201
|
Frankiw L, Baltimore D, Li G. Alternative mRNA splicing in cancer immunotherapy. Nat Rev Immunol 2019; 19:675-687. [PMID: 31363190 DOI: 10.1038/s41577-019-0195-7] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2019] [Indexed: 12/12/2022]
Abstract
Immunotherapies are yielding effective treatments for several previously untreatable cancers. Still, the identification of suitable antigens specific to the tumour that can be targets for cancer vaccines and T cell therapies is a challenge. Alternative processing of mRNA, a phenomenon that has been shown to alter the proteomic diversity of many cancers, may offer the potential of a broadened target space. Here, we discuss the promise of analysing mRNA processing events in cancer cells, with an emphasis on mRNA splicing, for the identification of potential new targets for cancer immunotherapy. Further, we highlight the challenges that must be overcome for this new avenue to have clinical applicability.
Collapse
Affiliation(s)
- Luke Frankiw
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| | - Guideng Li
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. .,Suzhou Institute of Systems Medicine, Suzhou, China.
| |
Collapse
|
202
|
Bioinformatic methods for cancer neoantigen prediction. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 164:25-60. [PMID: 31383407 DOI: 10.1016/bs.pmbts.2019.06.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor cells accumulate aberrations not present in normal cells, leading to presentation of neoantigens on MHC molecules on their surface. These non-self neoantigens distinguish tumor cells from normal cells to the immune system and are thus targets for cancer immunotherapy. The rapid development of molecular profiling platforms, such as next-generation sequencing, has enabled the generation of large datasets characterizing tumor cells. The simultaneous development of algorithms has enabled rapid and accurate processing of these data. Bioinformatic software tools encoding the algorithms can be strung together in a workflow to identify neoantigens. Here, with a focus on high-throughput sequencing, we review state-of-the art bioinformatic tools along with the steps and challenges involved in neoantigen identification and recognition.
Collapse
|
203
|
Totsch SK, Schlappi C, Kang KD, Ishizuka AS, Lynn GM, Fox B, Beierle EA, Whitley RJ, Markert JM, Gillespie GY, Bernstock JD, Friedman GK. Oncolytic herpes simplex virus immunotherapy for brain tumors: current pitfalls and emerging strategies to overcome therapeutic resistance. Oncogene 2019; 38:6159-6171. [PMID: 31289361 PMCID: PMC6771414 DOI: 10.1038/s41388-019-0870-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/21/2019] [Accepted: 06/22/2019] [Indexed: 12/25/2022]
Abstract
Malignant tumors of the central nervous system (CNS) continue to be a leading cause of cancer-related mortality in both
children and adults. Traditional therapies for malignant brain tumors consist of surgical resection and adjuvant chemoradiation;
such approaches are often associated with extreme morbidity. Accordingly, novel, targeted therapeutics for neoplasms of the CNS,
such as immunotherapy with oncolytic engineered herpes simplex virus (HSV) therapy, are urgently warranted. Herein, we discuss
treatment challenges related to HSV virotherapy delivery, entry, replication, and spread, and in so doing focus on host antiviral
immune responses and the immune microenvironment. Strategies to overcome such challenges including viral re-engineering,
modulation of the immunoregulatory microenvironment and combinatorial therapies with virotherapy, such as checkpoint inhibitors,
radiation, and vaccination are also examined in detail.
Collapse
Affiliation(s)
- Stacie K Totsch
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles Schlappi
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kyung-Don Kang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Brandon Fox
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Elizabeth A Beierle
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Richard J Whitley
- Division of Pediatric Infectious Disease, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - G Yancey Gillespie
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joshua D Bernstock
- Avidea Technologies, Inc, Baltimore, MD, USA. .,Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Gregory K Friedman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
204
|
Cimen Bozkus C, Roudko V, Finnigan JP, Mascarenhas J, Hoffman R, Iancu-Rubin C, Bhardwaj N. Immune Checkpoint Blockade Enhances Shared Neoantigen-Induced T-cell Immunity Directed against Mutated Calreticulin in Myeloproliferative Neoplasms. Cancer Discov 2019; 9:1192-1207. [PMID: 31266769 DOI: 10.1158/2159-8290.cd-18-1356] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 05/08/2019] [Accepted: 06/27/2019] [Indexed: 12/30/2022]
Abstract
Somatic frameshift mutations in the calreticulin (CALR) gene are key drivers of cellular transformation in myeloproliferative neoplasms (MPN). All patients carrying these mutations (CALR + MPN) share an identical sequence in the C-terminus of the mutated CALR protein (mut-CALR), with the potential for utility as a shared neoantigen. Here, we demonstrate that although a subset of patients with CALR + MPN develop specific T-cell responses against the mut-CALR C-terminus, PD-1 or CTLA4 expression abrogates the full complement of responses. Significantly, blockade of PD-1 and CLTA4 ex vivo by mAbs and of PD-1 in vivo by pembrolizumab administration restores mut-CALR-specific T-cell immunity in some patients with CALR + MPN. Moreover, mut-CALR elicits antigen-specific responses from both CD4+ and CD8+ T cells, confirming its broad applicability as an immunogen. Collectively, these results establish mut-CALR as a shared, MPN-specific neoantigen and inform the design of novel immunotherapies targeting mut-CALR. SIGNIFICANCE: Current treatment modalities for MPN are not effective in eliminating malignant cells. Here, we show that mutations in the CALR gene, which drive transformation in MPN, elicit T-cell responses that can be further enhanced by checkpoint blockade, suggesting immunotherapies could be employed to eliminate CALR + malignant cells in MPN.This article is highlighted in the In This Issue feature, p. 1143.
Collapse
Affiliation(s)
- Cansu Cimen Bozkus
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Vladimir Roudko
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John P Finnigan
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Camelia Iancu-Rubin
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nina Bhardwaj
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
205
|
Garcia-Garijo A, Fajardo CA, Gros A. Determinants for Neoantigen Identification. Front Immunol 2019; 10:1392. [PMID: 31293573 PMCID: PMC6601353 DOI: 10.3389/fimmu.2019.01392] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/03/2019] [Indexed: 12/22/2022] Open
Abstract
All tumors accumulate genetic alterations, some of which can give rise to mutated, non-self peptides presented by human leukocyte antigen (HLA) molecules and elicit T-cell responses. These immunogenic mutated peptides, or neoantigens, are foreign in nature and display exquisite tumor specificity. The correlative evidence suggesting they play an important role in the effectiveness of various cancer immunotherapies has triggered the development of vaccines and adoptive T-cell therapies targeting them. However, the systematic identification of personalized neoantigens in cancer patients, a critical requisite for the success of these therapies, remains challenging. A growing amount of evidence supports that only a small fraction of all tumor somatic non-synonymous mutations (NSM) identified represent bona fide neoantigens; mutated peptides that are processed, presented on the cell surface HLA molecules of cancer cells and are capable of triggering immune responses in patients. Here, we provide an overview of the existing strategies to identify candidate neoantigens and to evaluate their immunogenicity, two factors that impact on neoantigen identification. We will focus on their strengths and limitations to allow readers to rationally select and apply the most suitable method for their specific laboratory setting.
Collapse
Affiliation(s)
| | | | - Alena Gros
- Tumor Immunology and Immunotherapy, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
206
|
Tissue patrol by resident memory CD8+ T cells in human skin. Nat Immunol 2019; 20:756-764. [DOI: 10.1038/s41590-019-0404-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 04/16/2019] [Indexed: 12/13/2022]
|
207
|
Induction of neoantigen-reactive T cells from healthy donors. Nat Protoc 2019; 14:1926-1943. [DOI: 10.1038/s41596-019-0170-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 03/21/2019] [Indexed: 12/21/2022]
|
208
|
Yang W, Lee KW, Srivastava RM, Kuo F, Krishna C, Chowell D, Makarov V, Hoen D, Dalin MG, Wexler L, Ghossein R, Katabi N, Nadeem Z, Cohen MA, Tian SK, Robine N, Arora K, Geiger H, Agius P, Bouvier N, Huberman K, Vanness K, Havel JJ, Sims JS, Samstein RM, Mandal R, Tepe J, Ganly I, Ho AL, Riaz N, Wong RJ, Shukla N, Chan TA, Morris LGT. Immunogenic neoantigens derived from gene fusions stimulate T cell responses. Nat Med 2019; 25:767-775. [PMID: 31011208 PMCID: PMC6558662 DOI: 10.1038/s41591-019-0434-2] [Citation(s) in RCA: 273] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/22/2019] [Indexed: 12/27/2022]
Abstract
Anti-tumor immunity is driven by self versus non-self discrimination. Many immunotherapeutic approaches to cancer have taken advantage of tumor neoantigens derived from somatic mutations. Here, we demonstrate that gene fusions are a source of immunogenic neoantigens that can mediate responses to immunotherapy. We identified an exceptional responder with metastatic head and neck cancer who experienced a complete response to immune checkpoint inhibitor therapy, despite a low mutational load and minimal pre-treatment immune infiltration in the tumor. Using whole-genome sequencing and RNA sequencing, we identified a novel gene fusion and demonstrated that it produces a neoantigen that can specifically elicit a host cytotoxic T cell response. In a cohort of head and neck tumors with low mutation burden, minimal immune infiltration and prevalent gene fusions, we also identified gene fusion-derived neoantigens that generate cytotoxic T cell responses. Finally, analyzing additional datasets of fusion-positive cancers, including checkpoint-inhibitor-treated tumors, we found evidence of immune surveillance resulting in negative selective pressure against gene fusion-derived neoantigens. These findings highlight an important class of tumor-specific antigens and have implications for targeting gene fusion events in cancers that would otherwise be less poised for response to immunotherapy, including cancers with low mutational load and minimal immune infiltration.
Collapse
Affiliation(s)
- Wei Yang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ken-Wing Lee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Raghvendra M Srivastava
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fengshen Kuo
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chirag Krishna
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Diego Chowell
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vladimir Makarov
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Douglas Hoen
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin G Dalin
- Department of Pediatrics, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Leonard Wexler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nora Katabi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zaineb Nadeem
- Department of Surgery (Head and Neck Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marc A Cohen
- Department of Surgery (Head and Neck Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - S Ken Tian
- New York Genome Center, New York, NY, USA
| | | | | | | | | | - Nancy Bouvier
- Integrated Genomics Operation, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kety Huberman
- Integrated Genomics Operation, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katelynd Vanness
- Integrated Genomics Operation, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan J Havel
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jennifer S Sims
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert M Samstein
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rajarsi Mandal
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery (Head and Neck Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justin Tepe
- Department of Surgery (Head and Neck Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ian Ganly
- Department of Surgery (Head and Neck Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem Riaz
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard J Wong
- Department of Surgery (Head and Neck Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neerav Shukla
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Luc G T Morris
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Surgery (Head and Neck Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
209
|
Magalhaes I, Carvalho-Queiroz C, Hartana CA, Kaiser A, Lukic A, Mints M, Nilsson O, Grönlund H, Mattsson J, Berglund S. Facing the future: challenges and opportunities in adoptive T cell therapy in cancer. Expert Opin Biol Ther 2019; 19:811-827. [PMID: 30986360 DOI: 10.1080/14712598.2019.1608179] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION In recent years, immunotherapy for the treatment of solid cancer has emerged as a promising therapeutic alternative. Adoptive cell therapy (ACT), especially T cell-based, has been found to cause tumor regression and even cure in a percentage of treated patients. Checkpoint inhibitors further underscore the potential of the T cell compartment in the treatment of cancer. Not all patients respond to these treatments; however, many challenges remain. AREAS COVERED This review covers the challenges and progress in tumor antigen target identification and selection, and cell product manufacturing for T cell ACT. Tumor immune escape mechanisms and strategies to overcome those in the context of T cell ACT are also discussed. EXPERT OPINION The immunotherapy toolbox is rapidly expanding and improving, and the future promises further breakthroughs in the T cell ACT field. The heterogeneity of the tumor microenvironment and the multiplicity of tumor immune escape mechanisms pose formidable challenges to successful T cell immunotherapy in solid tumors, however. Individualized approaches and strategies combining treatments targeting different immunotherapeutic aspects will be needed in order to expand the applicability and improve the response rates in future.
Collapse
Affiliation(s)
- Isabelle Magalhaes
- a Department of Oncology-Pathology , Karolinska Institutet , Stockholm , Sweden
| | - Claudia Carvalho-Queiroz
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Ciputra Adijaya Hartana
- c Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital , Cambridge , MA , USA
| | - Andreas Kaiser
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Ana Lukic
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Michael Mints
- a Department of Oncology-Pathology , Karolinska Institutet , Stockholm , Sweden.,d Department of Surgical and Perioperative Sciences , Umeå University, Umeå, Sweden.,e Blood and Marrow Transplant Program, Medical Oncology and Hematology , Princess Margaret Cancer Center , Toronto , Canada.,f Department of Medicine , University of Toronto , Toronto , Canada
| | - Ola Nilsson
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Hans Grönlund
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Jonas Mattsson
- a Department of Oncology-Pathology , Karolinska Institutet , Stockholm , Sweden.,f Department of Medicine , University of Toronto , Toronto , Canada
| | - Sofia Berglund
- a Department of Oncology-Pathology , Karolinska Institutet , Stockholm , Sweden.,b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| |
Collapse
|
210
|
Ren SS, Deng JW, Hong M, Ren YL, Fu HJ, Liu YN, Chen Z. Ethical considerations of cellular immunotherapy for cancer. J Zhejiang Univ Sci B 2019; 20:23-31. [PMID: 30614227 DOI: 10.1631/jzus.b1800421] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
With the rapid development of immunology, molecular biology, and associated technologies such as next-generation sequencing, cellular immunotherapy has recently become the fourth major cancer treatment. Immunotherapies based on T cells, natural killer cells, and dendritic cells play key roles in cancer immunotherapy. However, their application in clinical practice raises several ethical issues. Thus, studies should focus on proper adherence to basic ethical principles that can effectively guide and solve related clinical problems in the course of treatment, improve treatment effects, and protect the rights and interests of patients. In this review, we discuss cellular immunotherapy-related ethical issues and highlight the ethical practices and current status of cellular immunotherapy in China. These considerations may supplement existing ethical standards in cancer immunotherapy.
Collapse
Affiliation(s)
- Sang-Sang Ren
- Health Policy and Hospital Management Research Center, Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jing-Wen Deng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Meng Hong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yan-Li Ren
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hai-Jing Fu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yan-Ning Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhi Chen
- Health Policy and Hospital Management Research Center, Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China.,State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
211
|
Kong P, Wang J, Song Z, Liu S, He W, Jiang C, Xie Q, Yang L, Xia X, Xia L. Circulating Lymphocytes, PD-L1 Expression on Tumor-infiltrating Lymphocytes, and Survival of Colorectal Cancer Patients with Different Mismatch Repair Gene Status. J Cancer 2019; 10:1745-1754. [PMID: 31205530 PMCID: PMC6547994 DOI: 10.7150/jca.25187] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/02/2019] [Indexed: 12/26/2022] Open
Abstract
Clinical outcomes of checkpoint blockade immunotherapy on colorectal cancer (CRC) are influenced by mismatch repair (MMR) gene status, which is associated with distinct tumor immune infiltrates and systemic inflammatory response status. However, the prognostic value of PD-L1 expression and the systemic inflammatory response for patients with MMR deficiency has not been fully investigated. In this study, we examined the association of systemic inflammatory markers, PD-1/PD-L1 pathway expression, microsatellite instability (MSI) status, and clinicopathological characteristics of CRC with patient survival between MMR-deficient (dMMR) group (N=168) and MMR-proficient (pMMR) group (N=169). We found a large proportion of dMMR CRC patients displayed increased level of systemic inflammatory markers such as C-reactive protein, Neutrophil/Lymphocyte Ratio (NLR), Glasgow Prognostic Score (GPS), and low expression of PD-L1 in tumor stroma. Several systemic inflammatory markers were associated with AJCC stage only in dMMR patients. Similarly, Tumor infiltrating lymphocyte (TIL) PD-L1 or stroma PD-L1 expression was associated with AJCC stage only in dMMR patients. Circulating serum lymphocytes and TIL PD-L1 expression are both independent prognosis predictors for CRC patients. Overall, we found that dMMR CRC displayed a comprehensively distinct tumor immune microenvironment and systemic inflammatory response makers. PD-L1 expression at different location has different impacts on CRC patient survival, and the TIL PD-L1 expression might be a potential predictor for dMMR CRC patient response to anti-PD-1 therapy.
Collapse
Affiliation(s)
- Pengfei Kong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.,Department of the VIP region, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Jing Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.,Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Ze Song
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.,Department of the Medical Imaging and Interventional Radiology, Sun Yat-sen University Cancer Centre, Guangzhou, 510000, PR China
| | - Shousheng Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.,Department of the VIP region, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Wenzhuo He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.,Department of the VIP region, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Chang Jiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.,Department of the VIP region, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Qiankun Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.,Department of the VIP region, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Lin Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.,Department of the VIP region, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Liangping Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.,Department of the VIP region, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| |
Collapse
|
212
|
Abstract
Engineered immune-cell-based cancer therapies have demonstrated robust efficacy in B cell malignancies, but challenges such as the lack of ideal targetable tumour antigens, tumour-mediated immunosuppression and severe toxicity still hinder their therapeutic efficacy and broad applicability. Synthetic biology can be used to overcome these challenges and create more robust, effective adaptive therapies that enable the specific targeting of cancer cells while sparing healthy cells. In this Progress article, we review recently developed gene circuit therapies for cancer using immune cells, nucleic acids and bacteria as chassis. We conclude by discussing outstanding challenges and future directions for realizing these gene circuit therapies in the clinic.
Collapse
Affiliation(s)
- Ming-Ru Wu
- Synthetic Biology Group, Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Barbara Jusiak
- Synthetic Biology Group, Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Timothy K Lu
- Synthetic Biology Group, Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Biophysics Program, Harvard University, Boston, MA, USA.
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
213
|
Cao S, Wylie KM, Wyczalkowski MA, Karpova A, Ley J, Sun S, Mashl RJ, Liang WW, Wang X, Johnson K, DiPersio JF, Gay H, Ratner L, Chen F, Adkins DR, Ding L. Dynamic host immune response in virus-associated cancers. Commun Biol 2019; 2:109. [PMID: 30911684 PMCID: PMC6430765 DOI: 10.1038/s42003-019-0352-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/07/2019] [Indexed: 02/06/2023] Open
Abstract
Viruses drive carcinogenesis in human cancers through diverse mechanisms that have not been fully elucidated but include promoting immune escape. Here we investigated associations between virus-positivity and immune pathway alteration for 2009 tumors across six virus-related cancer types. Analysis revealed that for 3 of 72 human papillomavirus (HPV)-positive head and neck squamous cell carcinoma (HNSC) the HPV genome integrated in immune checkpoint genes PD-L1 or PD-L2, driving elevated expression in the corresponding gene. In addition to the previously described upregulation of the PD-1 immunosuppressive pathway in Epstein-Barr virus (EBV)-positive stomach tumors, we also observed upregulation of the PD-1 pathway in cytomegalovirus (CMV)-positive tumors. Furthermore, we found signatures of T-cell and B-cell response in HPV-positive HNSC and EBV-positive stomach tumors and HPV-positive HNSC patients were associated with better survival when T-cell signals were detected. Our work reveals that viral infection may recruit immune effector cells, and upregulate PD-1 and CTLA-4 immunosuppressive pathways.
Collapse
Affiliation(s)
- Song Cao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110 USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108 USA
| | - Kristine M. Wylie
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108 USA
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63110 USA
| | - Matt A. Wyczalkowski
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110 USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108 USA
| | - Alla Karpova
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110 USA
| | - Jessica Ley
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110 USA
| | - Sam Sun
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110 USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108 USA
| | - R. Jay Mashl
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110 USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108 USA
| | - Wen-Wei Liang
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110 USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108 USA
| | - Xiaowei Wang
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO 63110 USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110 USA
| | - Kimberly Johnson
- Brown School Master of Public Health Program, Washington University in St. Louis, St. Louis, MO 63130 USA
| | - John F. DiPersio
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110 USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110 USA
| | - Hiram Gay
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110 USA
| | - Lee Ratner
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110 USA
| | - Feng Chen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110 USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110 USA
| | - Douglas R. Adkins
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110 USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110 USA
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110 USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108 USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110 USA
- Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110 USA
| |
Collapse
|
214
|
Matsuda T, Miyauchi E, Hsu YW, Nagayama S, Kiyotani K, Zewde M, Park JH, Kato T, Harada M, Matsui S, Ueno M, Fukuda K, Suzuki N, Hazama S, Nagano H, Takeuchi H, Vigneswaran WT, Kitagawa Y, Nakamura Y. TCR sequencing analysis of cancer tissues and tumor draining lymph nodes in colorectal cancer patients. Oncoimmunology 2019; 8:e1588085. [PMID: 31069156 DOI: 10.1080/2162402x.2019.1588085] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/21/2019] [Accepted: 02/16/2019] [Indexed: 12/15/2022] Open
Abstract
Tumor draining lymph nodes (TDLNs) are located in the routes of lymphatic drainage from a primary tumor and have the highest risk of metastasis in various types of solid tumors. TDLNs are also considered as a tissue to activate the antitumor immunity, where antigen-specific effector T cells are generated. However, T cell receptor (TCR) repertoires in TDLNs have not been well characterized. We collected 23 colorectal cancer tumors with 203 lymph nodes with/without metastatic cancer cells (67 were metastasis-positive and the remaining 136 were metastasis-negative) and performed TCR sequencing. Metastasis-positive TDLNs showed a significantly lower TCR diversity and shared TCR clonotypes more frequently with primary tumor tissues compared to metastasis-negative TDLNs. Principal component analysis indicated that TDLNs with metastasis showed similar TCR repertoires. These findings suggest that cancer-reactive T cell clones could be enriched in the metastasis-positive TDLNs.
Collapse
Affiliation(s)
- Tatsuo Matsuda
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Department of Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Eisaku Miyauchi
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Yu-Wen Hsu
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - Satoshi Nagayama
- Department of Gastroenterological Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kazuma Kiyotani
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makda Zewde
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jae-Hyun Park
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Taigo Kato
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Makiko Harada
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Shimpei Matsui
- Department of Gastroenterological Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masashi Ueno
- Department of Gastroenterological Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kazumasa Fukuda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Nobuaki Suzuki
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Shoichi Hazama
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hiroya Takeuchi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Wickii T Vigneswaran
- Department of Thoracic and Cardiovascular Surgery, Loyola University Medical Center, Maywood, IL, USA
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yusuke Nakamura
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
215
|
Albers JJ, Ammon T, Gosmann D, Audehm S, Thoene S, Winter C, Secci R, Wolf A, Stelzl A, Steiger K, Ruland J, Bassermann F, Kupatt C, Anton M, Krackhardt AM. Gene editing enables T-cell engineering to redirect antigen specificity for potent tumor rejection. Life Sci Alliance 2019; 2:2/2/e201900367. [PMID: 30877233 PMCID: PMC6421629 DOI: 10.26508/lsa.201900367] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 01/04/2023] Open
Abstract
Targeted integration of a tumor-reactive T-cell receptor into the TRAC locus using CRISPR-Cas9 and AAV6 redirects primary human T cells against tumor cells in vitro and in vivo. Adoptive transfer of TCR transgenic T cells holds great promise for treating various cancers. So far, mainly semi-randomly integrating vectors have been used to genetically modify T cells. These carry the risk of insertional mutagenesis, and the sole addition of an exogenous TCR potentially results in the mispairing of TCR chains with endogenous ones. Established approaches using nonviral vectors, such as transposons, already reduce the risk of insertional mutagenesis but have not accomplished site-specific integration. Here, we used CRISPR-Cas9 RNPs and adeno-associated virus 6 for gene targeting to deliver an engineered TCR gene specifically to the TCR alpha constant locus, thus placing it under endogenous transcriptional control. Our data demonstrate that this approach replaces the endogenous TCR, functionally redirects the edited T cells’ specificity in vitro, and facilitates potent tumor rejection in an in vivo xenograft model.
Collapse
Affiliation(s)
- Julian J Albers
- Klinik und Poliklinik für Innere Medizin III, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Tim Ammon
- Experimental Hematology Group, Klinik und Poliklinik für Innere Medizin III, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Dario Gosmann
- Klinik und Poliklinik für Innere Medizin III, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Stefan Audehm
- Klinik und Poliklinik für Innere Medizin III, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Silvia Thoene
- Institut für Klinische Chemie und Pathobiochemie, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany.,German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christof Winter
- Institut für Klinische Chemie und Pathobiochemie, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany.,German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ramona Secci
- Institut für Klinische Chemie und Pathobiochemie, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Anja Wolf
- Klinik und Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Anja Stelzl
- Klinik und Poliklinik für Innere Medizin III, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Katja Steiger
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Technische Universität München, Munich, Germany.,German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Ruland
- Institut für Klinische Chemie und Pathobiochemie, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany.,German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Florian Bassermann
- Klinik und Poliklinik für Innere Medizin III, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany.,German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner-Site Munich Heart Alliance, Munich, Germany
| | - Martina Anton
- Institut für Molekulare Immunologie und Experimentelle Onkologie und Therapieforschung, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Angela M Krackhardt
- Klinik und Poliklinik für Innere Medizin III, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany .,German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
216
|
Calvo Tardón M, Allard M, Dutoit V, Dietrich PY, Walker PR. Peptides as cancer vaccines. Curr Opin Pharmacol 2019; 47:20-26. [PMID: 30831470 DOI: 10.1016/j.coph.2019.01.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/30/2022]
Abstract
Cancer vaccines based on synthetic peptides are a safe, well-tolerated immunotherapy able to specifically stimulate tumor-reactive T cells. However, their clinical efficacy does not approach that achieved with other immunotherapies such as immune checkpoint blockade. Nevertheless, major advances have been made in selecting tumor antigens to target, identifying epitopes binding to classical and non-classical HLA molecules, and incorporating these into optimal sized peptides for formulation into a vaccine. Limited potency of currently used adjuvants and the immunosuppressive tumor microenvironment are now understood to be major impediments to vaccine efficacy that need to be overcome. Rationally designed combination therapies are now being tested and should ultimately enable peptide vaccination to be added to immuno-oncology treatment options.
Collapse
Affiliation(s)
- Marta Calvo Tardón
- Center for Translational Research in Onco-Hematology, Division of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Mathilde Allard
- Center for Translational Research in Onco-Hematology, Division of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Valérie Dutoit
- Center for Translational Research in Onco-Hematology, Department of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Pierre-Yves Dietrich
- Center for Translational Research in Onco-Hematology, Department of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Paul R Walker
- Center for Translational Research in Onco-Hematology, Division of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.
| |
Collapse
|
217
|
Iiizumi S, Ohtake J, Murakami N, Kouro T, Kawahara M, Isoda F, Hamana H, Kishi H, Nakamura N, Sasada T. Identification of Novel HLA Class II-Restricted Neoantigens Derived from Driver Mutations. Cancers (Basel) 2019; 11:cancers11020266. [PMID: 30813491 PMCID: PMC6406322 DOI: 10.3390/cancers11020266] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/30/2022] Open
Abstract
Neoantigens derived from tumor-specific genetic mutations might be suitable targets for cancer immunotherapy because of their high immunogenicity. In the current study, we evaluated the immunogenicity of 10 driver mutations that are frequently expressed in various cancers using peripheral blood mononuclear cells from healthy donors (n = 25). Of the 10 synthetic peptides (27-mer) derived from these mutations, the six peptides from KRAS-G12D, KRAS-G12R, KRAS-G13D, NRAS-Q61R, PIK3CA-H1047R, and C-Kit-D816V induced T cell responses, suggesting that frequent driver mutations are not always less immunogenic. In particular, immune responses to PIK3CA-H1047R, C-Kit-D816V, KRAS-G13D, and NRAS-Q61R were observed in more than 10% of the donors. All six peptides induced human leukocyte antigen (HLA) class II-restricted CD4+ T cell responses; notably, PIK3CA-H1047R contained at least two different CD4+ T cell epitopes restricted to different HLA class II alleles. In addition, PIK3CA-H1047R and C-Kit-D816V induced antigen-specific CD8+ T cells as well, indicating that they might contain both HLA class I- and class II-restricted epitopes. Since the identified neoantigens might be shared by patients with various types of cancers and are not easily lost due to immune escape, they have the potential to be promising off-the-shelf cancer immunotherapy targets in patients with the corresponding mutations.
Collapse
Affiliation(s)
- Susumu Iiizumi
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
- Research & Early Development Division, BrightPath Biotherapeutics Co., Ltd., 3-25-22 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan.
| | - Junya Ohtake
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
| | - Naoko Murakami
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
- Research & Early Development Division, BrightPath Biotherapeutics Co., Ltd., 3-25-22 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan.
| | - Taku Kouro
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
| | - Mamoru Kawahara
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
| | - Fumiko Isoda
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
- Research & Early Development Division, BrightPath Biotherapeutics Co., Ltd., 3-25-22 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan.
| | - Hiroshi Hamana
- Department of Innovative Cancer Immunotherapy, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Hiroyuki Kishi
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Norihiro Nakamura
- Research & Early Development Division, BrightPath Biotherapeutics Co., Ltd., 3-25-22 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan.
| | - Tetsuro Sasada
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
| |
Collapse
|
218
|
Aurisicchio L, Salvatori E, Lione L, Bandini S, Pallocca M, Maggio R, Fanciulli M, De Nicola F, Goeman F, Ciliberto G, Conforti A, Luberto L, Palombo F. Poly-specific neoantigen-targeted cancer vaccines delay patient derived tumor growth. J Exp Clin Cancer Res 2019; 38:78. [PMID: 30764846 PMCID: PMC6376688 DOI: 10.1186/s13046-019-1084-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/06/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Personalized cancer vaccines based on neoantigens have reached the clinical trial stage in melanoma. Different vaccination protocols showed efficacy in preclinical models without a clear indication of the quality and the number of neoantigens required for an effective cancer vaccine. METHODS In an effort to develop potent and efficacious neoantigen-based vaccines, we have developed different neoantigen minigene (NAM) vaccine vectors to determine the rules for a successful neoantigen cancer vaccine (NCV) delivered by plasmid DNA and electroporation. Immune responses were analyzed at the level of single neoantigen by flow cytometry and correlated with tumor growth. Adoptive T cell transfer, from HLA-2.1.1 mice, was used to demonstrate the efficacy of the NCV pipeline against human-derived tumors. RESULTS In agreement with previous bodies of evidence, immunogenicity was driven by predicted affinity. A strong poly-functional and poly-specific immune response was observed with high affinity neoantigens. However, only a high poly-specific vaccine vector was able to completely protect mice from subsequent tumor challenge. More importantly, this pipeline - from the selection of neoantigens to vaccine design - applied to a new model of patient derived tumor xenograft resulted in therapeutic treatment. CONCLUSIONS These results suggest a feasible strategy for a neoantigen cancer vaccine that is simple and applicable for clinical developments.
Collapse
Affiliation(s)
| | | | - Lucia Lione
- Takis, Rome, Italy
- Università Magna Grecia, Catanzaro, Italy
| | | | - Matteo Pallocca
- UOSD SAFU, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | | | | | | | - Frauke Goeman
- Oncogenomic and Epigenetic Unit, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | | | | | | | | |
Collapse
|
219
|
Schober K, Buchholz VR, Busch DH. TCR repertoire evolution during maintenance of CMV-specific T-cell populations. Immunol Rev 2019; 283:113-128. [PMID: 29664573 DOI: 10.1111/imr.12654] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During infections and cancer, the composition of the T-cell receptor (TCR) repertoire of antigen-specific CD8+ T cells changes over time. TCR avidity is thought to be a major driver of this process, thereby interacting with several additional regulators of T-cell responses to form a composite immune response architecture. Infections with latent viruses, such as cytomegalovirus (CMV), can lead to large T-cell responses characterized by an oligoclonal TCR repertoire. Here, we review the current status of experimental studies and theoretical models of TCR repertoire evolution during CMV infection. We will particularly discuss the degree to which this process may be determined through structural TCR avidity. As engineered TCR-redirected T cells have moved into the spotlight for providing more effective immunotherapies, it is essential to understand how the key features of a given TCR influence T-cell expansion and maintenance in settings of infection or malignancy. Deeper insights into these mechanisms will improve our basic understanding of T-cell immunology and help to identify optimal TCRs for immunotherapy.
Collapse
Affiliation(s)
- Kilian Schober
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Veit R Buchholz
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany.,Focus Group 'Clinical Cell Processing and Purification', Institute for Advanced Study, TUM, Munich, Germany.,National Centre for Infection Research (DZIF), Munich, Germany
| |
Collapse
|
220
|
Ren L, Leisegang M, Deng B, Matsuda T, Kiyotani K, Kato T, Harada M, Park JH, Saloura V, Seiwert T, Vokes E, Agrawal N, Nakamura Y. Identification of neoantigen-specific T cells and their targets: implications for immunotherapy of head and neck squamous cell carcinoma. Oncoimmunology 2019; 8:e1568813. [PMID: 30906664 PMCID: PMC6422382 DOI: 10.1080/2162402x.2019.1568813] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/23/2018] [Accepted: 11/02/2018] [Indexed: 01/04/2023] Open
Abstract
To develop a practically applicable method for T-cell receptor (TCR)-engineered T cell immunotherapy targeting neoantigens, we have been attempting to identify neoantigen-specific T cell receptors (TCRs) and establish TCR-engineered T cells in a 3-4-month period. In this study, we report the characterization of T cell repertoires in tumor microenvironment (TME) and identification of neoantigen-specific TCRs after stimulation of patient-derived T cells. We screened 15 potential neoantigen peptides and successfully identified two CD8+HLA-dextramer+ T cells, which recognized MAGOHBG17A and ZCCHC14P368L. All three dominant TCR clonotypes from MAGOHBG17A-HLA dextramer-sorted CD8+ T cells were also found in T cells in TME, while none of dominant TCR clonotypes from ZCCHC14P368L-HLA dextramer-sorted CD8+ T cells was found in the corresponding TME. The most dominant TCRA/TCRB pairs for these two neoantigens were cloned into HLA-matched healthy donors' T lymphocytes to generate TCR-engineered T cells. The functional assay showed MAGOHBG17A TCR-engineered T cells could be significantly activated in a mutation-specific, HLA-restricted and peptide-dose-dependent manner while ZCCHC14P368L TCR-engineered T cells could not. Our data showed neoantigen-reactive T cell clonotypes that were identified in the patient's peripheral blood could be present in the corresponding TME and might be good TCRs targeting neoantigens.
Collapse
Affiliation(s)
- Lili Ren
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Cytotherapy Laboratory, Shenzhen People's Hospital (The second Clinical Medical College of Jinan University), Shenzhen, China
| | - Matthias Leisegang
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Boya Deng
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Tatsuo Matsuda
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Kazuma Kiyotani
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Taigo Kato
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Makiko Harada
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jae-Hyun Park
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | | | - Tanguy Seiwert
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Everett Vokes
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Nishant Agrawal
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Yusuke Nakamura
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,Department of Surgery, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
221
|
Lugli E, Kvistborg P, Galletti G. Cancer neoantigens targeted by adoptive T cell transfer: private no more. J Clin Invest 2019; 129:949-951. [PMID: 30714989 DOI: 10.1172/jci126295] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Effector T cell responses directed toward cancer neoantigens mediate tumor regression following checkpoint blockade or adoptive T cell immunotherapy, but are generally "private", thus requiring considerable effort for their identification. In this issue of the JCI, Malekzadeh et al. show that a fraction of patients with epithelial cancers mount antigen-specific T cell responses to "hot spot" p53 mutations that in some cases are shared among patients. This work suggests that other genes frequently mutated in human cancer can be immunogenic, thus offering a rapid way to screen for cancer neoantigens that can be targeted by subsequent T cell-based therapies.
Collapse
Affiliation(s)
- Enrico Lugli
- Laboratory of Translational Immunology and.,Flow Cytometry Core, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Pia Kvistborg
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | | |
Collapse
|
222
|
van der Lee DI, Reijmers RM, Honders MW, Hagedoorn RS, de Jong RC, Kester MG, van der Steen DM, de Ru AH, Kweekel C, Bijen HM, Jedema I, Veelken H, van Veelen PA, Heemskerk MH, Falkenburg JF, Griffioen M. Mutated nucleophosmin 1 as immunotherapy target in acute myeloid leukemia. J Clin Invest 2019; 129:774-785. [PMID: 30640174 PMCID: PMC6355238 DOI: 10.1172/jci97482] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/06/2018] [Indexed: 12/15/2022] Open
Abstract
The most frequent subtype of acute myeloid leukemia (AML) is defined by mutations in the nucleophosmin 1 (NPM1) gene. Mutated NPM1 (ΔNPM1) is an attractive target for immunotherapy, since it is an essential driver gene and 4 bp frameshift insertions occur in the same hotspot in 30%-35% of AMLs, resulting in a C-terminal alternative reading frame of 11 aa. By searching the HLA class I ligandome of primary AMLs, we identified multiple ΔNPM1-derived peptides. For one of these peptides, HLA-A*02:01-binding CLAVEEVSL, we searched for specific T cells in healthy individuals using peptide-HLA tetramers. Tetramer-positive CD8+ T cells were isolated and analyzed for reactivity against primary AMLs. From one clone with superior antitumor reactivity, we isolated the T cell receptor (TCR) and demonstrated specific recognition and lysis of HLA-A*02:01-positive ΔNPM1 AML after retroviral transfer to CD8+ and CD4+ T cells. Antitumor efficacy of TCR-transduced T cells was confirmed in immunodeficient mice engrafted with a human AML cell line expressing ΔNPM1. In conclusion, the data show that ΔNPM1-derived peptides are presented on AML and that CLAVEEVSL is a neoantigen that can be efficiently targeted on AML by ΔNPM1 TCR gene transfer. Immunotherapy targeting ΔNPM1 may therefore contribute to treatment of AML.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Female
- HLA-A2 Antigen/genetics
- HLA-A2 Antigen/immunology
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Mutation
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- Nuclear Proteins/genetics
- Nuclear Proteins/immunology
- Nucleophosmin
- Peptides/genetics
- Peptides/immunology
- Transduction, Genetic
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Arnoud H. de Ru
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | | | | | | | | | - Peter A. van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | | | | | | |
Collapse
|
223
|
Jiang N, Schonnesen AA, Ma KY. Ushering in Integrated T Cell Repertoire Profiling in Cancer. Trends Cancer 2019; 5:85-94. [PMID: 30755308 PMCID: PMC6544389 DOI: 10.1016/j.trecan.2018.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 12/28/2022]
Abstract
Advances in immune profiling techniques have dramatically changed the cancer immunotherapy and monitoring landscape. High-throughput protein and gene expression technologies have paved the way for the discovery of therapeutic targets and biomarkers, and have made monitoring therapeutic response possible through the ability to independently assay the phenotype, specificity, exhaustion status, and lineage of single T cells. Although valuable insights into response profiling have been gained with current technologies, it has become evident that single-method profiling is insufficient to accurately capture an antitumor T cell response. We discuss and propose new methods that combine multiple axes of analysis to provide a comprehensive analysis of T cell repertoire in the fight against cancer.
Collapse
Affiliation(s)
- Ning Jiang
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA; LIVESTRONG Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA.
| | - Alexandra A Schonnesen
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Ke-Yue Ma
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
224
|
Alcazer V, Bonaventura P, Tonon L, Wittmann S, Caux C, Depil S. Neoepitopes-based vaccines: challenges and perspectives. Eur J Cancer 2019; 108:55-60. [DOI: 10.1016/j.ejca.2018.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/27/2018] [Accepted: 12/05/2018] [Indexed: 12/30/2022]
|
225
|
Bradley P, Thomas PG. Using T Cell Receptor Repertoires to Understand the Principles of Adaptive Immune Recognition. Annu Rev Immunol 2019; 37:547-570. [PMID: 30699000 DOI: 10.1146/annurev-immunol-042718-041757] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adaptive immune recognition is mediated by antigen receptors on B and T cells generated by somatic recombination during lineage development. The high level of diversity resulting from this process posed technical limitations that previously limited the comprehensive analysis of adaptive immune recognition. Advances over the last ten years have produced data and approaches allowing insights into how T cells develop, evolutionary signatures of recombination and selection, and the features of T cell receptors that mediate epitope-specific binding and T cell activation. The size and complexity of these data have necessitated the generation of novel computational and analytical approaches, which are transforming how T cell immunology is conducted. Here we review the development and application of novel biological, theoretical, and computational methods for understanding T cell recognition and discuss the potential for improved models of receptor:antigen interactions.
Collapse
Affiliation(s)
- Philip Bradley
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA; .,Institute for Protein Design, University of Washington, Seattle, Washington 98195, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA;
| |
Collapse
|
226
|
Li G, Bethune MT, Wong S, Joglekar AV, Leonard MT, Wang JK, Kim JT, Cheng D, Peng S, Zaretsky JM, Su Y, Luo Y, Heath JR, Ribas A, Witte ON, Baltimore D. T cell antigen discovery via trogocytosis. Nat Methods 2019; 16:183-190. [PMID: 30700903 DOI: 10.1038/s41592-018-0305-7] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/13/2018] [Indexed: 01/04/2023]
Abstract
T cell receptor (TCR) ligand discovery is essential for understanding and manipulating immune responses to tumors. We developed a cell-based selection platform for TCR ligand discovery that exploits a membrane transfer phenomenon called trogocytosis. We discovered that T cell membrane proteins are transferred specifically to target cells that present cognate peptide-major histocompatibility complex (MHC) molecules. Co-incubation of T cells expressing an orphan TCR with target cells collectively presenting a library of peptide-MHCs led to specific labeling of cognate target cells, enabling isolation of these target cells and sequencing of the cognate TCR ligand. We validated this method for two clinically employed TCRs and further used the platform to identify the cognate neoepitope for a subject-derived neoantigen-specific TCR. Thus, target cell trogocytosis is a robust tool for TCR ligand discovery that will be useful for studying basic tumor immunology and identifying new targets for immunotherapy.
Collapse
Affiliation(s)
- Guideng Li
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA. .,Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. .,Suzhou Institute of Systems Medicine, Suzhou, China.
| | - Michael T Bethune
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| | - Stephanie Wong
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Alok V Joglekar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Michael T Leonard
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jessica K Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jocelyn T Kim
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Donghui Cheng
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Songming Peng
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jesse M Zaretsky
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yapeng Su
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.,Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Yicheng Luo
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - James R Heath
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Parker Institute for Cancer Immunotherapy (PICI) Center, California Institute of Technology, Pasadena, CA, USA
| | - Antoni Ribas
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Division of Hematology & Oncology, Department of Medicine, and Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.,Parker Institute for Cancer Immunotherapy (PICI) Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Owen N Witte
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Parker Institute for Cancer Immunotherapy (PICI) Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA. .,Parker Institute for Cancer Immunotherapy (PICI) Center, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
227
|
Xu Y, Salazar GT, Zhang N, An Z. T-cell receptor mimic (TCRm) antibody therapeutics against intracellular proteins. Antib Ther 2019; 2:22-32. [PMID: 33928218 PMCID: PMC7990144 DOI: 10.1093/abt/tbz001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/03/2019] [Accepted: 01/10/2019] [Indexed: 12/21/2022] Open
Abstract
T-cell receptor mimic (TCRm) antibodies combine the capacity of a T cell to target intracellular antigens with other capacities unique to antibodies. Neoantigens are abnormal proteins that arise as a consequence of somatic mutations. Technological advances promote the development of neoantigen-targeting therapies including TCRm antibody therapies. This review summarizes key characteristics of TCRm antibodies, in particular those targeting neoantigens, and further introduces discussion of obstacles that must be overcome to advance TCRm therapeutics.
Collapse
Affiliation(s)
- Yixiang Xu
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Georgina To'a Salazar
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
228
|
Arora J, McLaren PJ, Chaturvedi N, Carrington M, Fellay J, Lenz TL. HIV peptidome-wide association study reveals patient-specific epitope repertoires associated with HIV control. Proc Natl Acad Sci U S A 2019; 116:944-949. [PMID: 30602460 PMCID: PMC6338868 DOI: 10.1073/pnas.1812548116] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Genetic variation in the peptide-binding groove of the highly polymorphic HLA class I molecules has repeatedly been associated with HIV-1 control and progression to AIDS, accounting for up to 12% of the variation in HIV-1 set point viral load (spVL). This suggests a key role in disease control for HLA presentation of HIV-1 epitopes to cytotoxic T cells. However, a comprehensive understanding of the relevant HLA-bound HIV epitopes is still elusive. Here we describe a peptidome-wide association study (PepWAS) approach that integrates HLA genotypes and spVL data from 6,311 HIV-infected patients to interrogate the entire HIV-1 proteome (3,252 unique peptides) for disease-relevant peptides. This PepWAS approach predicts a core set of epitopes associated with spVL, including known epitopes but also several previously uncharacterized disease-relevant peptides. More important, each patient presents only a small subset of these predicted core epitopes through their individual HLA-A and HLA-B variants. Eventually, the individual differences in these patient-specific epitope repertoires account for the variation in spVL that was previously associated with HLA genetic variation. PepWAS thus enables a comprehensive functional interpretation of the robust but little-understood association between HLA and HIV-1 control, prioritizing a short list of disease-associated epitopes for the development of targeted therapy.
Collapse
Affiliation(s)
- Jatin Arora
- Research Group for Evolutionary Immunogenomics, Max Planck Institute for Evolutionary Biology, 24306 Plön, Germany
| | - Paul J McLaren
- JC Wilt Infectious Diseases Research Center, Public Health Agency of Canada, Winnipeg, R3E 0W3, Canada
- National HIV and Retrovirology Laboratory, Public Health Agency of Canada, Winnipeg, R3E 0W3, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, R3E 0J9, Canada
| | - Nimisha Chaturvedi
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Mary Carrington
- Cancer and Inflammation Program, Leidos Biomedical Research, Frederick National Laboratory, Frederick, MD 21702
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139-3583
| | - Jacques Fellay
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Tobias L Lenz
- Research Group for Evolutionary Immunogenomics, Max Planck Institute for Evolutionary Biology, 24306 Plön, Germany;
| |
Collapse
|
229
|
Yusko E, Vignali M, Wilson RK, Mardis ER, Hodi FS, Horak C, Chang H, Woods DM, Robins H, Weber J. Association of Tumor Microenvironment T-cell Repertoire and Mutational Load with Clinical Outcome after Sequential Checkpoint Blockade in Melanoma. Cancer Immunol Res 2019; 7:458-465. [PMID: 30635271 DOI: 10.1158/2326-6066.cir-18-0226] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 09/25/2018] [Accepted: 01/04/2019] [Indexed: 12/26/2022]
Abstract
To understand prognostic factors for outcome between differentially sequenced nivolumab and ipilimumab in a randomized phase II trial, we measured T-cell infiltration and PD-L1 by IHC, T-cell repertoire metrics, and mutational load within the tumor. We used next-generation sequencing (NGS) and assessed the association of those parameters with response and overall survival. Immunosequencing of the T-cell receptor β-chain locus (TCRβ) from DNA of 91 pretreatment tumor samples and an additional 22 pairs of matched pre- and posttreatment samples from patients who received nivolumab followed by ipilimumab (nivo/ipi), or the reverse (ipi/nivo), was performed to measure T-cell clonality and fraction. Mutational and neoantigen load were also assessed by NGS in 82 of the 91 patients. Tumors were stained using IHC for PD-L1+ and CD8+ T cells. Pretreatment tumor TCR clonality and neoantigen load were marginally associated with best response with nivo/ipi (P = 0.04 and 0.05, respectively), but not with ipi/nivo. Amalgamated pretreatment mutational load and tumor T-cell fraction were significantly associated with best response with nivo/ipi (P = 0.002). Pretreatment PD-L1 staining intensity and CD8+ T-cell counts were correlated with T-cell fraction and clonality, but not mutational or neoantigen load. Patients with increased T-cell fraction posttreatment at week 13 had a 30-fold increased likelihood of survival (P = 0.002). Mutational and neoantigen load, and T-cell infiltrate within the tumor, were associated with outcome of sequential checkpoint inhibition using nivolumab then ipilimumab, but not when ipilimumab was administered before nivolumab.
Collapse
Affiliation(s)
- Erik Yusko
- Adaptive Biotechnologies, Seattle, Washington
| | | | - Richard K Wilson
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Elaine R Mardis
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | | | | | - Han Chang
- Bristol-Myers Squibb, Princeton, New Jersey
| | - David M Woods
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Medical Center, New York, New York
| | - Harlan Robins
- Adaptive Biotechnologies, Seattle, Washington.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jeffrey Weber
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Medical Center, New York, New York.
| |
Collapse
|
230
|
Sun W, Lee J, Zhang S, Benyshek C, Dokmeci MR, Khademhosseini A. Engineering Precision Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801039. [PMID: 30643715 PMCID: PMC6325626 DOI: 10.1002/advs.201801039] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/10/2018] [Indexed: 05/18/2023]
Abstract
Advances in genomic sequencing and bioinformatics have led to the prospect of precision medicine where therapeutics can be advised by the genetic background of individuals. For example, mapping cancer genomics has revealed numerous genes that affect the therapeutic outcome of a drug. Through materials and cell engineering, many opportunities exist for engineers to contribute to precision medicine, such as engineering biosensors for diagnosis and health status monitoring, developing smart formulations for the controlled release of drugs, programming immune cells for targeted cancer therapy, differentiating pluripotent stem cells into desired lineages, fabricating bioscaffolds that support cell growth, or constructing "organs-on-chips" that can screen the effects of drugs. Collective engineering efforts will help transform precision medicine into a more personalized and effective healthcare approach. As continuous progress is made in engineering techniques, more tools will be available to fully realize precision medicine's potential.
Collapse
Affiliation(s)
- Wujin Sun
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Junmin Lee
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Shiming Zhang
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Cole Benyshek
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Mehmet R. Dokmeci
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
- Department of RadiologyUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Ali Khademhosseini
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
- Department of RadiologyUniversity of California–Los AngelesLos AngelesCA90095USA
- Jonsson Comprehensive Cancer CenterUniversity of California–Los Angeles10833 Le Conte AveLos AngelesCA90024USA
- Department of Chemical and Biomolecular EngineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center of NanotechnologyDepartment of PhysicsKing Abdulaziz UniversityJeddah21569Saudi Arabia
- Department of Bioindustrial TechnologiesCollege of Animal Bioscience and TechnologyKonkuk UniversitySeoul05029Republic of Korea
| |
Collapse
|
231
|
Depil S, Bonaventura P, Alcazer V, Tonon L. Nouvelles approches vaccinales en cancérologie. Bull Cancer 2019; 105 Suppl 1:S113-S120. [PMID: 30595192 DOI: 10.1016/s0007-4551(18)30396-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
THE NEW VACCINE APPROACHES IN ONCOLOGY The concept of cancer vaccine relies on the induction of an adaptive immune response against antigens presented by tumor cells, in order to allow a long-term control of the tumor. First generations of cancer vaccines have been associated with relatively disappointing clinical results. The therapeutic breakthrough provided by the immune checkpoint inhibitors targeting PD-1/PD-L1, and the demonstration of the key role played by mutation-associated neoantigens in anti-tumor T cell response, have led to a renaissance of cancer vaccination. This next generation of cancer vaccines is thus based on the use of mutated neoantigens specific to each tumor (personalized approach). Preclinical results have shown that these neoantigens are able to induce potent antitumor immune responses, explained by the absence of thymic selection against the reactive T cells. Several clinical trials are ongoing, with promising preliminary results. Nevertheless, the demonstration of an anti-tumor effect remains to be done in the clinic. The optimization of neoantigen selection process is still a major challenge. Finally, cancer vaccines may either reinforce the activity of anti-PD-1/PD-L1, by increasing the number of effector T cells in the tumor, or induce an adaptive T cell response in non-T cell infiltrated "cold tumors". Vaccination may also have a strong interest in an adjuvant setting to allow a long-term control of the residual disease.
Collapse
Affiliation(s)
- Stéphane Depil
- Centre Léon-Bérard, 28, Prom. Léa-et-Napoléon-Bullukian, 69008 Lyon, France; Université Claude-Bernard-Lyon-1, 43, boulevard du 11-Novembre-1918, 69100 Villeurbanne, France.
| | - Paola Bonaventura
- Centre Léon-Bérard, 28, Prom. Léa-et-Napoléon-Bullukian, 69008 Lyon, France
| | - Vincent Alcazer
- Centre Léon-Bérard, 28, Prom. Léa-et-Napoléon-Bullukian, 69008 Lyon, France
| | - Laurie Tonon
- Fondation Synergie-Lyon-Cancer, 28, rue Laennec, 69008 Lyon, France
| |
Collapse
|
232
|
Kalaora S, Samuels Y. Cancer Exome-Based Identification of Tumor Neo-Antigens Using Mass Spectrometry. Methods Mol Biol 2019; 1884:203-214. [PMID: 30465205 DOI: 10.1007/978-1-4939-8885-3_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neo-antigens expressed on tumors are targets for development of cancer immunotherapy strategies. Use of prediction algorithms to identify neo-antigens yields a significant number of peptides that must be validated in laborious and time-consuming methods; many prove to be false-positive identifications. The use of HLA peptidomics allows the isolation of the HLA-peptide complexes directly from cells and can be done on fresh tumor, patient-derived xerographs, or cell lines when the tissue sample is limited. This method can be used to identify both HLA class I and HLA class II or any different MHC from different species. Here we describe the steps to create the immune-affinity columns used from the process, the immunoprecipitation procedure, and also the isolation of the peptides that will be analyzed by mass spectrometry.
Collapse
Affiliation(s)
- Shelly Kalaora
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Yardena Samuels
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
233
|
Dahl O, Brydøy M. The pioneers behind immune checkpoint blockers awarded the Nobel Prize in physiology or medicine 2018. Acta Oncol 2019; 58:1-8. [PMID: 30698061 DOI: 10.1080/0284186x.2018.1555375] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Olav Dahl
- Department of Clinical Science Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Marianne Brydøy
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
234
|
Gwin WR, Disis ML, Ruiz-Garcia E. Immuno-Oncology in the Era of Personalized Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1168:117-129. [PMID: 31713168 DOI: 10.1007/978-3-030-24100-1_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Personalized medicine in oncology utilizes evidence derived from genetic, immune, and proteomic profiling to inform therapeutic options as well as provide prognostic information for each unique individual and their tumor. Our ability to biologically and immunologically define each patient's tumor has been driven by the development of assays characterizing the genomic and proteomic profiles of tumors that in turn have led to the development of large biologic databases and computational tools for the analysis of these large data sets. In Immuno-oncology, the introduction of checkpoint inhibitors and their approval across multiple tumor types has led to the recognition that the majority of patients will not clinically respond to these therapies but will remain at risk for the development of significant immunologic side effects. This challenge highlights the need for the development and validation of both predictive biomarkers for response to such therapies as well as biomarkers prognostic of disease course. Despite extensive investigation into predictive biomarkers using these biologic databases and computational methods, only recently has progress been made in this area. This progress is the first step allowing us to identify patients likely to benefit from these therapies and moving our field closer to a truly personalized approach to the use of immune therapies in oncology.
Collapse
Affiliation(s)
- William R Gwin
- Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| | - Mary L Disis
- Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| | - Erika Ruiz-Garcia
- Department of Gastrointestinal Medical Oncology & Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City, Mexico.
| |
Collapse
|
235
|
Wolf Y, Samuels Y. Cancer research in the era of immunogenomics. ESMO Open 2018; 3:e000475. [PMID: 30622743 PMCID: PMC6307593 DOI: 10.1136/esmoopen-2018-000475] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 12/14/2022] Open
Abstract
The most meaningful advancement in cancer treatment in recent years has been the emergence of immunotherapy. Checkpoint inhibitor blockade and adoptive T cell therapy have shown remarkable clinical effects in a wide range of tumour types. Despite these advances, many tumours do not respond to these treatments, which raises the need to further investigate how patients can benefit from immunotherapy. This effort can now take advantage of the recent technological progress in single-cell, high-throughput sequencing and computational efforts. In this review, we will discuss advances in different immunotherapies and the principles of cancer immunogenomics, with an emphasis on the detection of cancer neoantigens with human leucocyte antigen peptidomics, and how these principles can be further used for more efficient clinical output.
Collapse
Affiliation(s)
- Yochai Wolf
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
236
|
Recent Development and Clinical Application of Cancer Vaccine: Targeting Neoantigens. J Immunol Res 2018; 2018:4325874. [PMID: 30662919 PMCID: PMC6313977 DOI: 10.1155/2018/4325874] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/18/2018] [Indexed: 12/30/2022] Open
Abstract
Recently, increasing data show that immunotherapy could be a powerful weapon against cancers. Comparing to the traditional surgery, chemotherapy or radiotherapy, immunotherapy more specifically targets cancer cells, giving rise to the opportunities to the patients to have higher response rates and better quality of life and even to cure the disease. Cancer vaccines could be designed to target tumor-associated antigens (TAAs), cancer germline antigens, virus-associated antigens, or tumor-specific antigens (TSAs), which are also called neoantigens. The cancer vaccines could be cell-based (e.g., dendritic cell vaccine provenge (sipuleucel-T) targeting prostatic acid phosphatase for metastatic prostate cancer), peptide/protein-based, or gene- (DNA/RNA) based, with the different kinds of adjuvants. Neoantigens are tumor-specific and could be presented by MHC molecules and recognized by T lymphocytes, serving the ideal immune targets to increase the therapeutic specificity and decrease the risk of nonspecific autoimmunity. By targeting the shared antigens and private epitopes, the cancer vaccine has potential to treat the disease. Accordingly, personalized neoantigen-based immunotherapies are emerging. In this article, we review the literature and evidence of the advantage and application of cancer vaccine. We summarize the recent clinical trials of neoantigen cancer vaccines which were designed according to the patients' personal mutanome. With the rapid development of personalized immunotherapy, it is believed that tumors could be efficiently controlled and become curable in the new era of precision medicine.
Collapse
|
237
|
Bulik-Sullivan B, Busby J, Palmer CD, Davis MJ, Murphy T, Clark A, Busby M, Duke F, Yang A, Young L, Ojo NC, Caldwell K, Abhyankar J, Boucher T, Hart MG, Makarov V, Montpreville VTD, Mercier O, Chan TA, Scagliotti G, Bironzo P, Novello S, Karachaliou N, Rosell R, Anderson I, Gabrail N, Hrom J, Limvarapuss C, Choquette K, Spira A, Rousseau R, Voong C, Rizvi NA, Fadel E, Frattini M, Jooss K, Skoberne M, Francis J, Yelensky R. Deep learning using tumor HLA peptide mass spectrometry datasets improves neoantigen identification. Nat Biotechnol 2018; 37:nbt.4313. [PMID: 30556813 DOI: 10.1038/nbt.4313] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 11/06/2018] [Indexed: 12/30/2022]
Abstract
Neoantigens, which are expressed on tumor cells, are one of the main targets of an effective antitumor T-cell response. Cancer immunotherapies to target neoantigens are of growing interest and are in early human trials, but methods to identify neoantigens either require invasive or difficult-to-obtain clinical specimens, require the screening of hundreds to thousands of synthetic peptides or tandem minigenes, or are only relevant to specific human leukocyte antigen (HLA) alleles. We apply deep learning to a large (N = 74 patients) HLA peptide and genomic dataset from various human tumors to create a computational model of antigen presentation for neoantigen prediction. We show that our model, named EDGE, increases the positive predictive value of HLA antigen prediction by up to ninefold. We apply EDGE to enable identification of neoantigens and neoantigen-reactive T cells using routine clinical specimens and small numbers of synthetic peptides for most common HLA alleles. EDGE could enable an improved ability to develop neoantigen-targeted immunotherapies for cancer patients.
Collapse
Affiliation(s)
| | - Jennifer Busby
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Christine D Palmer
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Matthew J Davis
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Tyler Murphy
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Andrew Clark
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Michele Busby
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Fujiko Duke
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Aaron Yang
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Lauren Young
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Noelle C Ojo
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Kamilah Caldwell
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Jesse Abhyankar
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Thomas Boucher
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Meghan G Hart
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | | | | | - Olaf Mercier
- Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France
| | - Timothy A Chan
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Giorgio Scagliotti
- University of Turin, Department of Oncology at San Luigi Hospital, Orbassano (Turin), Italy
| | - Paolo Bironzo
- University of Turin, Department of Oncology at San Luigi Hospital, Orbassano (Turin), Italy
| | - Silvia Novello
- University of Turin, Department of Oncology at San Luigi Hospital, Orbassano (Turin), Italy
| | - Niki Karachaliou
- Instituto Oncologico Dr. Rosell - Hospital Universitari Quiron Dexeus Location, Barcelona, Spain
| | | | - Ian Anderson
- St Joseph Heritage Healthcare, Santa Rosa, California, USA
| | | | - John Hrom
- Hattiesburg Clinic/Forrest General Cancer Center, Hattiesburg, Mississippi, USA
| | | | | | | | - Raphael Rousseau
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Cynthia Voong
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Naiyer A Rizvi
- New York Presbyterian/Columbia University Medical Center, New York, New York, USA
| | - Elie Fadel
- Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France
| | - Mark Frattini
- New York Presbyterian/Columbia University Medical Center, New York, New York, USA
| | - Karin Jooss
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Mojca Skoberne
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Joshua Francis
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| | - Roman Yelensky
- Gritstone Oncology, Inc., Emeryville, California and Cambridge, Massachusetts, USA
| |
Collapse
|
238
|
Abstract
Malignant transformation of cells depends on accumulation of DNA damage. Over the past years we have learned that the T cell-based immune system frequently responds to the neoantigens that arise as a consequence of this DNA damage. Furthermore, recognition of neoantigens appears an important driver of the clinical activity of both T cell checkpoint blockade and adoptive T cell therapy as cancer immunotherapies. Here we review the evidence for the relevance of cancer neoantigens in tumor control and the biological properties of these antigens. We discuss recent technological advances utilized to identify neoantigens, and the T cells that recognize them, in individual patients. Finally, we discuss strategies that can be employed to exploit cancer neoantigens in clinical interventions.
Collapse
Affiliation(s)
- Ton N Schumacher
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; , .,Oncode Institute, 3521AL Utrecht, The Netherlands
| | - Wouter Scheper
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; , .,Oncode Institute, 3521AL Utrecht, The Netherlands
| | - Pia Kvistborg
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; ,
| |
Collapse
|
239
|
Vikas P, Borcherding N, Zhang W. The clinical promise of immunotherapy in triple-negative breast cancer. Cancer Manag Res 2018; 10:6823-6833. [PMID: 30573992 PMCID: PMC6292225 DOI: 10.2147/cmar.s185176] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease with poorer outcomes compared to other breast cancer subtypes. Contributing to the worse prognosis in TNBC is the higher rates of relapse and rapid progression after relapse. Advances in targeted therapeutics and conventional chemotherapy for TNBC have been stymied due to the lack of specific targets. Moreover, the responses to chemotherapy in TNBC lack durability, partially accounting for the higher rates of relapse. Immunotherapy, notably immune-checkpoint blockade, has shown to improve survival and maintain robust antitumor responses in both hematologic and solid malignancies. Unlike lung cancer, melanoma, and bladder cancer, most breast cancers are not inherently immunogenic and typically have low T cell infiltration. However, among breast cancer subtypes, TNBC is characterized by greater tumor immune infiltrate and higher degree of stromal and intratumoral tumor-infiltrating lymphocytes (TILs), a predictive marker for responses to immunotherapy. Moreover, in TNBC, the high number of stromal TILs is predictive of more favorable survival outcomes and response to chemotherapy. Immunotherapy is being extensively explored in TNBC and clinical trials are showing some promising results. This article focuses on the rationale for immunotherapy in TNBC, to explore and discuss preclinical data, results from early clinical trials, and to summarize some ongoing trials. We will also discuss the potential application of immunotherapy in TNBC from a clinician's perspective.
Collapse
Affiliation(s)
- Praveen Vikas
- Department of Internal Medicine, College of Medicine, University of Iowa, Iowa City, IA, USA,
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA,
| | - Nicholas Borcherding
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA,
- Department of Pathology, College of Medicine, University of Iowa, Iowa City, IA, USA
- Cancer Biology Graduate Program, College of Medicine, University of Iowa, Iowa City, IA, USA
- Medical Scientist Training Program, College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Weizhou Zhang
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA,
- Department of Pathology, College of Medicine, University of Iowa, Iowa City, IA, USA
- Cancer Biology Graduate Program, College of Medicine, University of Iowa, Iowa City, IA, USA
- Medical Scientist Training Program, College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
240
|
Grimm J, Bill M, Jentzsch M, Beinicke S, Häntschel J, Goldmann K, Schulz J, Cross M, Franke G, Behre G, Vucinic V, Pönisch W, Lange T, Niederwieser D, Schwind S. Clinical impact of clonal hematopoiesis in acute myeloid leukemia patients receiving allogeneic transplantation. Bone Marrow Transplant 2018; 54:1189-1197. [DOI: 10.1038/s41409-018-0413-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 12/21/2022]
|
241
|
Blaha DT, Anderson SD, Yoakum DM, Hager MV, Zha Y, Gajewski TF, Kranz DM. High-Throughput Stability Screening of Neoantigen/HLA Complexes Improves Immunogenicity Predictions. Cancer Immunol Res 2018; 7:50-61. [PMID: 30425106 DOI: 10.1158/2326-6066.cir-18-0395] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/31/2018] [Accepted: 11/09/2018] [Indexed: 12/30/2022]
Abstract
Mutated peptides (neoantigens) from a patient's cancer genome can serve as targets for T-cell immunity, but identifying which peptides can be presented by an MHC molecule and elicit T cells has been difficult. Although algorithms that predict MHC binding exist, they are not yet able to distinguish experimental differences in half-lives of the complexes (an immunologically relevant parameter, referred to here as kinetic stability). Improvement in determining actual neoantigen peptide/MHC stability could be important, as only a small fraction of peptides in most current vaccines are capable of eliciting CD8+ T-cell responses. Here, we used a rapid, high-throughput method to experimentally determine peptide/HLA thermal stability on a scale that will be necessary for analysis of neoantigens from thousands of patients. The method combined the use of UV-cleavable peptide/HLA class I complexes and differential scanning fluorimetry to determine the Tm values of neoantigen complexes. Measured Tm values were accurate and reproducible and were directly proportional to the half-lives of the complexes. Analysis of known HLA-A2-restricted immunogenic peptides showed that Tm values better correlated with immunogenicity than algorithm-predicted binding affinities. We propose that temperature stability information can be used as a guide for the selection of neoantigens in cancer vaccines in order to focus attention on those mutated peptides with the highest probability of being expressed on the cell surface.
Collapse
Affiliation(s)
- Dylan T Blaha
- Department of Biochemistry, University of Illinois, Urbana, Illinois
| | - Scott D Anderson
- Department of Biochemistry, University of Illinois, Urbana, Illinois
| | - Daniel M Yoakum
- Department of Biochemistry, University of Illinois, Urbana, Illinois
| | - Marlies V Hager
- Department of Biochemistry, University of Illinois, Urbana, Illinois
| | - Yuanyuan Zha
- Department of Pathology, Department of Medicine, and the Ben May Department of Cancer, University of Chicago, Chicago, Illinois
| | - Thomas F Gajewski
- Department of Pathology, Department of Medicine, and the Ben May Department of Cancer, University of Chicago, Chicago, Illinois
| | - David M Kranz
- Department of Biochemistry, University of Illinois, Urbana, Illinois.
| |
Collapse
|
242
|
Zhang SQ, Ma KY, Schonnesen AA, Zhang M, He C, Sun E, Williams CM, Jia W, Jiang N. High-throughput determination of the antigen specificities of T cell receptors in single cells. Nat Biotechnol 2018; 36:nbt.4282. [PMID: 30418433 PMCID: PMC6728224 DOI: 10.1038/nbt.4282] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 09/26/2018] [Indexed: 01/07/2023]
Abstract
We present tetramer-associated T-cell receptor sequencing (TetTCR-seq) to link T cell receptor (TCR) sequences to their cognate antigens in single cells at high throughput. Binding is determined using a library of DNA-barcoded antigen tetramers that is rapidly generated by in vitro transcription and translation. We applied TetTCR-seq to identify patterns in TCR cross-reactivity with cancer neoantigens and to rapidly isolate neoantigen-specific TCRs with no cross-reactivity to the wild-type antigen.
Collapse
Affiliation(s)
- Shu-Qi Zhang
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Ke-Yue Ma
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | | | - Mingliang Zhang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai 200233, China
| | - Chenfeng He
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Eric Sun
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Chad M. Williams
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai 200233, China
| | - Ning Jiang
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
- LIVESTRONG Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
243
|
Systematically benchmarking peptide-MHC binding predictors: From synthetic to naturally processed epitopes. PLoS Comput Biol 2018; 14:e1006457. [PMID: 30408041 PMCID: PMC6224037 DOI: 10.1371/journal.pcbi.1006457] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 08/22/2018] [Indexed: 12/19/2022] Open
Abstract
A number of machine learning-based predictors have been developed for identifying immunogenic T-cell epitopes based on major histocompatibility complex (MHC) class I and II binding affinities. Rationally selecting the most appropriate tool has been complicated by the evolving training data and machine learning methods. Despite the recent advances made in generating high-quality MHC-eluted, naturally processed ligandome, the reliability of new predictors on these epitopes has yet to be evaluated. This study reports the latest benchmarking on an extensive set of MHC-binding predictors by using newly available, untested data of both synthetic and naturally processed epitopes. 32 human leukocyte antigen (HLA) class I and 24 HLA class II alleles are included in the blind test set. Artificial neural network (ANN)-based approaches demonstrated better performance than regression-based machine learning and structural modeling. Among the 18 predictors benchmarked, ANN-based mhcflurry and nn_align perform the best for MHC class I 9-mer and class II 15-mer predictions, respectively, on binding/non-binding classification (Area Under Curves = 0.911). NetMHCpan4 also demonstrated comparable predictive power. Our customization of mhcflurry to a pan-HLA predictor has achieved similar accuracy to NetMHCpan. The overall accuracy of these methods are comparable between 9-mer and 10-mer testing data. However, the top methods deliver low correlations between the predicted versus the experimental affinities for strong MHC binders. When used on naturally processed MHC-ligands, tools that have been trained on elution data (NetMHCpan4 and MixMHCpred) shows better accuracy than pure binding affinity predictor. The variability of false prediction rate is considerable among HLA types and datasets. Finally, structure-based predictor of Rosetta FlexPepDock is less optimal compared to the machine learning approaches. With our benchmarking of MHC-binding and MHC-elution predictors using a comprehensive metrics, a unbiased view for establishing best practice of T-cell epitope predictions is presented, facilitating future development of methods in immunogenomics.
Collapse
|
244
|
Bethune MT, Li XH, Yu J, McLaughlin J, Cheng D, Mathis C, Moreno BH, Woods K, Knights AJ, Garcia-Diaz A, Wong S, Hu-Lieskovan S, Puig-Saus C, Cebon J, Ribas A, Yang L, Witte ON, Baltimore D. Isolation and characterization of NY-ESO-1-specific T cell receptors restricted on various MHC molecules. Proc Natl Acad Sci U S A 2018; 115:E10702-E10711. [PMID: 30348802 PMCID: PMC6233129 DOI: 10.1073/pnas.1810653115] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tumor-specific T cell receptor (TCR) gene transfer enables specific and potent immune targeting of tumor antigens. Due to the prevalence of the HLA-A2 MHC class I supertype in most human populations, the majority of TCR gene therapy trials targeting public antigens have employed HLA-A2-restricted TCRs, limiting this approach to those patients expressing this allele. For these patients, TCR gene therapy trials have resulted in both tantalizing successes and lethal adverse events, underscoring the need for careful selection of antigenic targets. Broad and safe application of public antigen-targeted TCR gene therapies will require (i) selecting public antigens that are highly tumor-specific and (ii) targeting multiple epitopes derived from these antigens by obtaining an assortment of TCRs restricted by multiple common MHC alleles. The canonical cancer-testis antigen, NY-ESO-1, is not expressed in normal tissues but is aberrantly expressed across a broad array of cancer types. It has also been targeted with A2-restricted TCR gene therapy without adverse events or notable side effects. To enable the targeting of NY-ESO-1 in a broader array of HLA haplotypes, we isolated TCRs specific for NY-ESO-1 epitopes presented by four MHC molecules: HLA-A2, -B07, -B18, and -C03. Using these TCRs, we pilot an approach to extend TCR gene therapies targeting NY-ESO-1 to patient populations beyond those expressing HLA-A2.
Collapse
Affiliation(s)
- Michael T Bethune
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Xiao-Hua Li
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Jiaji Yu
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095
- Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Jami McLaughlin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Donghui Cheng
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Colleen Mathis
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Blanca Homet Moreno
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, CA 90095
| | - Katherine Woods
- Cancer Immunobiology Laboratory, Olivia Newton-John Cancer Research Institute, Austin Hospital, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- Cancer Immunobiology Laboratory, Ludwig Institute for Cancer Research, Heidelberg, VIC 3084, Australia
| | - Ashley J Knights
- Cancer Immunobiology Laboratory, Ludwig Institute for Cancer Research, Heidelberg, VIC 3084, Australia
| | - Angel Garcia-Diaz
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, CA 90095
| | - Stephanie Wong
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Siwen Hu-Lieskovan
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, CA 90095
| | - Cristina Puig-Saus
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, CA 90095
| | - Jonathan Cebon
- Cancer Immunobiology Laboratory, Olivia Newton-John Cancer Research Institute, Austin Hospital, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- Cancer Immunobiology Laboratory, Ludwig Institute for Cancer Research, Heidelberg, VIC 3084, Australia
| | - Antoni Ribas
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, CA 90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Department of Medicine, University of California, Los Angeles, CA 90095
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, CA 90095
| | - Lili Yang
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095;
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Owen N Witte
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095;
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, CA 90095
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125;
| |
Collapse
|
245
|
Finn OJ, Rammensee HG. Is It Possible to Develop Cancer Vaccines to Neoantigens, What Are the Major Challenges, and How Can These Be Overcome? Neoantigens: Nothing New in Spite of the Name. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028829. [PMID: 29254980 DOI: 10.1101/cshperspect.a028829] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The term "neoantigen," as applied to molecules newly expressed on tumor cells, has a long history. The groundbreaking discovery of a cancer causing virus in chickens by Rous over 100 years ago, followed by discoveries of other tumor-causing viruses in animals, suggested a viral etiology of human cancers. The search for other oncogenic viruses in the 1960s and 1970s resulted in the discoveries of Epstein-Barr virus (EBV), hepatitis B virus (HBV), and human papilloma virus (HPV), and continues until the present time. Contemporaneously, the budding field of immunology was posing the question can the immune system of animals or humans recognize a tumor that develops from one's own tissues and what types of antigens would distinguish the tumor from normal cells. Molecules encoded by oncogenic viruses provided the most logical candidates and evidence was quickly gathered for both humoral and cellular recognition of viral antigens, referred to as neoantigens. Often, however, serologic responses to virus-bearing tumors revealed neoantigens unrelated to viral proteins and expressed on multiple tumor types, foreshadowing later findings of multiple changes in other genes in tumor cells creating nonviral neoantigens.
Collapse
Affiliation(s)
- Olivera J Finn
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tuebingen, 72074 Tuebingen; and German Cancer Consortium, DKFZ Partner Site, D-69120 Heidelberg, Germany
| |
Collapse
|
246
|
Kissick HT. Is It Possible to Develop Cancer Vaccines to Neoantigens, What Are the Major Challenges, and How Can These Be Overcome? Neoantigens as Vaccine Targets for Cancer. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a033704. [PMID: 29254981 DOI: 10.1101/cshperspect.a033704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent work by several groups has undoubtedly shown that we can produce cancer vaccines targeting neoantigens. However, each vaccine is essentially a single-use, patient-specific product, making this approach resource-intensive. For this reason, it is important to ask whether this approach will be any more successful than what has been attempted during the last 30 years using vaccines targeting self-epitopes. Here, we discuss what might be expected from neoantigen vaccines based on our experience in chronic viral infections, and how this new approach may be applied to cancer immunotherapy.
Collapse
Affiliation(s)
- Haydn T Kissick
- Departments of Urology and Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322
| |
Collapse
|
247
|
Hammerl D, Rieder D, Martens JWM, Trajanoski Z, Debets R. Adoptive T Cell Therapy: New Avenues Leading to Safe Targets and Powerful Allies. Trends Immunol 2018; 39:921-936. [PMID: 30309702 DOI: 10.1016/j.it.2018.09.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/12/2018] [Accepted: 09/12/2018] [Indexed: 12/30/2022]
Abstract
Adoptive transfer of TCR-engineered T cells is a potent therapy, able to induce clinical responses in different human malignancies. Nevertheless, treatment toxicities may occur and, in particular for solid tumors, responses may be variable and often not durable. To address these challenges, it is imperative to carefully select target antigens and to immunologically interrogate the corresponding tumors when designing optimal T cell therapies. Here, we review recent advances, covering both omics- and laboratory tools that can enable the selection of optimal T cell epitopes and TCRs as well as the identification of dominant immune evasive mechanisms within tumor tissues. Furthermore, we discuss how these techniques may aid in a rational design of effective combinatorial adoptive T cell therapies.
Collapse
Affiliation(s)
- Dora Hammerl
- Laboratory of Tumor Immunology, Erasmus MC-Cancer Institute, Rotterdam, The Netherlands
| | - Dietmar Rieder
- Division of Bioinformatics, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC-Cancer Institute, Rotterdam, The Netherlands
| | - Zlatko Trajanoski
- Division of Bioinformatics, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Reno Debets
- Laboratory of Tumor Immunology, Erasmus MC-Cancer Institute, Rotterdam, The Netherlands.
| |
Collapse
|
248
|
Want MY, Lugade AA, Battaglia S, Odunsi K. Nature of tumour rejection antigens in ovarian cancer. Immunology 2018; 155:202-210. [PMID: 29772069 PMCID: PMC6142289 DOI: 10.1111/imm.12951] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/12/2018] [Accepted: 04/19/2018] [Indexed: 12/16/2022] Open
Abstract
Major progress in the analysis of human immune responses to cancer has been made through the molecular characterization of human tumour antigens. The development of therapeutic strategies for eliciting immune-mediated rejection of tumours has accelerated due to the elucidation of the molecular basis for tumour cell recognition and destruction by immune cells. Of the various human tumour antigens defined to date in ovarian cancer, the cancer-testis (CT) family of antigens have been studied extensively preclinically and clinically because of their testis-restricted expression in normal tissues and ability to elicit robust immune responses. Recent developments in cancer sequencing technologies offer a unique opportunity to identify tumour mutations with the highest likelihood of being expressed and recognized by the immune system. Such mutations, or neoantigens, could potentially serve as specific immune targets for T-cell-mediated destruction of cancer cells. This review will highlight current work in selecting tumour rejection antigens in ovarian cancer for improving the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Muzamil Y. Want
- Center For ImmunotherapyRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | - Amit A. Lugade
- Center For ImmunotherapyRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | | | - Kunle Odunsi
- Center For ImmunotherapyRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| |
Collapse
|
249
|
Abstract
This review discusses the rapidly evolving field of immunotherapy research, focusing on the types of cancer antigens that can be recognised by the immune system and potential methods by which neoantigens can be exploited clinically to successfully target and clear tumour cells. Recent studies suggest that the likelihood of successful immunotherapeutic targeting of cancer will be reliant on immune response to neoantigens. This type of cancer-specific antigen arises from somatic variants that result in alteration of the expressed protein sequence. Massively parallel sequencing techniques now allow the rapid identification of these genomic mutations, and algorithms can be used to predict those that will be processed by the proteasome, bind to the transporter complex and encode peptides that bind strongly to individual MHC molecules. The emerging data from assessment of the immunogenicity of neoantigens suggests that only a minority of mutations will form targetable epitopes and therefore the potential for immunotherapeutic targeting will be greater in cancers with a higher frequency of protein-altering somatic variants. It is evident that neoantigens contribute to the success of some immunotherapeutic interventions and that there is significant scope for specific targeting of these antigens to develop new treatment approaches.
Collapse
Affiliation(s)
- Antonia L Pritchard
- Genetics and Immunology Research Group, An Lòchran, 10 Inverness Campus, Inverness, IV2 5NA, Scotland, UK.
| |
Collapse
|
250
|
van Belzen IAEM, Kesmir C. Immune biomarkers for predicting response to adoptive cell transfer as cancer treatment. Immunogenetics 2018; 71:71-86. [PMID: 30232514 PMCID: PMC6326979 DOI: 10.1007/s00251-018-1083-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 08/28/2018] [Indexed: 12/20/2022]
Abstract
Adoptive cell transfer (ACT) is a form of personalised immunotherapy which has shown promising results in metastasised cancer. For this treatment, autologous T lymphocytes are selected and stimulated in vitro before re-administration in large numbers. However, only a fraction of patients benefit from ACT, and it is not yet known what biomarkers can predict treatment outcome. In this review, we describe what tumour characteristics are associated with response to ACT. Based on the current knowledge, the best candidate biomarker for a good anti-tumour response seems to be a large number of neoantigens with a homogeneous distribution across the tumour in combination with sufficient MHC-I expression level. Additionally, it is necessary to be able to isolate a diverse population of T cells reactive to these neoantigens from tumour tissue or peripheral blood. Additional promising candidate biomarkers shared with other cancer immunotherapies are a large number of tumour-infiltrating cytotoxic and memory T cells, normal levels of glycolysis, and a pro-inflammatory cytokine profile within the tumour. Intense research in this field will hopefully result in identification of more biomarkers for cancers with low mutational load.
Collapse
Affiliation(s)
- Ianthe A E M van Belzen
- Theoretical Biology and Bioinformatics, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - Can Kesmir
- Theoretical Biology and Bioinformatics, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| |
Collapse
|