine production and profound cytotoxic effects against HLA-A*11:01+ malignant cell lines harboring PIK3CAH1047L. Our data demonstrate the immunogenicity of an HLA-A*11:01-restricted PIK3CA hotspot mutation and its targeting therapeutic potential, together with promising candidates of TCR-T cell therapy.
Collapse
Affiliation(s)
- Meiying Shen
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Siyin Chen
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Xiaojian Han
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Yanan Hao
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Junfan Wang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Luo Li
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Tong Chen
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Bozhi Wang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Lin Zou
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Tong Zhang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Wanli Zhang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Xiaxia Han
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Wang Wang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China
| | - Haochen Yu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kang Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengchun Liu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Aishun Jin
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
12
|
Linette GP, Bear AS, Carreno BM. Facts and Hopes in Immunotherapy Strategies Targeting Antigens Derived from KRAS Mutations. Clin Cancer Res 2024; 30:2017-2024. [PMID: 38266167 PMCID: PMC11094419 DOI: 10.1158/1078-0432.ccr-23-1212] [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: 11/20/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024]
Abstract
In this commentary, we advance the notion that mutant KRAS (mKRAS) is an ideal tumor neoantigen that is amenable for targeting by the adaptive immune system. Recent progress highlights key advances on various fronts that validate mKRAS as a molecular target and support further pursuit as an immunological target. Because mKRAS is an intracellular membrane localized protein and not normally expressed on the cell surface, we surmise that proteasome degradation will generate short peptides that bind to HLA class I (HLA-I) molecules in the endoplasmic reticulum for transport through the Golgi for display on the cell surface. T-cell receptors (TCR)αβ and antibodies have been isolated that specifically recognize mKRAS encoded epitope(s) or haptenated-mKRAS peptides in the context of HLA-I on tumor cells. Case reports using adoptive T-cell therapy provide proof of principle that KRAS G12D can be successfully targeted by the immune system in patients with cancer. Among the challenges facing investigators is the requirement of precision medicine to identify and match patients to available mKRAS peptide/HLA therapeutics and to increase the population coverage by targeting additional mKRAS epitopes. Ultimately, we envision mKRAS-directed immunotherapy as an effective treatment option for selected patients that will complement and perhaps synergize with small-molecule mKRAS inhibitors and targeted mKRAS degraders.
Collapse
Affiliation(s)
- Gerald P. Linette
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Adham S. Bear
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Beatriz M. Carreno
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
13
|
Shafer P, Leung WK, Woods M, Choi JM, Rodriguez-Plata CM, Maknojia A, Mosquera A, Somes LK, Joubert J, Manliguez A, Ranjan R, Burt B, Lee HS, Zhang B, Fuqua S, Rooney C, Leen AM, Hoyos V. Incongruity between T cell receptor recognition of breast cancer hotspot mutations ESR1 Y537S and D538G following exogenous peptide loading versus endogenous antigen processing. Cytotherapy 2024; 26:266-275. [PMID: 38231165 PMCID: PMC10922969 DOI: 10.1016/j.jcyt.2023.12.002] [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: 08/14/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 01/18/2024]
Abstract
T cell receptor engineered T cell (TCR T) therapies have shown recent efficacy against certain types of solid metastatic cancers. However, to extend TCR T therapies to treat more patients across additional cancer types, new TCRs recognizing cancer-specific antigen targets are needed. Driver mutations in AKT1, ESR1, PIK3CA, and TP53 are common in patients with metastatic breast cancer (MBC) and if immunogenic could serve as ideal tumor-specific targets for TCR T therapy to treat this disease. Through IFN-γ ELISpot screening of in vitro expanded neopeptide-stimulated T cell lines from healthy donors and MBC patients, we identified reactivity towards 11 of 13 of the mutations. To identify neopeptide-specific TCRs, we then performed single-cell RNA sequencing of one of the T cell lines following neopeptide stimulation. Here, we identified an ESR1 Y537S specific T cell clone, clonotype 16, and an ESR1 Y537S/D538G dual-specific T cell clone, clonotype 21, which were HLA-B*40:02 and HLA-C*01:02 restricted, respectively. TCR Ts expressing these TCRs recognized and killed target cells pulsed with ESR1 neopeptides with minimal activity against ESR1 WT peptide. However, these TCRs failed to recognize target cells expressing endogenous mutant ESR1. To investigate the basis of this lack of recognition we performed immunopeptidomics analysis of a mutant-overexpressing lymphoblastoid cell line and found that the ESR1 Y537S neopeptide was not endogenously processed, despite binding to HLA-B*40:02 when exogenously pulsed onto the target cell. These results indicate that stimulation of T cells that likely derive from the naïve repertoire with pulsed minimal peptides may lead to the expansion of clones that recognize non-processed peptides, and highlights the importance of using methods that selectively expand T cells with specificity for antigens that are efficiently processed and presented.
Collapse
Affiliation(s)
- Paul Shafer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA
| | - Wingchi K Leung
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA
| | - Mae Woods
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA
| | - Jong Min Choi
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Carlos M Rodriguez-Plata
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA
| | - Arushana Maknojia
- Division of Infectious Disease, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Andres Mosquera
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA
| | - Lauren K Somes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA
| | - Jarrett Joubert
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA
| | - Anthony Manliguez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA
| | - Rashi Ranjan
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA
| | - Bryan Burt
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Hyun-Sung Lee
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Bing Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Suzanne Fuqua
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Cliona Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA
| | - Ann M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA
| | - Valentina Hoyos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA.
| |
Collapse
|
14
|
Katsikis PD, Ishii KJ, Schliehe C. Challenges in developing personalized neoantigen cancer vaccines. Nat Rev Immunol 2024; 24:213-227. [PMID: 37783860 PMCID: PMC12001822 DOI: 10.1038/s41577-023-00937-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2023] [Indexed: 10/04/2023]
Abstract
The recent success of cancer immunotherapies has highlighted the benefit of harnessing the immune system for cancer treatment. Vaccines have a long history of promoting immunity to pathogens and, consequently, vaccines targeting cancer neoantigens have been championed as a tool to direct and amplify immune responses against tumours while sparing healthy tissue. In recent years, extensive preclinical research and more than one hundred clinical trials have tested different strategies of neoantigen discovery and vaccine formulations. However, despite the enthusiasm for neoantigen vaccines, proof of unequivocal efficacy has remained beyond reach for the majority of clinical trials. In this Review, we focus on the key obstacles pertaining to vaccine design and tumour environment that remain to be overcome in order to unleash the true potential of neoantigen vaccines in cancer therapy.
Collapse
Affiliation(s)
- Peter D Katsikis
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands.
| | - Ken J Ishii
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan
- International Vaccine Design Center (vDesC), The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan
| | - Christopher Schliehe
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
15
|
Meng W, Schreiber RD, Lichti CF. Recent advances in immunopeptidomic-based tumor neoantigen discovery. Adv Immunol 2023; 160:1-36. [PMID: 38042584 DOI: 10.1016/bs.ai.2023.10.001] [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] [Indexed: 12/04/2023]
Abstract
The role of aberrantly expressed proteins in tumors in driving immune-mediated control of cancer has been well documented for more than five decades. Today, we know that both aberrantly expressed normal proteins as well as mutant proteins (neoantigens) can function as tumor antigens in both humans and mice. Next-generation sequencing (NGS) and high-resolution mass spectrometry (MS) technologies have made significant advances since the early 2010s, enabling detection of rare but clinically relevant neoantigens recognized by T cells. MS profiling of tumor-specific immunopeptidomes remains the most direct method to identify mutant peptides bound to cellular MHC. However, the need for use of large numbers of cells or significant amounts of tumor tissue to achieve neoantigen detection has historically limited the application of MS. Newer, more sensitive MS technologies have recently demonstrated the capacities to detect neoantigens from fewer cells. Here, we highlight recent advancements in immunopeptidomics-based characterization of tumor-specific neoantigens. Various tumor antigen categories and neoantigen identification approaches are also discussed. Furthermore, we summarize recent reports that achieved successful tumor neoantigen detection by MS using a variety of starting materials, MS acquisition modes, and novel ion mobility devices.
Collapse
Affiliation(s)
- Wei Meng
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, United States
| | - Robert D Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, United States.
| | - Cheryl F Lichti
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, United States.
| |
Collapse
|
16
|
Lu D, Chen Y, Jiang M, Wang J, Li Y, Ma K, Sun W, Zheng X, Qi J, Jin W, Chen Y, Chai Y, Zhang CWH, Liang H, Tan S, Gao GF. KRAS G12V neoantigen specific T cell receptor for adoptive T cell therapy against tumors. Nat Commun 2023; 14:6389. [PMID: 37828002 PMCID: PMC10570350 DOI: 10.1038/s41467-023-42010-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
KRAS mutations are broadly recognized as promising targets for tumor therapy. T cell receptors (TCRs) can specifically recognize KRAS mutant neoantigens presented by human lymphocyte antigen (HLA) and mediate T cell responses to eliminate tumor cells. In the present study, we identify two TCRs specific for the 9-mer KRAS-G12V mutant neoantigen in the context of HLA-A*11:01. The TCR-T cells are constructed and display cytokine secretion and cytotoxicity upon co-culturing with varied tumor cells expressing the KRAS-G12V mutation. Moreover, 1-2C TCR-T cells show anti-tumor activity in preclinical models in female mice. The 9-mer KRAS-G12V mutant peptide exhibits a distinct conformation from the 9-mer wildtype peptide and its 10-mer counterparts. Specific recognition of the G12V mutant by TCR depends both on distinct conformation from wildtype peptide and on direct interaction with residues from TCRs. Our study reveals the mechanisms of presentation and TCR recognition of KRAS-G12V mutant peptide and describes TCRs with therapeutic potency for tumor immunotherapy.
Collapse
Affiliation(s)
- Dan Lu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Department of Immunology, Beijing Children's Hospital, Capital Medical University, National Centre for Children's Health, Beijing, China
| | - Yuan Chen
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Min Jiang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Jie Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yiting Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Keke Ma
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Wenqiao Sun
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xing Zheng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Wenjing Jin
- YKimmu (Beijing) Biotechnology Co., Ltd, Beijing, China
| | - Yu Chen
- YKimmu (Beijing) Biotechnology Co., Ltd, Beijing, China
| | - Yan Chai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | | | - Hao Liang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Shuguang Tan
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
- Shenzhen Children's Hospital, Shenzhen, Guangdong, China.
- Beijing Life Science Academy, Beijing, China.
| | - George F Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
- Beijing Life Science Academy, Beijing, China.
| |
Collapse
|
17
|
Schendel DJ. Evolution by innovation as a driving force to improve TCR-T therapies. Front Oncol 2023; 13:1216829. [PMID: 37810959 PMCID: PMC10552759 DOI: 10.3389/fonc.2023.1216829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/16/2023] [Indexed: 10/10/2023] Open
Abstract
Adoptive cell therapies continually evolve through science-based innovation. Specialized innovations for TCR-T therapies are described here that are embedded in an End-to-End Platform for TCR-T Therapy Development which aims to provide solutions for key unmet patient needs by addressing challenges of TCR-T therapy, including selection of target antigens and suitable T cell receptors, generation of TCR-T therapies that provide long term, durable efficacy and safety and development of efficient and scalable production of patient-specific (personalized) TCR-T therapy for solid tumors. Multiple, combinable, innovative technologies are used in a systematic and sequential manner in the development of TCR-T therapies. One group of technologies encompasses product enhancements that enable TCR-T therapies to be safer, more specific and more effective. The second group of technologies addresses development optimization that supports discovery and development processes for TCR-T therapies to be performed more quickly, with higher quality and greater efficiency. Each module incorporates innovations layered onto basic technologies common to the field of immunology. An active approach of "evolution by innovation" supports the overall goal to develop best-in-class TCR-T therapies for treatment of patients with solid cancer.
Collapse
Affiliation(s)
- Dolores J. Schendel
- Medigene Immunotherapies GmbH, Planegg, Germany
- Medigene AG, Planegg, Germany
| |
Collapse
|
18
|
Carabaño M, Liu T, Liu A, Lee JC, Cheng L, Wang LJ, Huang CK, Lu S. Association of HLA-A, B, and C alleles and cancer susceptibility in 179 solid malignancies. Am J Transl Res 2023; 15:5642-5652. [PMID: 37854217 PMCID: PMC10579009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/04/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND The major histocompatibility complex (MHC) genes are known to be capable of influencing the susceptibility of many cancers. All mammalian cells, including cancer cells, express MHC class I molecules consisting of human leukocyte antigens (HLA) A, B, and C. The tumor susceptibility of HLA-A, B, and C alleles has not been studied extensively in solid tumors. METHODS HLA-A, B, and C genotypes of 179 solid tumors were collected from Caris Comprehensive Tumor Profiling reports, including 45 GU, 44 GI, 28 pancreaticobiliary, 21 thoracic, 15 breast, 13 Gyn, among others. The tumors were mainly from Caucasians (82%). The HLA allele frequencies in the tumors were compared to those of respective ethnic populations in the US National Marrow Donor Program (NMDP) database. Fisher's exact tests were performed, adjusted P values were calculated using Benjamini-Hochberg's method for false discovery rate (FDR), and Prevalence ratios (PRs) were calculated to quantify associations. RESULTS Twenty-one alleles were not listed in the NMDP. Among them, A*11:303 alone was present in 11 carcinomas, and B*08:222 was seen in 4 tumors. Among the alleles listed in the NMDP, C*08:02, B*14:02, A*03:02, and B*44:06 were significantly associated with tumors in Caucasian Americans (PR: 2.50-170), while B*44:02 appeared protective (PR: 0.36). Alleles with less significant associations were listed. CONCLUSIONS From the HLA-A, B, and C data of the 179 tumors, we identified several susceptible alleles and one protective allele. Of interest, 21 alleles were not listed in the NMDP. The limited cases prevented our analysis from identifying cancer-susceptible alleles in other races.
Collapse
Affiliation(s)
- Miguel Carabaño
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown UniversityProvidence, RI 02903, USA
| | - Tao Liu
- Department of Biostatistics, Brown UniversityProvidence, RI 02912, USA
| | - Abraham Liu
- Department of Biostatistics, Brown UniversityProvidence, RI 02912, USA
| | - Jim ChunHao Lee
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown UniversityProvidence, RI 02903, USA
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown UniversityProvidence, RI 02903, USA
| | - Li-Juan Wang
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown UniversityProvidence, RI 02903, USA
| | - Chiung-Kuei Huang
- Department of Pathology and Laboratory Medicine, Tulane University School of MedicineNew Orleans, LA 70112, USA
| | - Shaolei Lu
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown UniversityProvidence, RI 02903, USA
| |
Collapse
|
19
|
Ade CM, Sporn MJ, Das S, Yu Z, Hanada KI, Qi YA, Maity T, Zhang X, Guha U, Andresson T, Yang JC. Identification of neoepitope reactive T-cell receptors guided by HLA-A*03:01 and HLA-A*11:01 immunopeptidomics. J Immunother Cancer 2023; 11:e007097. [PMID: 37758652 PMCID: PMC10537849 DOI: 10.1136/jitc-2023-007097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Tumor-specific mutated proteins can create immunogenic non-self, mutation-containing 'neoepitopes' that are attractive targets for adoptive T-cell therapies. To avoid the complexity of defining patient-specific, private neoepitopes, there has been major interest in targeting common shared mutations in driver genes using off-the-shelf T-cell receptors (TCRs) engineered into autologous lymphocytes. However, identifying the precise naturally processed neoepitopes to pursue is a complex and challenging process. One method to definitively demonstrate whether an epitope is presented at the cell surface is to elute peptides bound to a specific major histocompatibility complex (MHC) allele and analyze them by mass spectrometry (MS). These MS data can then be prospectively applied to isolate TCRs specific to the neoepitope. METHODS We created mono-allelic cell lines expressing one class I HLA allele and one common mutated oncogene in order to eliminate HLA deconvolution requirements and increase the signal of recovered peptides. MHC-bound peptides on the surface of these cell lines were immunoprecipitated, purified, and analyzed using liquid chromatography-tandem mass spectrometry, producing a list of mutation-containing minimal epitopes. To validate the immunogenicity of these neoepitopes, HLA-transgenic mice were vaccinated using the minimal peptides identified by MS in order to generate neoepitope-reactive TCRs. Specificity of these candidate TCRs was confirmed by peptide titration and recognition of transduced targets. RESULTS We identified precise neoepitopes derived from mutated isoforms of KRAS, EGFR, BRAF, and PIK3CA presented by HLA-A*03:01 and/or HLA-A*11:01 across multiple biological replicates. From our MS data, we were able to successfully isolate murine TCRs that specifically recognize four HLA-A*11:01 restricted neoepitopes (KRAS G13D, PIK3CA E545K, EGFR L858R and BRAF V600E) and three HLA-A*03:01 restricted neoepitopes (KRAS G12V, EGFR L858R and BRAF V600E). CONCLUSIONS Our data show that an MS approach can be used to demonstrate which shared oncogene-derived neoepitopes are processed and presented by common HLA alleles, and those MS data can rapidly be used to develop TCRs against these common tumor-specific antigens. Although further characterization of these neoepitope-specific murine TCRs is required, ultimately, they have the potential to be used clinically for adoptive cell therapy.
Collapse
Affiliation(s)
- Catherine M Ade
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Matthew J Sporn
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Sudipto Das
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Zhiya Yu
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Ken-Ichi Hanada
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Yue A Qi
- Thoracic and GI Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Tapan Maity
- Thoracic and GI Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
- Laboratory of Cell Biology, National Cancer Institute, Bethesda, MD, USA
| | - Xu Zhang
- Thoracic and GI Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - Udayan Guha
- Thoracic and GI Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
- NextCure Inc, Beltsville, MD, USA
| | - Thorkell Andresson
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - James C Yang
- Surgery Branch, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
20
|
Wright KM, DiNapoli SR, Miller MS, Aitana Azurmendi P, Zhao X, Yu Z, Chakrabarti M, Shi W, Douglass J, Hwang MS, Hsiue EHC, Mog BJ, Pearlman AH, Paul S, Konig MF, Pardoll DM, Bettegowda C, Papadopoulos N, Kinzler KW, Vogelstein B, Zhou S, Gabelli SB. Hydrophobic interactions dominate the recognition of a KRAS G12V neoantigen. Nat Commun 2023; 14:5063. [PMID: 37604828 PMCID: PMC10442379 DOI: 10.1038/s41467-023-40821-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 08/10/2023] [Indexed: 08/23/2023] Open
Abstract
Specificity remains a major challenge to current therapeutic strategies for cancer. Mutation associated neoantigens (MANAs) are products of genetic alterations, making them highly specific therapeutic targets. MANAs are HLA-presented (pHLA) peptides derived from intracellular mutant proteins that are otherwise inaccessible to antibody-based therapeutics. Here, we describe the cryo-EM structure of an antibody-MANA pHLA complex. Specifically, we determine a TCR mimic (TCRm) antibody bound to its MANA target, the KRASG12V peptide presented by HLA-A*03:01. Hydrophobic residues appear to account for the specificity of the mutant G12V residue. We also determine the structure of the wild-type G12 peptide bound to HLA-A*03:01, using X-ray crystallography. Based on these structures, we perform screens to validate the key residues required for peptide specificity. These experiments led us to a model for discrimination between the mutant and the wild-type peptides presented on HLA-A*03:01 based exclusively on hydrophobic interactions.
Collapse
Affiliation(s)
- Katharine M Wright
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA
- Discovery Chemistry, Protein and Structural Chemistry, Merck & Co, Inc, West Point, PA, 19846, USA
| | - Sarah R DiNapoli
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Michelle S Miller
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
| | - P Aitana Azurmendi
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA
| | - Xiaowei Zhao
- Janelia Research Campus, HHMI,19700 Helix Drive, Ashburn, VA, 20147, USA
| | - Zhiheng Yu
- Janelia Research Campus, HHMI,19700 Helix Drive, Ashburn, VA, 20147, USA
| | - Mayukh Chakrabarti
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - WuXian Shi
- Energy & Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY, 11973, USA
- Case Center for Synchrotron Biosciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jacqueline Douglass
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Michael S Hwang
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Emily Han-Chung Hsiue
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Novartis Institutes for BioMedical Research, 250 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Brian J Mog
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Alexander H Pearlman
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Suman Paul
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Division of Hematologic Malignancies and Bone Marrow Transplantation, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Maximilian F Konig
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Drew M Pardoll
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Chetan Bettegowda
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Nickolas Papadopoulos
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kenneth W Kinzler
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Bert Vogelstein
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Shibin Zhou
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA.
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Sandra B Gabelli
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Discovery Chemistry, Protein and Structural Chemistry, Merck & Co, Inc, West Point, PA, 19846, USA.
| |
Collapse
|
21
|
Abelin JG, Bergstrom EJ, Rivera KD, Taylor HB, Klaeger S, Xu C, Verzani EK, Jackson White C, Woldemichael HB, Virshup M, Olive ME, Maynard M, Vartany SA, Allen JD, Phulphagar K, Harry Kane M, Rachimi S, Mani DR, Gillette MA, Satpathy S, Clauser KR, Udeshi ND, Carr SA. Workflow enabling deepscale immunopeptidome, proteome, ubiquitylome, phosphoproteome, and acetylome analyses of sample-limited tissues. Nat Commun 2023; 14:1851. [PMID: 37012232 PMCID: PMC10070353 DOI: 10.1038/s41467-023-37547-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
Serial multi-omic analysis of proteome, phosphoproteome, and acetylome provides insights into changes in protein expression, cell signaling, cross-talk and epigenetic pathways involved in disease pathology and treatment. However, ubiquitylome and HLA peptidome data collection used to understand protein degradation and antigen presentation have not together been serialized, and instead require separate samples for parallel processing using distinct protocols. Here we present MONTE, a highly sensitive multi-omic native tissue enrichment workflow, that enables serial, deep-scale analysis of HLA-I and HLA-II immunopeptidome, ubiquitylome, proteome, phosphoproteome, and acetylome from the same tissue sample. We demonstrate that the depth of coverage and quantitative precision of each 'ome is not compromised by serialization, and the addition of HLA immunopeptidomics enables the identification of peptides derived from cancer/testis antigens and patient specific neoantigens. We evaluate the technical feasibility of the MONTE workflow using a small cohort of patient lung adenocarcinoma tumors.
Collapse
Affiliation(s)
- Jennifer G Abelin
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA.
| | - Erik J Bergstrom
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Keith D Rivera
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Hannah B Taylor
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Susan Klaeger
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Charles Xu
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Eva K Verzani
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - C Jackson White
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Hilina B Woldemichael
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Maya Virshup
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Meagan E Olive
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Myranda Maynard
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Stephanie A Vartany
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Joseph D Allen
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Kshiti Phulphagar
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - M Harry Kane
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Suzanna Rachimi
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - D R Mani
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Michael A Gillette
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
- Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Shankha Satpathy
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Karl R Clauser
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Namrata D Udeshi
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA.
| | - Steven A Carr
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
22
|
Martinov T, Greenberg PD. Targeting Driver Oncogenes and Other Public Neoantigens Using T Cell Receptor-Based Cellular Therapy. ANNUAL REVIEW OF CANCER BIOLOGY 2023; 7:331-351. [PMID: 37655310 PMCID: PMC10470615 DOI: 10.1146/annurev-cancerbio-061521-082114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
T cell reactivity to tumor-specific neoantigens can drive endogenous and therapeutically induced antitumor immunity. However, most tumor-specific neoantigens are unique to each patient (private) and targeting them requires personalized therapy. A smaller subset of neoantigens includes epitopes that span recurrent mutation hotspots, translocations, or gene fusions in oncogenic drivers and tumor suppressors, as well as epitopes that arise from viral oncogenic proteins. Such antigens are likely to be shared across patients (public), uniformly expressed within a tumor, and required for cancer cell survival and fitness. Although a limited number of these public neoantigens are naturally immunogenic, recent studies affirm their clinical utility. In this review, we highlight efforts to target mutant KRAS, mutant p53, and epitopes derived from oncogenic viruses using T cells engineered with off-the-shelf T cell receptors. We also discuss the challenges and strategies to achieving more effective T cell therapies, particularly in the context of solid tumors.
Collapse
Affiliation(s)
- Tijana Martinov
- Program in Immunology and Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Philip D Greenberg
- Program in Immunology and Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Immunology Department, University of Washington, Seattle, Washington, USA
| |
Collapse
|
23
|
Expansion of KRAS hotspot mutations reactive T cells from human pancreatic tumors using autologous T cells as the antigen-presenting cells. Cancer Immunol Immunother 2022; 72:1301-1313. [PMID: 36436020 DOI: 10.1007/s00262-022-03335-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/18/2022] [Indexed: 11/28/2022]
Abstract
Adoptive cell therapy (ACT) with expanded tumor-infiltrating lymphocytes (TIL) or TCR gene-modified T cells (TCR-T) that recognize mutant KRAS neo-antigens can mediate tumor regression in patients with advanced pancreatic ductal adenocarcinoma (PDAC) (Tran et al in N Engl J Med, 375:2255-2262, 2016; Leidner et al in N Engl J Med, 386:2112-2119, 2022). The mutant KRAS-targeted ACT holds great potential to achieve durable clinical responses for PDAC, which has had no meaningful improvement over 40 years. However, the wide application of mutant KRAS-centric ACT is currently limited by the rarity of TIL that recognize the mutant KRAS. In addition, PDAC is generally recognized as a poorly immunogenic tumor, and TILs in PDAC are less abundant than in immunogenic tumors such as melanoma. To increase the success rate of TIL production, we adopted a well-utilized K562-based artificial APC (aAPC) that expresses 4-1BBL as the costimulatory molecules to enhance the TIL production from PDCA. However, stimulation with K562-based aAPC led to a rapid loss of specificity to mutant KRAS. To selectively expand neo-antigen-specific T cells, particularly mKRAS, from the TILs, we used tandem mini gene-modified autologous T cells (TMG-T) as the novel aAPC. Using this modified IVS protocol, we successfully generated TIL cultures specifically reactive to mKRAS (G12V). We believe that autologous TMG-T cells provide a reliable source of autologous APC to expand a rare population of neoantigen-specific T cells in TILs.
Collapse
|
24
|
Bernhardt M, Cruz-Garcia Y, Rech A, Meierjohann S, Erhard F, Schilling B, Schlosser A. Extending the Mass Spectrometry-Detectable Landscape of MHC Peptides by Use of Restricted Access Material. Anal Chem 2022; 94:14214-14222. [PMID: 36194871 DOI: 10.1021/acs.analchem.2c02198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Mass spectrometry-based immunopeptidomics enables the comprehensive identification of major histocompatibility complex (MHC) peptides from a cell culture as well as from tissue or tumor samples and is applied for the identification of tumor-specific and viral T-cell epitopes. Although mass spectrometry is generally considered an "unbiased" method for MHC peptide identification, the physicochemical properties of MHC peptides can greatly influence their detectability. Here, we demonstrate that highly hydrophobic peptides are lost during sample preparation when C18 solid-phase extraction (SPE) is used for separating MHC peptides from proteins. To overcome this limitation, we established an optimized protocol involving restricted access material (RAM). Compared to C18-SPE, RAM-SPE improved the overall MHC peptide recovery and extended the landscape of mass spectrometry-detectable MHC peptides toward more hydrophobic peptides.
Collapse
Affiliation(s)
- Melissa Bernhardt
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, Julius Maximilians University Würzburg, 97080 Würzburg, Germany
| | - Yiliam Cruz-Garcia
- Department of Biochemistry and Molecular Biology, Julius Maximilians University Würzburg, 97080 Würzburg, Germany
| | - Anne Rech
- Department of Dermatology, Venereology, and Allergology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Svenja Meierjohann
- Institute of Pathology, Julius Maximilians University Würzburg, 97080 Würzburg, Germany
| | - Florian Erhard
- Institute for Virology and Immunobiology, Julius Maximilians University Würzburg, 97080 Würzburg, Germany
| | - Bastian Schilling
- Department of Dermatology, Venereology, and Allergology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Andreas Schlosser
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, Julius Maximilians University Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
25
|
Freed-Pastor WA, Aguirre AJ. Getting a handle on KRAS inhibitor resistance with hapten-mediated anti-tumor immunity. Cancer Cell 2022; 40:908-910. [PMID: 36099887 DOI: 10.1016/j.ccell.2022.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Covalent inhibitors of oncogenic KRASG12C have demonstrated impressive clinical responses; however, therapeutic resistance has been commonly observed. In this issue, Zhang and colleagues demonstrate that small molecule KRASG12C inhibitors can generate haptenated major histocompatibility complex (MHC) class I:peptide complexes, which represent attractive targets for immune-based therapies to combat pharmacologic resistance.
Collapse
Affiliation(s)
- William A Freed-Pastor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Andrew J Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Abstract
ABSTRACT The holy grail of cancer therapeutics is the destruction of cancer cells while avoiding harm to normal cells. Cancer is unique from normal tissues because of the presence of somatic mutations that accumulate during tumorigenesis. Some nonsynonymous mutations can give rise to mutated peptide antigens (hereafter referred to as neoantigens) that can be specifically recognized by T cells. Thus, the immunological targeting of neoantigens represents a safe and promising strategy to treat patients with cancer. This article reviews the clinical application of adoptive cell therapy targeting neoantigens in patients with epithelial cancers.
Collapse
|
27
|
Leidner R, Sanjuan Silva N, Huang H, Sprott D, Zheng C, Shih YP, Leung A, Payne R, Sutcliffe K, Cramer J, Rosenberg SA, Fox BA, Urba WJ, Tran E. Neoantigen T-Cell Receptor Gene Therapy in Pancreatic Cancer. N Engl J Med 2022; 386:2112-2119. [PMID: 35648703 PMCID: PMC9531755 DOI: 10.1056/nejmoa2119662] [Citation(s) in RCA: 315] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A patient with progressive metastatic pancreatic cancer was treated with a single infusion of 16.2×109 autologous T cells that had been genetically engineered to clonally express two allogeneic HLA-C*08:02-restricted T-cell receptors (TCRs) targeting mutant KRAS G12D expressed by the tumors. The patient had regression of visceral metastases (overall partial response of 72% according to the Response Evaluation Criteria in Solid Tumors, version 1.1); the response was ongoing at 6 months. The engineered T cells constituted more than 2% of all the circulating peripheral-blood T cells 6 months after the cell transfer. In this patient, TCR gene therapy targeting the KRAS G12D driver mutation mediated the objective regression of metastatic pancreatic cancer. (Funded by the Providence Portland Medical Foundation.).
Collapse
Affiliation(s)
- Rom Leidner
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - Nelson Sanjuan Silva
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - Huayu Huang
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - David Sprott
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - Chunhong Zheng
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - Yi-Ping Shih
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - Amy Leung
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - Roxanne Payne
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - Kim Sutcliffe
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - Julie Cramer
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - Steven A Rosenberg
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - Bernard A Fox
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - Walter J Urba
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| | - Eric Tran
- From the Earle A. Chiles Research Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., B.A.F., W.J.U., E.T.), Providence Cancer Institute (R.L., N.S.S., H.H., D.S., C.Z., Y.-P.S., A.L., R.P., K.S., J.C., B.A.F., W.J.U., E.T.), Portland, OR; and the Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (S.A.R.)
| |
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
|