251
|
Huang D, Chen J, Yang L, Ouyang Q, Li J, Lao L, Zhao J, Liu J, Lu Y, Xing Y, Chen F, Su F, Yao H, Liu Q, Su S, Song E. NKILA lncRNA promotes tumor immune evasion by sensitizing T cells to activation-induced cell death. Nat Immunol 2018; 19:1112-1125. [PMID: 30224822 DOI: 10.1038/s41590-018-0207-y] [Citation(s) in RCA: 328] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 08/07/2018] [Indexed: 12/29/2022]
Abstract
Activation-induced cell death (AICD) of T lymphocytes can be exploited by cancers to escape immunological destruction. We demonstrated that tumor-specific cytotoxic T lymphocytes (CTLs) and type 1 helper T (TH1) cells, rather than type 2 helper T cells and regulatory T cells, were sensitive to AICD in breast and lung cancer microenvironments. NKILA, an NF-κB-interacting long noncoding RNA (lncRNA), regulates T cell sensitivity to AICD by inhibiting NF-κB activity. Mechanistically, calcium influx in stimulated T cells via T cell-receptor signaling activates calmodulin, thereby removing deacetylase from the NKILA promoter and enhancing STAT1-mediated transcription. Administering CTLs with NKILA knockdown effectively inhibited growth of breast cancer patient-derived xenografts in mice by increasing CTL infiltration. Clinically, NKILA overexpression in tumor-specific CTLs and TH1 cells correlated with their apoptosis and shorter patient survival. Our findings underscore the importance of lncRNAs in determining tumor-mediated T cell AICD and suggest that engineering lncRNAs in adoptively transferred T cells might provide a novel antitumor immunotherapy.
Collapse
Affiliation(s)
- Di Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Linbin Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qian Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiaqian Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Liyan Lao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jinghua Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yiwen Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yue Xing
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fei Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fengxi Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Herui Yao
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shicheng Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China. .,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China. .,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China. .,Program in Molecular Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
252
|
Kalaora S, Wolf Y, Feferman T, Barnea E, Greenstein E, Reshef D, Tirosh I, Reuben A, Patkar S, Levy R, Quinkhardt J, Omokoko T, Qutob N, Golani O, Zhang J, Mao X, Song X, Bernatchez C, Haymaker C, Forget MA, Creasy C, Greenberg P, Carter BW, Cooper ZA, Rosenberg SA, Lotem M, Sahin U, Shakhar G, Ruppin E, Wargo JA, Friedman N, Admon A, Samuels Y. Combined Analysis of Antigen Presentation and T-cell Recognition Reveals Restricted Immune Responses in Melanoma. Cancer Discov 2018; 8:1366-1375. [PMID: 30209080 DOI: 10.1158/2159-8290.cd-17-1418] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/26/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022]
Abstract
The quest for tumor-associated antigens (TAA) and neoantigens is a major focus of cancer immunotherapy. Here, we combine a neoantigen prediction pipeline and human leukocyte antigen (HLA) peptidomics to identify TAAs and neoantigens in 16 tumors derived from seven patients with melanoma and characterize their interactions with their tumor-infiltrating lymphocytes (TIL). Our investigation of the antigenic and T-cell landscapes encompassing the TAA and neoantigen signatures, their immune reactivity, and their corresponding T-cell identities provides the first comprehensive analysis of cancer cell T-cell cosignatures, allowing us to discover remarkable antigenic and TIL similarities between metastases from the same patient. Furthermore, we reveal that two neoantigen-specific clonotypes killed 90% of autologous melanoma cells, both in vitro and in vivo, showing that a limited set of neoantigen-specific T cells may play a central role in melanoma tumor rejection. Our findings indicate that combining HLA peptidomics with neoantigen predictions allows robust identification of targetable neoantigens, which could successfully guide personalized cancer immunotherapies.Significance: As neoantigen targeting is becoming more established as a powerful therapeutic approach, investigating these molecules has taken center stage. Here, we show that a limited set of neoantigen-specific T cells mediates tumor rejection, suggesting that identifying just a few antigens and their corresponding T-cell clones could guide personalized immunotherapy. Cancer Discov; 8(11); 1366-75. ©2018 AACR. This article is highlighted in the In This Issue feature, p. 1333.
Collapse
Affiliation(s)
- Shelly Kalaora
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yochai Wolf
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Feferman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Erez Greenstein
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Dan Reshef
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alexandre Reuben
- Departments of Surgical Oncology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sushant Patkar
- Cancer Data Science Lab, National Cancer Institute, NIH, Rockville, Maryland
| | - Ronen Levy
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Tana Omokoko
- BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Nouar Qutob
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ofra Golani
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Jianhua Zhang
- Departments of Surgical Oncology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xizeng Mao
- Departments of Surgical Oncology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xingzhi Song
- Departments of Surgical Oncology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chantale Bernatchez
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cara Haymaker
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marie-Andrée Forget
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Caitlin Creasy
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Polina Greenberg
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Brett W Carter
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zachary A Cooper
- Departments of Surgical Oncology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Michal Lotem
- Sharett Institute of Oncology, Hadassah Medical School, Jerusalem, Israel
| | - Ugur Sahin
- TRON-Translational Oncology at the University Medical Center of Johannes Gutenberg University GmbH, Mainz, Germany
| | - Guy Shakhar
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eytan Ruppin
- Cancer Data Science Lab, National Cancer Institute, NIH, Rockville, Maryland
| | - Jennifer A Wargo
- Departments of Surgical Oncology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nir Friedman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Arie Admon
- Department of Biology, Technion, Haifa, Israel
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
253
|
Liao M, Chen P, Liao Y, Li J, Yao W, Sun T, Liao W, Su L. Preoperative high-sensitivity C-reactive protein to lymphocyte ratio index plays a vital role in the prognosis of hepatocellular carcinoma after surgical resection. Onco Targets Ther 2018; 11:5591-5600. [PMID: 30237725 PMCID: PMC6135434 DOI: 10.2147/ott.s167857] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background At present, the predictive ability of the prognostic indicator of hepatocellular carcinoma (HCC) is still limited. This study aims to analyze the relationship between the preoperative high-sensitivity C-reactive protein to lymphocyte ratio (HCLR) and the clinicopathologic characteristics of HCC. Patients and methods A total of 229 HCC patients undergoing surgical resection were retrospectively analyzed. The majority of the patients (132/229) had tumors larger than 5 cm, and 45 out of 229 had more than one tumor focus. Receiver operating characteristic curve analysis was used to decide the cutoff value of HCLR. The overall survival (OS) and progression-free survival (PFS) rates were evaluated by adopting the Kaplan–Meier method. Results The cutoff value of HCLR for the best discrimination of HCC prognosis was 1.3 with a sensitivity of 75.5% and a specificity of 71.8%. The area under the receiver operating characteristic curve was 0.791 (95% CI, 0.731–0.840). Preoperative HCLR at a high level (>1.3) was positively correlated with large tumor size, TNM stage, microvascular invasion, and recurrence. The mean OS and PFS in patients with HCLR >1.3 were significantly shorter than in those with HCLR ≤1.3. Univariate and multivariate analyses revealed the HCLR was an independent predictor of OS and PFS. Conclusion HCLR was an important independent predictor of dismal prognosis in HCC patients and can be used as a sensitive indicator for the dynamic monitoring of postoperative patients.
Collapse
Affiliation(s)
- Minjun Liao
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China, , .,Guangxi Medical University, Nanning, Guangxi, People's Republic of China,
| | - Pu Chen
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China, ,
| | - Yan Liao
- Disease Prevention and Control Center of Guilin, Guilin, Guangxi, People's Republic of China
| | - Jun Li
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China, ,
| | - Wenmin Yao
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China, ,
| | - Tian Sun
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China, ,
| | - Weijia Liao
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China, ,
| | - Lili Su
- Department of Clinical Laboratory, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, Guangxi, People's Republic of China
| |
Collapse
|
254
|
Zamora AE, Crawford JC, Thomas PG. Hitting the Target: How T Cells Detect and Eliminate Tumors. THE JOURNAL OF IMMUNOLOGY 2018; 200:392-399. [PMID: 29311380 DOI: 10.4049/jimmunol.1701413] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 11/16/2017] [Indexed: 12/21/2022]
Abstract
The successes of antitumor immuno-based therapies and the application of next-generation sequencing to mutation profiling have produced insights into the specific targets of antitumor T cells. Mutated proteins have tremendous potential as targets for interventions using autologous T cells or engineered cell therapies and may serve as important correlates of efficacy for immunoregulatory interventions including immune checkpoint blockade. As mutated self, tumors present an exceptional case for host immunity, which has primarily evolved in response to foreign pathogens. Tumor Ags' resemblance to self may limit immune recognition, but key features appear to be the same between antipathogen and antitumor responses. Determining which targets will make efficacious Ags and which responses might be elicited therapeutically are key questions for the field. Here we discuss current knowledge on antitumor specificity, the mutations that provide immunogenic targets, and how cross-reactivity and immunodominance may contribute to variation in immune responses among tumor types.
Collapse
Affiliation(s)
- Anthony E Zamora
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| |
Collapse
|
255
|
Koşaloğlu-Yalçın Z, Lanka M, Frentzen A, Logandha Ramamoorthy Premlal A, Sidney J, Vaughan K, Greenbaum J, Robbins P, Gartner J, Sette A, Peters B. Predicting T cell recognition of MHC class I restricted neoepitopes. Oncoimmunology 2018; 7:e1492508. [PMID: 30377561 PMCID: PMC6204999 DOI: 10.1080/2162402x.2018.1492508] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/31/2018] [Accepted: 06/20/2018] [Indexed: 12/17/2022] Open
Abstract
Epitopes that arise from a somatic mutation, also called neoepitopes, are now known to play a key role in cancer immunology and immunotherapy. Recent advances in high-throughput sequencing have made it possible to identify all mutations and thereby all potential neoepitope candidates in an individual cancer. However, most of these neoepitope candidates are not recognized by T cells of cancer patients when tested in vivo or in vitro, meaning they are not immunogenic. Especially in patients with a high mutational load, usually hundreds of potential neoepitopes are detected, highlighting the need to further narrow down this candidate list. In our study, we assembled a dataset of known, naturally processed, immunogenic neoepitopes to dissect the properties that make these neoepitopes immunogenic. The tools to use and thresholds to apply for prioritizing neoepitopes have so far been largely based on experience with epitope identification in other settings such as infectious disease and allergy. Here, we performed a detailed analysis on our dataset of curated immunogenic neoepitopes to establish the appropriate tools and thresholds in the cancer setting. To this end, we evaluated different predictors for parameters that play a role in a neoepitope's immunogenicity and suggest that using binding predictions and length-rescaling yields the best performance in discriminating immunogenic neoepitopes from a background set of mutated peptides. We furthermore show that almost all neoepitopes had strong predicted binding affinities (as expected), but more surprisingly, the corresponding non-mutated peptides had nearly as high affinities. Our results provide a rational basis for parameters in neoepitope filtering approaches that are being commonly used. Abbreviations: SNV: single nucleotide variant; nsSNV: nonsynonymous single nucleotide variant; ROC: receiver operating characteristic; AUC: area under ROC curve; HLA: human leukocyte antigen; MHC: major histocompatibility complex; PD-1: Programmed cell death protein 1; PD-L1 or CTLA-4: cytotoxic T-lymphocyte associated protein 4.
Collapse
Affiliation(s)
- Zeynep Koşaloğlu-Yalçın
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Manasa Lanka
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Angela Frentzen
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | | | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Kerrie Vaughan
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Jason Greenbaum
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Paul Robbins
- Surgery Branch, National Cancer Institute, Bethesda, MD, USA
| | - Jared Gartner
- Surgery Branch, National Cancer Institute, Bethesda, MD, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
256
|
Pogorelyy MV, Fedorova AD, McLaren JE, Ladell K, Bagaev DV, Eliseev AV, Mikelov AI, Koneva AE, Zvyagin IV, Price DA, Chudakov DM, Shugay M. Exploring the pre-immune landscape of antigen-specific T cells. Genome Med 2018; 10:68. [PMID: 30144804 PMCID: PMC6109350 DOI: 10.1186/s13073-018-0577-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/06/2018] [Indexed: 12/26/2022] Open
Abstract
Background Adaptive immune responses to newly encountered pathogens depend on the mobilization of antigen-specific clonotypes from a vastly diverse pool of naive T cells. Using recent advances in immune repertoire sequencing technologies, models of the immune receptor rearrangement process, and a database of annotated T cell receptor (TCR) sequences with known specificities, we explored the baseline frequencies of T cells specific for defined human leukocyte antigen (HLA) class I-restricted epitopes in healthy individuals. Methods We used a database of TCR sequences with known antigen specificities and a probabilistic TCR rearrangement model to estimate the baseline frequencies of TCRs specific to distinct antigens epitopespecificT-cells. We verified our estimates using a publicly available collection of TCR repertoires from healthy individuals. We also interrogated a database of immunogenic and non-immunogenic peptides is used to link baseline T-cell frequencies with epitope immunogenicity. Results Our findings revealed a high degree of variability in the prevalence of T cells specific for different antigens that could be explained by the physicochemical properties of the corresponding HLA class I-bound peptides. The occurrence of certain rearrangements was influenced by ancestry and HLA class I restriction, and umbilical cord blood samples contained higher frequencies of common pathogen-specific TCRs. We also identified a quantitative link between specific T cell frequencies and the immunogenicity of cognate epitopes presented by defined HLA class I molecules. Conclusions Our results suggest that the population frequencies of specific T cells are strikingly non-uniform across epitopes that are known to elicit immune responses. This inference leads to a new definition of epitope immunogenicity based on specific TCR frequencies, which can be estimated with a high degree of accuracy in silico, thereby providing a novel framework to integrate computational and experimental genomics with basic and translational research efforts in the field of T cell immunology. Electronic supplementary material The online version of this article (10.1186/s13073-018-0577-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Alla D Fedorova
- Department of Genomics of Adaptive Immunity, IBCH RAS, Moscow, Russia
| | - James E McLaren
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Dmitri V Bagaev
- Department of Genomics of Adaptive Immunity, IBCH RAS, Moscow, Russia
| | - Alexey V Eliseev
- Department of Genomics of Adaptive Immunity, IBCH RAS, Moscow, Russia.,Department of Molecular Technologies, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Artem I Mikelov
- Department of Genomics of Adaptive Immunity, IBCH RAS, Moscow, Russia.,Center for Data-Intensive Biomedicine and Biotechnology, Skoltech, Moscow, Russia
| | - Anna E Koneva
- Department of Genomics of Adaptive Immunity, IBCH RAS, Moscow, Russia
| | - Ivan V Zvyagin
- Department of Genomics of Adaptive Immunity, IBCH RAS, Moscow, Russia.,Department of Molecular Technologies, Pirogov Russian National Research Medical University, Moscow, Russia
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK.,Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, UK
| | - Dmitry M Chudakov
- Department of Genomics of Adaptive Immunity, IBCH RAS, Moscow, Russia.,Department of Molecular Technologies, Pirogov Russian National Research Medical University, Moscow, Russia.,Center for Data-Intensive Biomedicine and Biotechnology, Skoltech, Moscow, Russia.,Central European Institute of Technology, CEITEC, Brno, Czech Republic
| | - Mikhail Shugay
- Department of Genomics of Adaptive Immunity, IBCH RAS, Moscow, Russia. .,Department of Molecular Technologies, Pirogov Russian National Research Medical University, Moscow, Russia. .,Center for Data-Intensive Biomedicine and Biotechnology, Skoltech, Moscow, Russia.
| |
Collapse
|
257
|
Hutchison S, Pritchard AL. Identifying neoantigens for use in immunotherapy. Mamm Genome 2018; 29:714-730. [PMID: 30167844 PMCID: PMC6267674 DOI: 10.1007/s00335-018-9771-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/08/2018] [Indexed: 12/14/2022]
Abstract
This review focuses on the types of cancer antigens that can be recognised by the immune system and form due to alterations in the cancer genome, including cancer testis, overexpressed and neoantigens. Specifically, neoantigens can form when cancer cell-specific mutations occur that result in alterations of the protein from ‘self’. This type of antigen can result in an immune response sufficient to clear tumour cells when activated. Furthermore, studies have reported that the likelihood of successful immunotherapeutic targeting of cancer by many different methods was reliant on immune response to neoantigens. The recent resurgence of interest in the immune response to tumour cells, in conjunction with technological advances, has resulted in a large increase in the predicted, identified and functionally confirmed neoantigens. This growth in identified neoantigen sequences has increased the contents of training sets for algorithms, which in turn improves the prediction of which genetic mutations may form neoantigens. Additionally, algorithms predicting how proteins will be processed into peptide epitopes by the proteasome and which peptides bind to the transporter complex are also improving with this research. Now that large screens of all the tumour-specific protein altering mutations are possible, the emerging data from assessment of the immunogenicity of neoantigens suggest that only a minority of variants will form targetable epitopes. The potential for immunotherapeutic targeting of neoantigens will therefore be greater in cancers with a higher frequency of protein altering somatic variants. There is considerable potential in the use of neoantigens to treat patients, either alone or in combination with other immunotherapies and with continued advancements, these potentials will be realised.
Collapse
Affiliation(s)
- Sharon Hutchison
- Genetics and Immunology Research Group, University of the Highlands and Islands, An Lòchran, 10 Inverness Campus, Inverness, IV2 5NA, Scotland, UK
| | - Antonia L Pritchard
- Genetics and Immunology Research Group, University of the Highlands and Islands, An Lòchran, 10 Inverness Campus, Inverness, IV2 5NA, Scotland, UK.
| |
Collapse
|
258
|
Barros L, Pretti MA, Chicaybam L, Abdo L, Boroni M, Bonamino MH. Immunological-based approaches for cancer therapy. Clinics (Sao Paulo) 2018; 73:e429s. [PMID: 30133560 PMCID: PMC6097086 DOI: 10.6061/clinics/2018/e429s] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/05/2018] [Indexed: 02/06/2023] Open
Abstract
The immunologic landscape of tumors has been continuously unveiled, providing a new look at the interactions between cancer cells and the immune system. Emerging tumor cells are constantly eliminated by the immune system, but some cells establish a long-term equilibrium phase leading to tumor immunoediting and, eventually, evasion. During this process, tumor cells tend to acquire more mutations. Bearing a high mutation burden leads to a greater number of neoantigens with the potential to initiate an immune response. Although many tumors evoke an immune response, tumor clearance by the immune system does not occur due to a suppressive tumor microenvironment. The mechanisms by which tumors achieve the ability to evade immunologic control vary. Understanding these differences is crucial for the improvement and application of new immune-based therapies. Much effort has been placed in developing in silico algorithms to predict tumor immunogenicity and to characterize the microenvironment via high-throughput sequencing and gene expression techniques. Each sequencing source, transcriptomics, and genomics yields a distinct level of data, helping to elucidate the tumor-based immune responses and guiding the fine-tuning of current and upcoming immune-based therapies. In this review, we explore some of the immunological concepts behind the new immunotherapies and the bioinformatic tools to study the immunological aspects of tumors, focusing on neoantigen determination and microenvironment deconvolution. We further discuss the immune-based therapies already in clinical use, those underway for future clinical application, the next steps in immunotherapy, and how the characterization of the tumor immune contexture can impact therapies aiming to promote or unleash immune-based tumor elimination.
Collapse
Affiliation(s)
- Luciana Barros
- Programa de Carcinogenese Molecular, Coordenacao de Pesquisa, Instituto Nacional de Cancer (INCA), Rio de Janeiro, RJ, BR
| | - Marco Antonio Pretti
- Programa de Carcinogenese Molecular, Coordenacao de Pesquisa, Instituto Nacional de Cancer (INCA), Rio de Janeiro, RJ, BR
| | | | - Luiza Abdo
- Programa de Carcinogenese Molecular, Coordenacao de Pesquisa, Instituto Nacional de Cancer (INCA), Rio de Janeiro, RJ, BR
| | - Mariana Boroni
- Laboratorio de Bioinformatica e Biologia Computacional, Coordenacao de Pesquisa, Instituto Nacional de Cancer (INCA), Rio de Janeiro, RJ, BR
| | - Martin Hernán Bonamino
- Laboratorio de Bioinformatica e Biologia Computacional, Coordenacao de Pesquisa, Instituto Nacional de Cancer (INCA), Rio de Janeiro, RJ, BR
- *Corresponding author. E-mail: /
| |
Collapse
|
259
|
Bykova NA, Malko DB, Efimov GA. In Silico Analysis of the Minor Histocompatibility Antigen Landscape Based on the 1000 Genomes Project. Front Immunol 2018; 9:1819. [PMID: 30166983 PMCID: PMC6105694 DOI: 10.3389/fimmu.2018.01819] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/24/2018] [Indexed: 12/30/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is routinely used to treat hematopoietic malignancies. The eradication of residual tumor cells during engraftment is mediated by donor cytotoxic T lymphocytes reactive to alloantigens. In a HLA-matched transplantation context, alloantigens are encoded by various polymorphic genes situated outside the HLA locus, also called minor histocompatibility antigens (MiHAs). Recently, MiHAs have been recognized as promising targets for post-transplantation T-cell immunotherapy as they have several appealing advantages over tumor-associated antigens (TAAs) and neoantigens, i.e., they are more abundant than TAAs, which potentially facilitates multiple targeting; and unlike neoantigens, they are encoded by germline polymorphisms, some of which are common and thus, suitable for off-the-shelf therapy. The genetic sources of MiHAs are nonsynonymous polymorphisms that cause differences between the recipient and donor proteomes and subsequently, the immunopeptidomes. Systematic description of the alloantigen landscape in HLA-matched transplantation is still lacking as previous studies focused only on a few immunogenic and common MiHAs. Here, we perform a thorough in silico analysis of the public genomic data to classify genetic polymorphisms that lead to MiHA formation and estimate the number of potentially available MiHA mismatches. Our findings suggest that a donor/recipient pair is expected to have at least several dozen mismatched strong MHC-binding SNP-associated peptides per HLA allele (116 ± 26 and 65 ± 15 for non-related pairs and siblings respectively in European populations as predicted by two independent algorithms). Over 70% of them are encoded by relatively frequent polymorphisms (minor allele frequency > 0.1) and thus, may be targetable by off-the-shelf therapeutics. We showed that the most appealing targets (probability of mismatch over 20%) reside in the asymmetric allele frequency region, which spans from 0.15 to 0.47 and corresponds to an order of several hundred (213 ± 47) possible targets per HLA allele that can be considered for immunogenicity validation. Overall, these findings demonstrate the significant potential of MiHAs as targets for T-cell immunotherapy and emphasize the need for the systematic discovery of novel MiHAs.
Collapse
Affiliation(s)
- Nadia A Bykova
- Laboratory of Transplantation Immunology, National Research Center for Hematology, Moscow, Russia
| | - Dmitry B Malko
- Laboratory of Transplantation Immunology, National Research Center for Hematology, Moscow, Russia
| | - Grigory A Efimov
- Laboratory of Transplantation Immunology, National Research Center for Hematology, Moscow, Russia
| |
Collapse
|
260
|
Gettinger SN, Choi J, Mani N, Sanmamed MF, Datar I, Sowell R, Du VY, Kaftan E, Goldberg S, Dong W, Zelterman D, Politi K, Kavathas P, Kaech S, Yu X, Zhao H, Schlessinger J, Lifton R, Rimm DL, Chen L, Herbst RS, Schalper KA. A dormant TIL phenotype defines non-small cell lung carcinomas sensitive to immune checkpoint blockers. Nat Commun 2018; 9:3196. [PMID: 30097571 PMCID: PMC6086912 DOI: 10.1038/s41467-018-05032-8] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 06/07/2018] [Indexed: 02/07/2023] Open
Abstract
The biological determinants of sensitivity and resistance to immune checkpoint blockers are not completely understood. To elucidate the role of intratumoral T-cells and their association with the tumor genomic landscape, we perform paired whole exome DNA sequencing and multiplexed quantitative immunofluorescence (QIF) in pre-treatment samples from non-small cell lung carcinoma (NSCLC) patients treated with PD-1 axis blockers. QIF is used to simultaneously measure the level of CD3+ tumor infiltrating lymphocytes (TILs), in situ T-cell proliferation (Ki-67 in CD3) and effector capacity (Granzyme-B in CD3). Elevated mutational load, candidate class-I neoantigens or intratumoral CD3 signal are significantly associated with favorable response to therapy. Additionally, a "dormant" TIL signature is associated with survival benefit in patients treated with immune checkpoint blockers characterized by elevated TILs with low activation and proliferation. We further demonstrate that dormant TILs can be reinvigorated upon PD-1 blockade in a patient-derived xenograft model.
Collapse
Affiliation(s)
- S N Gettinger
- Medical Oncology and Yale Cancer Center, New Haven, CT, 06511, USA
| | - J Choi
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - N Mani
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06511, USA
- Translational Immuno-oncology Laboratory, Yale Cancer Center, New Haven, CT, 06511, USA
| | - M F Sanmamed
- Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - I Datar
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06511, USA
- Translational Immuno-oncology Laboratory, Yale Cancer Center, New Haven, CT, 06511, USA
| | - Ryan Sowell
- Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Victor Y Du
- Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - E Kaftan
- Medical Oncology and Yale Cancer Center, New Haven, CT, 06511, USA
- Translational Immuno-oncology Laboratory, Yale Cancer Center, New Haven, CT, 06511, USA
| | - S Goldberg
- Medical Oncology and Yale Cancer Center, New Haven, CT, 06511, USA
| | - W Dong
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - D Zelterman
- Yale School of Public Health, New Haven, CT, 06511, USA
| | - K Politi
- Medical Oncology and Yale Cancer Center, New Haven, CT, 06511, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - P Kavathas
- Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
| | - S Kaech
- Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - X Yu
- Yale School of Public Health, New Haven, CT, 06511, USA
| | - H Zhao
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06511, USA
- Yale School of Public Health, New Haven, CT, 06511, USA
| | - J Schlessinger
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - R Lifton
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - D L Rimm
- Medical Oncology and Yale Cancer Center, New Haven, CT, 06511, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - L Chen
- Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - R S Herbst
- Medical Oncology and Yale Cancer Center, New Haven, CT, 06511, USA
| | - K A Schalper
- Medical Oncology and Yale Cancer Center, New Haven, CT, 06511, USA.
- Department of Pathology, Yale School of Medicine, New Haven, CT, 06511, USA.
- Translational Immuno-oncology Laboratory, Yale Cancer Center, New Haven, CT, 06511, USA.
| |
Collapse
|
261
|
Gfeller D, Bassani-Sternberg M. Predicting Antigen Presentation-What Could We Learn From a Million Peptides? Front Immunol 2018; 9:1716. [PMID: 30090105 PMCID: PMC6068240 DOI: 10.3389/fimmu.2018.01716] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/12/2018] [Indexed: 12/30/2022] Open
Abstract
Antigen presentation lies at the heart of immune recognition of infected or malignant cells. For this reason, important efforts have been made to predict which peptides are more likely to bind and be presented by the human leukocyte antigen (HLA) complex at the surface of cells. These predictions have become even more important with the advent of next-generation sequencing technologies that enable researchers and clinicians to rapidly determine the sequences of pathogens (and their multiple variants) or identify non-synonymous genetic alterations in cancer cells. Here, we review recent advances in predicting HLA binding and antigen presentation in human cells. We argue that the very large amount of high-quality mass spectrometry data of eluted (mainly self) HLA ligands generated in the last few years provides unprecedented opportunities to improve our ability to predict antigen presentation and learn new properties of HLA molecules, as demonstrated in many recent studies of naturally presented HLA-I ligands. Although major challenges still lie on the road toward the ultimate goal of predicting immunogenicity, these experimental and computational developments will facilitate screening of putative epitopes, which may eventually help decipher the rules governing T cell recognition.
Collapse
Affiliation(s)
- David Gfeller
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| |
Collapse
|
262
|
Egorov ES, Kasatskaya SA, Zubov VN, Izraelson M, Nakonechnaya TO, Staroverov DB, Angius A, Cucca F, Mamedov IZ, Rosati E, Franke A, Shugay M, Pogorelyy MV, Chudakov DM, Britanova OV. The Changing Landscape of Naive T Cell Receptor Repertoire With Human Aging. Front Immunol 2018; 9:1618. [PMID: 30087674 PMCID: PMC6066563 DOI: 10.3389/fimmu.2018.01618] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/29/2018] [Indexed: 12/22/2022] Open
Abstract
Human aging is associated with a profound loss of thymus productivity, yet naïve T lymphocytes still maintain their numbers by division in the periphery for many years. The extent of such proliferation may depend on the cytokine environment, including IL-7 and T-cell receptor (TCR) “tonic” signaling mediated by self pMHCs recognition. Additionally, intrinsic properties of distinct subpopulations of naïve T cells could influence the overall dynamics of aging-related changes within the naïve T cell compartment. Here, we investigated the differences in the architecture of TCR beta repertoires for naïve CD4, naïve CD8, naïve CD4+CD25−CD31+ (enriched with recent thymic emigrants, RTE), and mature naïve CD4+CD25−CD31− peripheral blood subsets between young and middle-age/old healthy individuals. In addition to observing the accumulation of clonal expansions (as was shown previously), we reveal several notable changes in the characteristics of T cell repertoire. We observed significant decrease of CDR3 length, NDN insert, and number of non-template added N nucleotides within TCR beta CDR3 with aging, together with a prominent change of physicochemical properties of the central part of CDR3 loop. These changes were similar across CD4, CD8, RTE-enriched, and mature CD4 subsets of naïve T cells, with minimal or no difference observed between the latter two subsets for individuals of the same age group. We also observed an increase in “publicity” (fraction of shared clonotypes) of CD4, but not CD8 naïve T cell repertoires. We propose several explanations for these phenomena built upon previous studies of naïve T-cell homeostasis, and call for further studies of the mechanisms causing the observed changes and of consequences of these changes in respect of the possible holes formed in the landscape of naïve T cell TCR repertoire.
Collapse
Affiliation(s)
- Evgeny S Egorov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Sofya A Kasatskaya
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.,Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Vasiliy N Zubov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Mark Izraelson
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | | | | | - Andrea Angius
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | - Ilgar Z Mamedov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Elisa Rosati
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Mikhail Shugay
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.,Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | - Dmitriy M Chudakov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.,Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Olga V Britanova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
263
|
Novel frontiers in detecting cancer metastasis. Clin Exp Metastasis 2018; 35:403-412. [PMID: 30022365 DOI: 10.1007/s10585-018-9918-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 05/21/2018] [Indexed: 12/15/2022]
Abstract
Cancer microenvironment is the critical battle ground between the cancer cells and host response. Thus, more emphasis is directed to study the relationship between cancer cells and the stromal cells. Multiplex microscopy is an emerging technique in which multiple cell populations within the cancer microenvironment may be stained so that spatial relationship between cancer cells and, in particular, the immune cells may be studied during different stages of cancer development. Recent discovery of mutational burden and neoantigens in cancer has opened new landscapes in the interaction of host immune cells and cancer neoantigens. The emerging role of miRNAs may become an added dimension to study cancer beyond traditional pathway of DNA directed RNA being associated with the malignant behavior of cancer. Circulating tumor cells, cancer markers and ctDNA can be used as markers for circulating cancer cells in the blood. Further studies are needed to validate if liquid biopsy of cancer may become a routine clinical tool to screen cancer or follow patients for recurrence or responses to treatment.
Collapse
|
264
|
Pawelec G. Does patient age influence anti-cancer immunity? Semin Immunopathol 2018; 41:125-131. [DOI: 10.1007/s00281-018-0697-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 07/06/2018] [Indexed: 12/30/2022]
|
265
|
Tubb VM, Schrikkema DS, Croft NP, Purcell AW, Linnemann C, Freriks MR, Chen F, Long HM, Lee SP, Bendle GM. Isolation of T cell receptors targeting recurrent neoantigens in hematological malignancies. J Immunother Cancer 2018; 6:70. [PMID: 30001747 PMCID: PMC6044029 DOI: 10.1186/s40425-018-0386-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/04/2018] [Indexed: 12/25/2022] Open
Abstract
Mutation-derived neoantigens represent an important class of tumour-specific, tumour rejection antigens, and are attractive targets for TCR gene therapy of cancer. The majority of such mutations are patient-specific and targeting these requires a fully personalized approach. However, some mutations are found recurrently among cancer patients, and represent potential targets for neoantigen-specific TCR gene therapy that is more widely applicable. Therefore, we have investigated if some cancer mutations found recurrently in hematological malignancies encode immunogenic neoantigens presented by common European Caucasoid HLA class I alleles and can form targets for TCR gene therapy. We initially focused on identifying HLA class I neoepitopes derived from calreticulin (CALR) exon 9 mutations, found in ~ 80% of JAK2wt myeloproliferative neoplasms (MPN). Based on MHC class I peptide predictions, a number of peptides derived from mutant CALR (mCALR) were predicted to bind to HLA-A*03:01 and HLA-B*07:02. However, using mass spectrometry and ex vivo pMHC multimer staining of PBMC from MPN patients with CALR exon 9 mutations, we found no evidence that these peptides were naturally processed and presented on the surface of mCALR-expressing target cells. We next developed a protocol utilizing pMHC multimers to isolate CD8+ T cells from healthy human donor PBMC that are specific for mCALR and additional putative neoepitopes found recurrently in hematological malignancies. Using this approach, CD8+ T cells specific for HLA-A*03:01- and HLA-B*07:02-presented mCALR peptides and an HLA-A*11:01-presented mutant FBXW7 (mFBXW7) peptide were successfully isolated. TCRs isolated from mCALR-specific CD8+ T cell populations were not able to recognize target cells engineered to express mCALR. In contrast, mFBXW7-specific CD8+ T cells were able to recognize target cells engineered to express mFBXW7. In conclusion, while we found no evidence for mCALR derived neoepitope presentation in the context of the HLA class I alleles studied, our data suggests that the recurrent pR465H mutation in FBXW7 may encode an HLA-A*11:01 presented neoepitope, and warrants further investigation as a target for T cell based immunotherapy of cancer.
Collapse
Affiliation(s)
- Vanessa M Tubb
- Institute of Immunology and Immunotherapy, Cancer Immunology and Immunotherapy Centre, University of Birmingham, Birmingham, UK.
| | - Deborah S Schrikkema
- Institute of Immunology and Immunotherapy, Cancer Immunology and Immunotherapy Centre, University of Birmingham, Birmingham, UK
| | - Nathan P Croft
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Anthony W Purcell
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Carsten Linnemann
- Division of Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Manon R Freriks
- Division of Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Frederick Chen
- Centre for Clinical Haematology, Queen Elizabeth Hospital NHS Foundation Trust, Birmingham, UK
| | - Heather M Long
- Institute of Immunology and Immunotherapy, Cancer Immunology and Immunotherapy Centre, University of Birmingham, Birmingham, UK
| | - Steven P Lee
- Institute of Immunology and Immunotherapy, Cancer Immunology and Immunotherapy Centre, University of Birmingham, Birmingham, UK
| | - Gavin M Bendle
- Institute of Immunology and Immunotherapy, Cancer Immunology and Immunotherapy Centre, University of Birmingham, Birmingham, UK
| |
Collapse
|
266
|
Matko S, Manderla J, Bonsack M, Schmitz M, Bornhauser M, Tonn T, Odendahl M. PRAME peptide-specific CD8 + T cells represent the predominant response against leukemia-associated antigens in healthy individuals. Eur J Immunol 2018; 48:1400-1411. [PMID: 29738081 DOI: 10.1002/eji.201747399] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 04/10/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022]
Abstract
Antigen-specific T cells isolated from healthy individuals (HIs) have shown great therapeutic potential upon adoptive transfer for the treatment of viremia in immunosuppressed patients. The lack of comprehensive data on the prevalence and characteristics of leukemia-associated antigen (LAA)-specific T cells in HIs still limits such an approach for tumor therapy. Therefore, we have investigated T-cell responses against prominent candidates comprising Wilms' tumor protein 1 (WT1), preferentially expressed antigen in melanoma (PRAME), Survivin, NY-ESO, and p53 by screening PBMCs from HIs using intracellular IFN-γ staining following provocation with LAA peptide mixes. Here, we found predominantly poly-functional effector/effector memory CCR7- /CD45RA+/- /CD8+ LAA peptide-specific T cells with varying CD95 expression in 34 of 100 tested HIs, whereas CD4+ T cells responses were restricted to 5. Most frequent LAA peptide-specific T cell responses were directed against WT1 and PRAME peptides with a prevalence of 20 and 17%, respectively, showing the highest magnitude (0.16% ± 0.22% (mean ± SD)) for PRAME peptides. Cytotoxicity of PRAME peptide-specific T cells was demonstrated by specific killing of PRAME peptide-pulsed T2 cells. Furthermore, the proliferative capacity of PRAME peptide-specific T cells was confined to HIs responsive toward PRAME peptide challenge corroborating the accuracy of the screening results. In conclusion, we identified PRAME as a promising target antigen for adoptive leukemia therapy.
Collapse
Affiliation(s)
- Sarah Matko
- Experimental Transfusion Medicine, Medical Faculty Carl Gustav Carus, TU Dresden, Germany.,Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Julia Manderla
- Experimental Transfusion Medicine, Medical Faculty Carl Gustav Carus, TU Dresden, Germany.,Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Maria Bonsack
- Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| | - Marc Schmitz
- Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany.,German Cancer Research Center (DKFZ) partner site Dresden; and German Cancer Consortium (DKTK), Heidelberg, Germany.,Institute of Immunology, Medical Faculty, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Martin Bornhauser
- Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany.,German Cancer Research Center (DKFZ) partner site Dresden; and German Cancer Consortium (DKTK), Heidelberg, Germany.,Department of Medicine 1, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Torsten Tonn
- Experimental Transfusion Medicine, Medical Faculty Carl Gustav Carus, TU Dresden, Germany.,Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany.,German Cancer Research Center (DKFZ) partner site Dresden; and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Marcus Odendahl
- Experimental Transfusion Medicine, Medical Faculty Carl Gustav Carus, TU Dresden, Germany.,Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| |
Collapse
|
267
|
Borcherding N, Kolb R, Gullicksrud J, Vikas P, Zhu Y, Zhang W. Keeping Tumors in Check: A Mechanistic Review of Clinical Response and Resistance to Immune Checkpoint Blockade in Cancer. J Mol Biol 2018; 430:2014-2029. [PMID: 29800567 PMCID: PMC6071324 DOI: 10.1016/j.jmb.2018.05.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/26/2022]
Abstract
Immune checkpoints are a diverse set of inhibitory signals to the immune system that play a functional role in adaptive immune response and self-tolerance. Dysregulation of these pathways is a vital mechanism in the avoidance of immune destruction by tumor cells. Immune checkpoint blockade (ICB) refers to targeted strategies to disrupt the tumor co-opted immune suppression to enhance anti-tumor immunity. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1) are two immune checkpoints that have the widest range of antibody-based therapies. These therapies have gone from promising approaches to Food and Drug Administration-approved first- and second-line agents for a number of immunogenic cancers. The burgeoning investigations of ICB efficacy in blood and solid cancers have underscored the importance of identifying the predictors of response and resistance to ICB. Identification of response correlates is made complicated by the observations of mixed reactions, or different responses in multiple lesions from the same patient, and delayed responses that can occur over a year after the induction therapy. Factors that can influence response and resistance in ICB can illuminate underlying molecular mechanisms of immune activation and suppression. These same response predictors can guide the identification of patients who would benefit from ICB, reduce off-target immune-relate adverse events, and facilitate the use of combinatorial therapies to increase efficacy. Here we review the underlying principles of immune checkpoint therapy and results of single-agent ICB clinical trials, and summarize the predictors of response and resistance.
Collapse
Affiliation(s)
- Nicholas Borcherding
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA
| | - Ryan Kolb
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA
| | - Jodi Gullicksrud
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Praveen Vikas
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA
| | - Yuwen Zhu
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Weizhou Zhang
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA; Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA 52242-11, USA.
| |
Collapse
|
268
|
Tran T, Blanc C, Granier C, Saldmann A, Tanchot C, Tartour E. Therapeutic cancer vaccine: building the future from lessons of the past. Semin Immunopathol 2018; 41:69-85. [PMID: 29978248 DOI: 10.1007/s00281-018-0691-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022]
Abstract
Anti-cancer vaccines have raised many hopes from the start of immunotherapy but have not yet been clinically successful. The few positive results of anti-cancer vaccines have been observed in clinical situations of low tumor burden or preneoplastic lesions. Several new concepts and new results reposition this therapeutic approach in the field of immunotherapy. Indeed, cancers that respond to anti-PD-1/PD-L1 (20-30%) are those that are infiltrated by anti-tumor T cells with an inflammatory infiltrate. However, 70% of cancers do not appear to have an anti-tumor immune reaction in the tumor microenvironment. To induce this anti-tumor immunity, therapeutic combinations between vaccines and anti-PD-1/PD-L1 are being evaluated. In addition, the identification of neoepitopes against which the immune system is less tolerated is giving rise to a new enthusiasm by the first clinical results of the vaccine including these neoepitopes in humans. The ability of anti-cancer vaccines to induce a population of anti-tumor T cells called memory resident T cells that play an important role in immunosurveillance is also a new criterion to consider in the design of therapeutic vaccines.
Collapse
Affiliation(s)
- T Tran
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - C Blanc
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - C Granier
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - A Saldmann
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - C Tanchot
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Eric Tartour
- INSERM U970, Paris Cardiovascular Research Center (PARCC), Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
- Hôpital Européen Georges Pompidou, Laboratory of Immunology, Assistance Publique des Hôpitaux de Paris, Paris, France.
- Equipe Labellisée Ligue Nationale contre le Cancer, Paris, France.
| |
Collapse
|
269
|
Abstract
The recent advances in in vitro 3D culture technologies, such as organoids, have opened new avenues for the development of novel, more physiological human cancer models. Such preclinical models are essential for more efficient translation of basic cancer research into novel treatment regimens for patients with cancer. Wild-type organoids can be grown from embryonic and adult stem cells and display self-organizing capacities, phenocopying essential aspects of the organs they are derived from. Genetic modification of organoids allows disease modelling in a setting that approaches the physiological environment. Additionally, organoids can be grown with high efficiency from patient-derived healthy and tumour tissues, potentially enabling patient-specific drug testing and the development of individualized treatment regimens. In this Review, we evaluate tumour organoid protocols and how they can be utilized as an alternative model for cancer research.
Collapse
Affiliation(s)
- Jarno Drost
- Princess Máxima Centre for Paediatric Oncology, Utrecht, Netherlands.
| | - Hans Clevers
- Princess Máxima Centre for Paediatric Oncology, Utrecht, Netherlands
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
270
|
Legut M, Sewell AK. Designer T-cells and T-cell receptors for customized cancer immunotherapies. Curr Opin Pharmacol 2018; 41:96-103. [PMID: 29852403 DOI: 10.1016/j.coph.2018.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/14/2018] [Indexed: 12/27/2022]
Abstract
Cancer immunotherapy, focused on harnessing and empowering the immune system against tumours, has transformed modern oncology. One of the most promising avenues in development involves using genetically engineered T-cells to target cancer antigens via specific T-cell receptors (TCRs). TCRs have a naturally low affinity towards cancer-associated antigens, and therefore show scope for improvement. Here we describe approaches to procure TCRs with enhanced affinity and specificity towards cancer, using protein engineering or selection of natural TCRs from unadulterated repertoires. In particular, we discuss novel methods facilitating the targeting of tumour-specific mutations. Finally, we provide a prospective outlook on the potential development of novel, off-the-shelf immunotherapies by leveraging recent advances in genome editing.
Collapse
Affiliation(s)
- Mateusz Legut
- Division of Infection and Immunity, Cardiff University School of Medicine, Henry Wellcome Building, University Hospital Wales, Cardiff CF14 4XN, Wales, UK
| | - Andrew K Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Henry Wellcome Building, University Hospital Wales, Cardiff CF14 4XN, Wales, UK.
| |
Collapse
|
271
|
Creech AL, Ting YS, Goulding SP, Sauld JF, Barthelme D, Rooney MS, Addona TA, Abelin JG. The Role of Mass Spectrometry and Proteogenomics in the Advancement of HLA Epitope Prediction. Proteomics 2018; 18:e1700259. [PMID: 29314742 PMCID: PMC6033110 DOI: 10.1002/pmic.201700259] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/12/2017] [Indexed: 12/30/2022]
Abstract
A challenge in developing personalized cancer immunotherapies is the prediction of putative cancer-specific antigens. Currently, predictive algorithms are used to infer binding of peptides to human leukocyte antigen (HLA) heterodimers to aid in the selection of putative epitope targets. One drawback of current epitope prediction algorithms is that they are trained on datasets containing biochemical HLA-peptide binding data that may not completely capture the rules associated with endogenous processing and presentation. The field of MS has made great improvements in instrumentation speed and sensitivity, chromatographic resolution, and proteogenomic database search strategies to facilitate the identification of HLA-ligands from a variety of cell types and tumor tissues. As such, these advances have enabled MS profiling of HLA-binding peptides to be a tractable, orthogonal approach to lower throughput biochemical assays for generating comprehensive datasets to train epitope prediction algorithms. In this review, we will highlight the progress made in the field of HLA-ligand profiling enabled by MS and its impact on current and future epitope prediction strategies.
Collapse
|
272
|
Milam AAV, Bartleson JM, Donermeyer DL, Horvath S, Durai V, Raju S, Yu H, Redmann V, Zinselmeyer B, White JM, Murphy KM, Allen PM. Tuning T Cell Signaling Sensitivity Alters the Behavior of CD4 + T Cells during an Immune Response. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:3429-3437. [PMID: 29618523 PMCID: PMC5940509 DOI: 10.4049/jimmunol.1701422] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/13/2018] [Indexed: 11/19/2022]
Abstract
Intricate processes in the thymus and periphery help curb the development and activation of autoreactive T cells. The subtle signals that govern these processes are an area of great interest, but tuning TCR sensitivity for the purpose of affecting T cell behavior remains technically challenging. Previously, our laboratory described the derivation of two TCR-transgenic CD4 T cell mouse lines, LLO56 and LLO118, which recognize the same cognate Listeria epitope with the same affinity. Despite the similarity of the two TCRs, LLO56 cells respond poorly in a primary infection whereas LLO118 cells respond robustly. Phenotypic examination of both lines revealed a substantial difference in their surface of expression of CD5, which serves as a dependable readout of the self-reactivity of a cell. We hypothesized that the increased interaction with self by the CD5-high LLO56 was mediated through TCR signaling, and was involved in the characteristic weak primary response of LLO56 to infection. To explore this issue, we generated an inducible knock-in mouse expressing the self-sensitizing voltage-gated sodium channel Scn5a. Overexpression of Scn5a in peripheral T cells via the CD4-Cre promoter resulted in increased TCR-proximal signaling. Further, Scn5a-expressing LLO118 cells, after transfer into BL6 recipient mice, displayed an impaired response during infection relative to wild-type LLO118 cells. In this way, we were able to demonstrate that tuning of TCR sensitivity to self can be used to alter in vivo immune responses. Overall, these studies highlight the critical relationship between TCR-self-pMHC interaction and an immune response to infection.
Collapse
Affiliation(s)
- Ashley A Viehmann Milam
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Juliet M Bartleson
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - David L Donermeyer
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Stephen Horvath
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Vivek Durai
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Saravanan Raju
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Haiyang Yu
- Ludwig Institute for Cancer Research, La Jolla, CA 92093; and
| | - Veronika Redmann
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Bernd Zinselmeyer
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - J Michael White
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Kenneth M Murphy
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
- Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110
| | - Paul M Allen
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110;
| |
Collapse
|
273
|
Update on Tumor Neoantigens and Their Utility: Why It Is Good to Be Different. Trends Immunol 2018; 39:536-548. [PMID: 29751996 DOI: 10.1016/j.it.2018.04.005] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 12/18/2022]
Abstract
Antitumor rejection by the immune system is a complex process that is regulated by several factors. Among these factors are the quality and quantity of mutational events that occur in cancer cells. Perhaps one of the most important types of mutations that influence antitumor immunity is the neoantigen, that is, a non-self-antigen that arises as a result of somatic mutation. Recent work has demonstrated that neoantigens hold significant promise for developing new diagnostic and therapeutic modalities. Therapeutic targeting of neoantigens is important for achieving benefit following therapy with immune checkpoint blockade agents or for cancer vaccines targeting mutations. Here, we review our understanding of neoantigens and discuss new developments in the quest to use them in cancer immunotherapy.
Collapse
|
274
|
Matsuda T, Leisegang M, Park JH, Ren L, Kato T, Ikeda Y, Harada M, Kiyotani K, Lengyel E, Fleming GF, Nakamura Y. Induction of Neoantigen-Specific Cytotoxic T Cells and Construction of T-cell Receptor–Engineered T Cells for Ovarian Cancer. Clin Cancer Res 2018; 24:5357-5367. [DOI: 10.1158/1078-0432.ccr-18-0142] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/31/2018] [Accepted: 04/30/2018] [Indexed: 01/26/2023]
|
275
|
The perfect personalized cancer therapy: cancer vaccines against neoantigens. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:86. [PMID: 29678194 PMCID: PMC5910567 DOI: 10.1186/s13046-018-0751-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 04/03/2018] [Indexed: 02/07/2023]
Abstract
In the advent of Immune Checkpoint inhibitors (ICI) and of CAR-T adoptive T-cells, the new frontier in Oncology is Cancer Immunotherapy because of its ability to provide long term clinical benefit in metastatic disease in several solid and liquid tumor types. It is now clear that ICI acts by unmasking preexisting immune responses as well as by inducing de novo responses against tumor neoantigens. Thanks to theprogress made in genomics technologies and the evolution of bioinformatics, neoantigens represent ideal targets, due to their specific expression in cancer tissue and the potential lack of side effects. In this review, we discuss the promise of preclinical and clinical results with mutation-derived neoantigen cancer vaccines (NCVs) along with the current limitations from bioinformatics prediction to manufacturing an effective new therapeutic approach.
Collapse
|
276
|
Krackhardt AM, Anliker B, Hildebrandt M, Bachmann M, Eichmüller SB, Nettelbeck DM, Renner M, Uharek L, Willimsky G, Schmitt M, Wels WS, Schüssler-Lenz M. Clinical translation and regulatory aspects of CAR/TCR-based adoptive cell therapies-the German Cancer Consortium approach. Cancer Immunol Immunother 2018; 67:513-523. [PMID: 29380009 PMCID: PMC11028374 DOI: 10.1007/s00262-018-2119-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 01/20/2018] [Indexed: 12/17/2022]
Abstract
Adoptive transfer of T cells genetically modified by TCRs or CARs represents a highly attractive novel therapeutic strategy to treat malignant diseases. Various approaches for the development of such gene therapy medicinal products (GTMPs) have been initiated by scientists in recent years. To date, however, the number of clinical trials commenced in Germany and Europe is still low. Several hurdles may contribute to the delay in clinical translation of these therapeutic innovations including the significant complexity of manufacture and non-clinical testing of these novel medicinal products, the limited knowledge about the intricate regulatory requirements of the academic developers as well as limitations of funds for clinical testing. A suitable good manufacturing practice (GMP) environment is a key prerequisite and platform for the development, validation, and manufacture of such cell-based therapies, but may also represent a bottleneck for clinical translation. The German Cancer Consortium (DKTK) and the Paul-Ehrlich-Institut (PEI) have initiated joint efforts of researchers and regulators to facilitate and advance early phase, academia-driven clinical trials. Starting with a workshop held in 2016, stakeholders from academia and regulatory authorities in Germany have entered into continuing discussions on a diversity of scientific, manufacturing, and regulatory aspects, as well as the benefits and risks of clinical application of CAR/TCR-based cell therapies. This review summarizes the current state of discussions of this cooperative approach providing a basis for further policy-making and suitable modification of processes.
Collapse
Affiliation(s)
- Angela M Krackhardt
- Klinik und Poliklinik für Innere Medizin III, Hämatologie und Onkologie, Klinikum rechts der Isar, TU München, TUM School of Medicine, Munich, Germany.
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany.
| | - Brigitte Anliker
- Paul-Ehrlich-Institut (PEI, German Federal Institute for Vaccines and Biomedicines), Langen, Germany
| | - Martin Hildebrandt
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- TUMCells (Interdisciplinary Center for Cellular Therapies), TUM School of Medicine, Munich, Germany
| | - Michael Bachmann
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Helmholtz Zentrum Dresden Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Radio and Tumorimmunology, Dresden, Germany
- Nationales Centrum für Tumorerkrankungen (NCT), Heidelberg and Dresden, Germany
| | - Stefan B Eichmüller
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Nationales Centrum für Tumorerkrankungen (NCT), Heidelberg and Dresden, Germany
- GMP and T Cell Therapy Unit, DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
| | - Dirk M Nettelbeck
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
| | - Matthias Renner
- Paul-Ehrlich-Institut (PEI, German Federal Institute for Vaccines and Biomedicines), Langen, Germany
| | - Lutz Uharek
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Stem Cell Facility, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Gerald Willimsky
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Michael Schmitt
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Department of Internal Medicine V, GMP Core Facility, Heidelberg University Hospital, Heidelberg, Germany
| | - Winfried S Wels
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Martina Schüssler-Lenz
- Paul-Ehrlich-Institut (PEI, German Federal Institute for Vaccines and Biomedicines), Langen, Germany
| |
Collapse
|
277
|
A meta-analysis of HLA peptidome composition in different hematological entities: entity-specific dividing lines and "pan-leukemia" antigens. Oncotarget 2018; 8:43915-43924. [PMID: 28159928 PMCID: PMC5546449 DOI: 10.18632/oncotarget.14918] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/26/2016] [Indexed: 12/15/2022] Open
Abstract
Hematological malignancies (HM) are highly amenable targets for immunotherapeutic intervention and may be effectively treated by antigen-specific T-cell based treatment. Recent studies demonstrate that physiologically occurring anti-cancer T-cell responses in certain HM entities target broadly presented non-mutated epitopes. HLA ligands are thus implied as prime targets for broadly applicable and antigen-specific off-the-shelf compounds. With the aim of assessing the presence of common targets shared among different HM which may enable addressing a larger patient collective we conducted a meta-analysis of 83 mass spectrometry-based HLA peptidome datasets (comprising 40,361 unique peptide identifications) across four major HM (19 AML, 16 CML, 35 CLL, and 13 MM/MCL samples) and investigated similarities and differences within the HLA presented antigenic landscape. We found the cancer HLA peptidome datasets to cluster specifically along entity and lineage lines, suggesting that the immunopeptidome directly reflects the differences in the underlying (tumor-)biology. In line with these findings, we only detected a small set of entity-spanning antigens, which were predominantly characterized by low presentation frequencies within the different patient cohorts. These findings suggest that design of T-cell immunotherapies for the treatment of HM should ideally be conducted in an entity-specific fashion.
Collapse
|
278
|
Wieder T, Eigentler T, Brenner E, Röcken M. Immune checkpoint blockade therapy. J Allergy Clin Immunol 2018; 142:1403-1414. [PMID: 29596939 DOI: 10.1016/j.jaci.2018.02.042] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 02/09/2018] [Accepted: 02/27/2018] [Indexed: 12/30/2022]
Abstract
Immune checkpoints are accessory molecules that either promote or inhibit T-cell activation. Two inhibitory molecules, cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1), got high attention, as inhibition of CTLA-4 or PD-1 signaling provides the first immune therapy that significantly improves the survival of patients with metastatic solid cancers. Inhibition of CTLA-4 or PD-1 was first studied in and approved for patients with metastatic melanoma. Blocking immune checkpoints is also efficient in non-small-cell lung cancer, renal cell cancers, hypermutated gastrointestinal cancers, and others. Immune responses, whether directed against infections or against tumors, are divided into 2 phases: an initiation phase and an activation phase, where the immune system recognizes a danger signal and becomes activated by innate signals to fight the danger. This reaction is fundamental for the control of infections and cancer, but needs to be turned off once the danger is controlled, because persistence of this activation ultimately causes severe tissue damage. Therefore, each activation of the immune system is followed by a termination phase, where endogenous immune suppressor molecules arrest immune responses to prevent harmful damage. In the case of cancer immune therapies, therapeutic approaches classically enhanced the initiation and activation of immune responses to increase the emergence and the efficacy of cytotoxic T lymphocytes (CTL) against cancers. In sharp contrast, immune checkpoint blockade focuses on the termination of immune responses by inhibiting immune suppressor molecules. It thus prevents the termination of immune responses or even awakes those CTLs that became exhausted during an immune response. Therefore, blocking negatively regulating immune checkpoints restores the capacity of exhausted CTL to kill the cancer they infiltrate. In addition, they drive surviving cancer cells into a still poorly defined state of dormancy. As the therapy also awakes self-reactive CTL, one downside of the therapy is the induction of organ-specific autoimmune diseases. The second downside is the exorbitant drug price that withdraws patients in need from a therapy that was developed by academic research, which impairs further academic treatment development and financially charges the public health system.
Collapse
Affiliation(s)
- Thomas Wieder
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Thomas Eigentler
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Ellen Brenner
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Martin Röcken
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
279
|
Immune signatures predicting responses to immunomodulatory antibody therapy. Curr Opin Immunol 2018; 51:91-96. [PMID: 29550661 DOI: 10.1016/j.coi.2018.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 02/02/2018] [Accepted: 03/01/2018] [Indexed: 12/30/2022]
Abstract
Since the first immunomodulatory antibody was licensed by the FDA in 2011 for treating melanoma it has remained the case that only a certain proportion of cancer patients respond favourably to a particular therapy. Recent results from combining two or more different antibodies each targeting a different immune checkpoint indicate that the proportion of responding patients can be increased, but thus far there are no such therapies routinely yielding clinical benefit in 100% of patients in any cancer type. Therefore, predicting which patients will respond to a particular therapy remains of the utmost importance in order to maximise treatment efficacy and minimise side-effects and costs. Moreover, determining biomarkers predicting responses may provide insight into the mechanisms responsible for success or failure of that therapy. This article reviews seminal papers mostly from the past two years of progress in this area of intense investigation, and mostly in melanoma, the tumour type for which the largest body of data exists thus far.
Collapse
|
280
|
Wei MM, Wang YS, Ye XS. Carbohydrate-based vaccines for oncotherapy. Med Res Rev 2018; 38:1003-1026. [PMID: 29512174 DOI: 10.1002/med.21493] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/18/2018] [Accepted: 01/31/2018] [Indexed: 01/02/2023]
Abstract
Cancer is still one of the most serious threats to human worldwide. Aberrant patterns of glycosylation on the surface of cancer cells, which are correlated with various cancer development stages, can differentiate the abnormal tissues from the healthy ones. Therefore, tumor-associated carbohydrate antigens (TACAs) represent the desired targets for cancer immunotherapy. However, these carbohydrate antigens may not able to evoke powerful immune response to combat with cancer for their poor immunogenicity and immunotolerance. Different approaches have been developed to address these problems. In this review, we want to summarize the latest advances in TACAs based anticancer vaccines.
Collapse
Affiliation(s)
- Meng-Man Wei
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yong-Shi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
281
|
Kiyotani K, Chan HT, Nakamura Y. Immunopharmacogenomics towards personalized cancer immunotherapy targeting neoantigens. Cancer Sci 2018; 109:542-549. [PMID: 29288513 PMCID: PMC5834780 DOI: 10.1111/cas.13498] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/23/2017] [Accepted: 12/25/2017] [Indexed: 12/15/2022] Open
Abstract
Utilizing the host immune system to eradicate cancer cells has been the most investigated subject in the cancer research field in recent years. However, most of the studies have focused on highly variable responses from immunotherapies such as immune checkpoint inhibitors, from which the majority of patients experienced no or minimum clinical benefit. Advances in genomic sequencing technologies have improved our understanding of immunopharmacogenomics and allowed us to identify novel cancer-specific immune targets. Highly tumor-specific antigens, neoantigens, are generated by somatic mutations that are not present in normal cells. It is plausible that by targeting antigens with high tumor-specificity, such as neoantigens, the likelihood of toxic effects is very limited. However, understanding the interaction between neoantigens and the host immune system remains a significant challenge. This review focuses on the potential use of neoantigen-targeted immunotherapies in cancer treatment and the recent progress of different strategies in predicting, identifying, and validating neoantigens. Successful identification of highly tumor-specific antigens accelerates the development of personalized immunotherapy with no or minimum adverse effects and with a broader coverage of patients.
Collapse
Affiliation(s)
- Kazuma Kiyotani
- Project for ImmunogenomicsCancer Precision Medicine CenterJapanese Foundation for Cancer ResearchTokyoJapan
| | - Hiu Ting Chan
- Project for ImmunogenomicsCancer Precision Medicine CenterJapanese Foundation for Cancer ResearchTokyoJapan
| | - Yusuke Nakamura
- Department of MedicineThe University of ChicagoChicagoILUSA
- Department of SurgeryThe University of ChicagoChicagoILUSA
| |
Collapse
|
282
|
Ping Y, Liu C, Zhang Y. T-cell receptor-engineered T cells for cancer treatment: current status and future directions. Protein Cell 2018; 9:254-266. [PMID: 28108950 PMCID: PMC5829268 DOI: 10.1007/s13238-016-0367-1] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022] Open
Abstract
T-cell receptor (TCR)-engineered T cells are a novel option for adoptive cell therapy used for the treatment of several advanced forms of cancer. Work using TCR-engineered T cells began more than two decades ago, with numerous preclinical studies showing that such cells could mediate tumor lysis and eradication. The success of these trials provided the foundation for clinical trials, including recent clinical successes using TCR-engineered T cells to target New York esophageal squamous cell carcinoma (NY-ESO-1). These successes demonstrate the potential of this approach to treat cancer. In this review, we provide a perspective on the current and future applications of TCR-engineered T cells for the treatment of cancer. Our summary focuses on TCR activation and both pre-clinical and clinical applications of TCR-engineered T cells. We also discuss how to enhance the function of TCR-engineered T cells and prolong their longevity in the tumor microenvironment.
Collapse
Affiliation(s)
- Yu Ping
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chaojun Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, 450052, China.
| |
Collapse
|
283
|
Lee EJ, Nam GH, Lee NK, Kih M, Koh E, Kim YK, Hong Y, Kim S, Park SY, Jeong C, Yang Y, Kim IS. Nanocage-Therapeutics Prevailing Phagocytosis and Immunogenic Cell Death Awakens Immunity against Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:1705581. [PMID: 29333661 DOI: 10.1002/adma.201705581] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/18/2017] [Indexed: 06/07/2023]
Abstract
A growing appreciation of the relationship between the immune system and the tumorigenesis has led to the development of strategies aimed at "re-editing" the immune system to kill tumors. Here, a novel tactic is reported for overcoming the activation-energy threshold of the immunosuppressive tumor microenvironment and mediating the delivery and presentation of tumor neoantigens to the host's immune system. This nature-derived nanocage not only efficiently presents ligands that enhance cancer cell phagocytosis, but also delivers drugs that induce immunogenic cancer cell death. The designed nanocage-therapeutics induce the release of neoantigens and danger signals in dying tumor cells, and leads to enhancement of tumor cell phagocytosis and cross-priming of tumor specific T cells by neoantigen peptide-loaded antigen-presenting cells. Potent inhibition of tumor growth and complete eradication of tumors is observed through systemic tumor-specific T cell responses in tumor draining lymph nodes and the spleen and further, infiltration of CD8+ T cells into the tumor site. Remarkably, after removal of the primary tumor, all mice treated with this nanocage-therapeutics are protected against subsequent challenge with the same tumor cells, suggesting development of lasting, tumor-specific responses. This designed nanocage-therapeutics "awakens" the host's immune system and provokes a durable systemic immune response against cancer.
Collapse
Affiliation(s)
- Eun Jung Lee
- Department of Chemical Engineering, School of Applied Chemical Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Gi-Hoon Nam
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Na Kyeong Lee
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Minwoo Kih
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Eunee Koh
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Yoon Kyoung Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Yeonsun Hong
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Soyoun Kim
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Seung-Yoon Park
- Department of Biochemistry, School of Medicine, Dongguk University, Gyeongju, 38066, Republic of Korea
| | - Cherlhyun Jeong
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yoosoo Yang
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
284
|
Sioud M. T-cell cross-reactivity may explain the large variation in how cancer patients respond to checkpoint inhibitors. Scand J Immunol 2018; 87. [DOI: 10.1111/sji.12643] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 01/23/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Mouldy Sioud
- Department of Cancer Immunology; Oslo University Hospital; The Norwegian Radium Hospital; Montebello Oslo Norway
| |
Collapse
|
285
|
Brown JS, Sundar R, Lopez J. Combining DNA damaging therapeutics with immunotherapy: more haste, less speed. Br J Cancer 2018; 118:312-324. [PMID: 29123260 PMCID: PMC5808021 DOI: 10.1038/bjc.2017.376] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 09/04/2017] [Accepted: 09/04/2017] [Indexed: 12/14/2022] Open
Abstract
The idea that chemotherapy can be used in combination with immunotherapy may seem somewhat counterproductive, as it can theoretically eliminate the immune cells needed for antitumour immunity. However, much preclinical work has now demonstrated that in addition to direct cytotoxic effects on cancer cells, a proportion of DNA damaging agents may actually promote immunogenic cell death, alter the inflammatory milieu of the tumour microenvironment and/or stimulate neoantigen production, thereby activating an antitumour immune response. Some notable combinations have now moved forward into the clinic, showing promise in phase I-III trials, whereas others have proven toxic, and challenging to deliver. In this review, we discuss the emerging data of how DNA damaging agents can enhance the immunogenic properties of malignant cells, focussing especially on immunogenic cell death, and the expansion of neoantigen repertoires. We discuss how best to strategically combine DNA damaging therapeutics with immunotherapy, and the challenges of successfully delivering these combination regimens to patients. With an overwhelming number of chemotherapy/immunotherapy combination trials in process, clear hypothesis-driven trials are needed to refine the choice of combinations, and determine the timing and sequencing of agents in order to stimulate antitumour immunological memory and improve maintained durable response rates, with minimal toxicity.
Collapse
Affiliation(s)
- Jessica S Brown
- Royal Marsden NHS Foundation Trust, Downs Road, London SM2 5PT, UK
| | - Raghav Sundar
- Royal Marsden NHS Foundation Trust, Downs Road, London SM2 5PT, UK
- Department of Haematology-Oncology, National University Health System, Singapore
| | - Juanita Lopez
- Royal Marsden NHS Foundation Trust, Downs Road, London SM2 5PT, UK
- The Institute of Cancer Research, London SM2 5NG, UK
| |
Collapse
|
286
|
Cheung AS, Zhang DK, Koshy ST, Mooney DJ. Scaffolds that mimic antigen-presenting cells enable ex vivo expansion of primary T cells. Nat Biotechnol 2018; 36:160-169. [PMID: 29334370 PMCID: PMC5801009 DOI: 10.1038/nbt.4047] [Citation(s) in RCA: 264] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 12/01/2017] [Indexed: 12/22/2022]
Abstract
Therapeutic ex vivo T-cell expansion is limited by low rates and T-cell products of limited functionality. Here we describe a system that mimics natural antigen-presenting cells (APCs) and consists of a fluid lipid bilayer supported by mesoporous silica micro-rods. The lipid bilayer presents membrane-bound cues for T-cell receptor stimulation and costimulation, while the micro-rods enable sustained release of soluble paracrine cues. Using anti-CD3, anti-CD28, and interleukin-2, we show that the APC-mimetic scaffolds (APC-ms) promote two- to tenfold greater polyclonal expansion of primary mouse and human T cells compared with commercial expansion beads (Dynabeads). The efficiency of expansion depends on the density of stimulatory cues and the amount of material in the starting culture. Following a single stimulation, APC-ms enables antigen-specific expansion of rare cytotoxic T-cell subpopulations at a greater magnitude than autologous monocyte-derived dendritic cells after 2 weeks. APC-ms support over fivefold greater expansion of restimulated CD19 CAR-T cells than Dynabeads, with similar efficacy in a xenograft lymphoma model.
Collapse
Affiliation(s)
- Alexander S. Cheung
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- The Wyss Institute for Biologically Inspired Engineering Harvard University, Cambridge, Massachusetts, USA
| | - David K.Y. Zhang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- The Wyss Institute for Biologically Inspired Engineering Harvard University, Cambridge, Massachusetts, USA
| | - Sandeep T. Koshy
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- The Wyss Institute for Biologically Inspired Engineering Harvard University, Cambridge, Massachusetts, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, USA
| | - David J. Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- The Wyss Institute for Biologically Inspired Engineering Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
287
|
Gee MH, Han A, Lofgren SM, Beausang JF, Mendoza JL, Birnbaum ME, Bethune MT, Fischer S, Yang X, Gomez-Eerland R, Bingham DB, Sibener LV, Fernandes RA, Velasco A, Baltimore D, Schumacher TN, Khatri P, Quake SR, Davis MM, Garcia KC. Antigen Identification for Orphan T Cell Receptors Expressed on Tumor-Infiltrating Lymphocytes. Cell 2018; 172:549-563.e16. [PMID: 29275860 PMCID: PMC5786495 DOI: 10.1016/j.cell.2017.11.043] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/30/2017] [Accepted: 11/22/2017] [Indexed: 12/30/2022]
Abstract
The immune system can mount T cell responses against tumors; however, the antigen specificities of tumor-infiltrating lymphocytes (TILs) are not well understood. We used yeast-display libraries of peptide-human leukocyte antigen (pHLA) to screen for antigens of "orphan" T cell receptors (TCRs) expressed on TILs from human colorectal adenocarcinoma. Four TIL-derived TCRs exhibited strong selection for peptides presented in a highly diverse pHLA-A∗02:01 library. Three of the TIL TCRs were specific for non-mutated self-antigens, two of which were present in separate patient tumors, and shared specificity for a non-mutated self-antigen derived from U2AF2. These results show that the exposed recognition surface of MHC-bound peptides accessible to the TCR contains sufficient structural information to enable the reconstruction of sequences of peptide targets for pathogenic TCRs of unknown specificity. This finding underscores the surprising specificity of TCRs for their cognate antigens and enables the facile indentification of tumor antigens through unbiased screening.
Collapse
Affiliation(s)
- Marvin H Gee
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arnold Han
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shane M Lofgren
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John F Beausang
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Juan L Mendoza
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael E Birnbaum
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael T Bethune
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Suzanne Fischer
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinbo Yang
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Raquel Gomez-Eerland
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - David B Bingham
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leah V Sibener
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ricardo A Fernandes
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew Velasco
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ton N Schumacher
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Purvesh Khatri
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stephen R Quake
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Mark M Davis
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; The Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - K Christopher Garcia
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; The Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
288
|
Rech AJ, Balli D, Mantero A, Ishwaran H, Nathanson KL, Stanger BZ, Vonderheide RH. Tumor Immunity and Survival as a Function of Alternative Neopeptides in Human Cancer. Cancer Immunol Res 2018; 6:276-287. [PMID: 29339376 DOI: 10.1158/2326-6066.cir-17-0559] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 11/27/2017] [Accepted: 01/05/2018] [Indexed: 12/15/2022]
Abstract
The immune system exerts antitumor activity via T cell-dependent recognition of tumor-specific antigens. Although the number of tumor neopeptides-peptides derived from somatic mutations-often correlates with immune activity and survival, most classically defined high-affinity neopeptides (CDNs) are not immunogenic, and only rare CDNs have been linked to tumor rejection. Thus, the rules of tumor antigen recognition remain incompletely understood. Here, we analyzed neopeptides, immune activity, and clinical outcome from 6,324 patients across 27 tumor types. We characterized a class of "alternatively defined neopeptides" (ADNs), which are mutant peptides predicted to bind MHC (class I or II) with improved affinity relative to their nonmutated counterpart. ADNs are abundant and molecularly distinct from CDNs. The load of ADNs correlated with intratumoral T-cell responses and immune suppression, and ADNs were also strong predictors of patient survival across tumor types. These results expand the spectrum of mutation-derived tumor antigens with potential clinical relevance. Cancer Immunol Res; 6(3); 276-87. ©2018 AACR.
Collapse
Affiliation(s)
- Andrew J Rech
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Balli
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alejandro Mantero
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miami, Florida
| | - Hemant Ishwaran
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miami, Florida
| | - Katherine L Nathanson
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ben Z Stanger
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Vonderheide
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania. .,Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
289
|
Kato T, Matsuda T, Ikeda Y, Park JH, Leisegang M, Yoshimura S, Hikichi T, Harada M, Zewde M, Sato S, Hasegawa K, Kiyotani K, Nakamura Y. Effective screening of T cells recognizing neoantigens and construction of T-cell receptor-engineered T cells. Oncotarget 2018. [PMID: 29541393 PMCID: PMC5834292 DOI: 10.18632/oncotarget.24232] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Neoantigens are the main targets of tumor-specific T cells reactivated by immune checkpoint-blocking antibodies or when using tumor-infiltrating T cells for adoptive therapy. While cancers often accumulate hundreds of mutations and harbor several immunogenic neoantigens, the repertoire of mutation-specific T cells in patients might be restricted. To bypass suboptimal conditions, which impede the reactivation of existing T cells or the priming of neoantigen-specific T cells in a patient, we employ T cells of healthy donors with an overlapping HLA repertoire to target cancer neoantigens. In this study, we focus on streamlining the process of in vitro-induction of neoantigen-specific T cells and isolating their T cell receptors (TCRs) to establish a time-efficient protocol that will allow the patient to benefit from subsequent therapy. We first optimized the priming of T cells to omit multiple restimulations and extended culturing. Neoantigen-specific T cells were enriched using specific dextramers and next-generation sequencing was applied to determine the TCR repertoire. This allowed us to circumvent the laborious process of expanding T cell clones. Using this protocol, we successfully identified HLA-A-restricted TCRs specific for neoantigens found in an esophageal cancer cell line (TE-8) and a primary ovarian cancer. To verify TCR specificity, we generated TCR-engineered T cells and confirmed recognition of the tumor-derived neoantigens. Our results also emphasize the importance of neoepitope selection in order to avoid cross-reactivity to corresponding wild-type peptide sequences. In conclusion, we established a 2-week protocol for generating and identifying neoantigen-specific TCRs from third-party donors making this strategy applicable for clinical use.
Collapse
Affiliation(s)
- Taigo Kato
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Tatsuo Matsuda
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Yuji Ikeda
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Jae-Hyun Park
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Matthias Leisegang
- Institute of Immunology - Campus Buch, Charité - Universitätsmedizin Berlin, Berlin 13125, Germany.,Berlin Institute of Health, Berlin 10117, Germany
| | | | - Tetsuro Hikichi
- OncoTherapy Science Inc., Kawasaki, Kanagawa 213-0012, Japan
| | - Makiko Harada
- OncoTherapy Science Inc., Kawasaki, Kanagawa 213-0012, Japan
| | - Makda Zewde
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Sho Sato
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama 350-1298, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama 350-1298, Japan
| | - Kazuma Kiyotani
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Yusuke Nakamura
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA.,Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
290
|
Wang M, Yao LC, Cheng M, Cai D, Martinek J, Pan CX, Shi W, Ma AH, De Vere White RW, Airhart S, Liu ET, Banchereau J, Brehm MA, Greiner DL, Shultz LD, Palucka K, Keck JG. Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy. FASEB J 2018; 32:1537-1549. [PMID: 29146734 PMCID: PMC5892726 DOI: 10.1096/fj.201700740r] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Establishment of an in vivo small animal model of human tumor and human immune system interaction would enable preclinical investigations into the mechanisms underlying cancer immunotherapy. To this end, nonobese diabetic (NOD).Cg-PrkdcscidIL2rgtm1Wjl/Sz (null; NSG) mice were transplanted with human (h)CD34+ hematopoietic progenitor and stem cells, which leads to the development of human hematopoietic and immune systems [humanized NSG (HuNSG)]. HuNSG mice received human leukocyte antigen partially matched tumor implants from patient-derived xenografts [PDX; non–small cell lung cancer (NSCLC), sarcoma, bladder cancer, and triple-negative breast cancer (TNBC)] or from a TNBC cell line-derived xenograft (CDX). Tumor growth curves were similar in HuNSG compared with nonhuman immune-engrafted NSG mice. Treatment with pembrolizumab, which targets programmed cell death protein 1, produced significant growth inhibition in both CDX and PDX tumors in HuNSG but not in NSG mice. Finally, inhibition of tumor growth was dependent on hCD8+ T cells, as demonstrated by antibody-mediated depletion. Thus, tumor-bearing HuNSG mice may represent an important, new model for preclinical immunotherapy research.—Wang, M., Yao, L.-C., Cheng, M., Cai, D., Martinek, J., Pan, C.-X., Shi, W., Ma, A.-H., De Vere White, R. W., Airhart, S., Liu, E. T., Banchereau, J., Brehm, M. A., Greiner, D. L., Shultz, L. D., Palucka, K., Keck, J. G. Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy.
Collapse
Affiliation(s)
- Minan Wang
- Department of In Vivo Pharmacology Services, The Jackson Laboratory, Sacramento, California, USA
| | - Li-Chin Yao
- Department of In Vivo Pharmacology Services, The Jackson Laboratory, Sacramento, California, USA
| | - Mingshan Cheng
- Department of In Vivo Pharmacology Services, The Jackson Laboratory, Sacramento, California, USA
| | - Danying Cai
- Department of In Vivo Pharmacology Services, The Jackson Laboratory, Sacramento, California, USA
| | - Jan Martinek
- Department of Immunology, The Jackson Laboratory, Farmington, Connecticut, USA
| | - Chong-Xian Pan
- Department of Urology, Davis Comprehensive Cancer Center, University of California, Davis, California, USA
| | - Wei Shi
- Department of Urology, Davis Comprehensive Cancer Center, University of California, Davis, California, USA
| | - Ai-Hong Ma
- Department of Biochemistry and Molecular Medicine, Davis Comprehensive Cancer Center, University of California, Davis, Davis, California, USA
| | - Ralph W De Vere White
- Department of Urology, Davis Comprehensive Cancer Center, University of California, Davis, California, USA
| | - Susan Airhart
- Department of Immunology, The Jackson Laboratory, Farmington, Connecticut, USA
| | - Edison T Liu
- Department of Immunology, The Jackson Laboratory, Farmington, Connecticut, USA
| | - Jacques Banchereau
- Department of Immunology, The Jackson Laboratory, Farmington, Connecticut, USA
| | - Michael A Brehm
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts, USA; and
| | - Dale L Greiner
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts, USA; and
| | - Leonard D Shultz
- Department of Immunology, The Jackson Laboratory, Bar Harbor, Maine, USA
| | - Karolina Palucka
- Department of Immunology, The Jackson Laboratory, Farmington, Connecticut, USA
| | - James G Keck
- Department of In Vivo Pharmacology Services, The Jackson Laboratory, Sacramento, California, USA
| |
Collapse
|
291
|
Dendrou CA, Petersen J, Rossjohn J, Fugger L. HLA variation and disease. Nat Rev Immunol 2018; 18:325-339. [PMID: 29292391 DOI: 10.1038/nri.2017.143] [Citation(s) in RCA: 491] [Impact Index Per Article: 70.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fifty years since the first description of an association between HLA and human disease, HLA molecules have proven to be central to physiology, protective immunity and deleterious, disease-causing autoimmune reactivity. Technological advances have enabled pivotal progress in the determination of the molecular mechanisms that underpin the association between HLA genetics and functional outcome. Here, we review our current understanding of HLA molecules as the fundamental platform for immune surveillance and responsiveness in health and disease. We evaluate the scope for personalized antigen-specific disease prevention, whereby harnessing HLA-ligand interactions for clinical benefit is becoming a realistic prospect.
Collapse
Affiliation(s)
- Calliope A Dendrou
- Nuffield Department of Medicine, The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Jan Petersen
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Wellington Road, Clayton, Victoria 3800, Australia.,Infection and Immunity Programme and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Jamie Rossjohn
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Wellington Road, Clayton, Victoria 3800, Australia.,Infection and Immunity Programme and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, Victoria 3800, Australia.,Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Lars Fugger
- Danish National Research Foundation Centre PERSIMUNE, Rigshospitalet, University of Copenhagen, Copenhagen DK-2100, Denmark.,Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology and Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DS, UK
| |
Collapse
|
292
|
Added Value of Whole-Exome and Transcriptome Sequencing for Clinical Molecular Screenings of Advanced Cancer Patients With Solid Tumors. Cancer J 2018; 24:153-162. [DOI: 10.1097/ppo.0000000000000322] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
293
|
Pawelec G. Immune correlates of clinical outcome in melanoma. Immunology 2017; 153:415-422. [PMID: 29164593 DOI: 10.1111/imm.12870] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/07/2017] [Accepted: 11/14/2017] [Indexed: 12/14/2022] Open
Abstract
Melanoma has long been recognized as a potentially immunogenic tumour, but only recently has it become clear that the reason for this resides in its many ultraviolet (UV)-induced mutations and expression of multiple autoantigens which can be targeted by the immune system. The first successful applications of immune-based treatments included passive immunotherapy using high-dose interleukin (IL)-2 and/or adoptive transfer of natural killer (NK)-cells, as well as active immunotherapy using whole cell-derived or peptide vaccines. In the intervening decades, it has become clear that these approaches can lead to durable responses in stage III/IV melanoma, and even to functional cures - but only in a vanishingly small fraction of patients. With the advent of immune checkpoint blockade first with anti-cytotoxic T-lymphocyte 4 (CTLA-4), then with anti-programmed cell death 1 (PD-1) antibodies, and combinations thereof, the small percentage of responding patients may be increased to half, a major accomplishment in this refractory disease. Improved techniques for identifying mutation-derived neoantigens and thus more sophisticated active immunotherapies, probably combined with checkpoint blockade, currently hold great promise for further increasing the fraction of responding patients. As additional immunomodulatory antibodies and therapies become available, it will be increasingly important to develop diagnostic tools to determine which particular therapy is likely to elicit the best response for the individual patient. Practically speaking, therapy selection and efficacy monitoring on the basis of the results of a blood test would be most desirable. The purpose of this review is to consider the feasibility of identifying 'immune signatures' for predicting responses and determining mechanisms responsible for success or failure of these immunotherapies.
Collapse
Affiliation(s)
- Graham Pawelec
- Second Department of Internal Medicine, University of Tuebingen Center for Medical Research (ZMF), Tuebingen, Germany.,Division of Cancer Studies, King's College London, London, UK.,John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK.,Health Sciences North Research Institute, Sudbury, ON, Canada
| |
Collapse
|
294
|
Wirth TC, Kühnel F. Neoantigen Targeting-Dawn of a New Era in Cancer Immunotherapy? Front Immunol 2017; 8:1848. [PMID: 29312332 PMCID: PMC5742119 DOI: 10.3389/fimmu.2017.01848] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/06/2017] [Indexed: 12/30/2022] Open
Abstract
During their development and progression tumors acquire numerous mutations that, when translated into proteins give rise to neoantigens that can be recognized by T cells. Initially, neoantigens were not recognized as preferred targets for cancer immunotherapy due to their enormous diversity and the therefore limited options to develop “one fits all” pharmacologic solutions. In recent years, the experience obtained in clinical trials demonstrating a predictive role of neoantigens in checkpoint inhibition has changed our view on the clinical potential of neoantigens in cancer immunotherapy. Technological advances such as sequencing of whole cancer genomes, the development of reliable algorithms for epitope prediction, and an increasing number of immunotherapeutic options now facilitate the development of personalized tumor therapies directly targeting a patient’s neoantigenic burden. Preclinical studies in mice that support the excellent therapeutic potential of neoantigen-directed immunotherapies have provided blueprints on how this methodology can be translated into clinical applications in humans. Consistently, very recent clinical studies on personalized vaccinations targeting in silico predicted neoepitopes shed a first light on the therapeutic potential of personalized, neoantigen-directed immunotherapies. In our review, we discuss the various subtypes of tumor antigens with a focus on neoantigens and their potential in cancer immunotherapy. We will describe the current methods and techniques of detection as well as the structural requirements for neoantigens that are needed for their recognition by T cells and for tumor destruction. To assess the clinical potential of neoantigens, we will discuss their occurrence and functional relevance in spontaneous and hereditary cancers and their prognostic and predictive value. We will present in detail the existing immunotherapeutic options that exploit the neoantigen burden of tumors encompassing both preclinical efforts that provided convincing technological proof-of-concept and the current clinical studies confirming the potential of neoantigen-directed immunotherapies.
Collapse
Affiliation(s)
- Thomas C Wirth
- Clinic for Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Hannover, Germany
| | - Florian Kühnel
- Clinic for Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Hannover, Germany
| |
Collapse
|
295
|
Rao M, Zhenjiang L, Meng Q, Sinclair G, Dodoo E, Maeurer M. Mutant Epitopes in Cancer. Oncoimmunology 2017. [DOI: 10.1007/978-3-319-62431-0_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
296
|
Karpanen T, Olweus J. The Potential of Donor T-Cell Repertoires in Neoantigen-Targeted Cancer Immunotherapy. Front Immunol 2017; 8:1718. [PMID: 29321773 PMCID: PMC5732232 DOI: 10.3389/fimmu.2017.01718] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/21/2017] [Indexed: 12/30/2022] Open
Abstract
T cells can recognize peptides encoded by mutated genes, but analysis of tumor-infiltrating lymphocytes suggests that very few neoantigens spontaneously elicit T-cell responses. This may be an important reason why immune checkpoint inhibitors are mainly effective in tumors with a high mutational burden. Reasons for clinically insufficient responses to neoantigens might be inefficient priming, inhibition, or deletion of the cognate T cells. Responses can be dramatically improved by cancer immunotherapy such as checkpoint inhibition, but often with temporary effects. By contrast, T cells from human leukocyte antigen (HLA)-matched donors can cure diseases such as chronic myeloid leukemia. The therapeutic effect is mediated by donor T cells recognizing polymorphic peptides for which the donor and patient are disparate, presented on self-HLA. Donor T-cell repertoires are unbiased by the immunosuppressive environment of the tumor. A recent study demonstrated that T cells from healthy individuals are able to respond to neoantigens that are ignored by tumor-infiltrating T cells of melanoma patients. In this review, we discuss possible reasons why neoantigens escape host T cells and how these limitations may be overcome by utilization of donor-derived T-cell repertoires to facilitate rational design of neoantigen-targeted immunotherapy.
Collapse
Affiliation(s)
- Terhi Karpanen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, and K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| | - Johanna Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, and K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| |
Collapse
|
297
|
Bethune MT, Joglekar AV. Personalized T cell-mediated cancer immunotherapy: progress and challenges. Curr Opin Biotechnol 2017; 48:142-152. [DOI: 10.1016/j.copbio.2017.03.024] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/15/2017] [Accepted: 03/19/2017] [Indexed: 12/26/2022]
|
298
|
Bräunlein E, Krackhardt AM. Identification and Characterization of Neoantigens As Well As Respective Immune Responses in Cancer Patients. Front Immunol 2017; 8:1702. [PMID: 29250075 PMCID: PMC5714868 DOI: 10.3389/fimmu.2017.01702] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/17/2017] [Indexed: 12/16/2022] Open
Abstract
Cancer immunotherapy has recently emerged as a powerful tool for the treatment of diverse advanced malignancies. In particular, therapeutic application of immune checkpoint modulators, such as anti-CTLA4 or anti-PD-1/PD-L1 antibodies, have shown efficacy in a broad range of malignant diseases. Although pharmacodynamics of these immune modulators are complex, recent studies strongly support the notion that altered peptide ligands presented on tumor cells representing neoantigens may play an essential role in tumor rejection by T cells activated by anti-CTLA4 and anti-PD-1 antibodies. Neoantigens may have diverse sources as viral and mutated proteins. Moreover, posttranslational modifications and altered antigen processing may also contribute to the neoantigenic peptide ligand landscape. Different approaches of target identification are currently applied in combination with subsequent characterization of autologous and non-self T-cell responses against such neoantigens. Additional efforts are required to elucidate key characteristics and interdependences of neoantigens, immunodominance, respective T-cell responses, and the tumor microenvironment in order to define decisive determinants involved in effective T-cell-mediated tumor rejection. This review focuses on our current knowledge of identification and characterization of such neoantigens as well as respective T-cell responses. It closes with challenges to be addressed in future relevant for further improvement of immunotherapeutic strategies in malignant diseases.
Collapse
Affiliation(s)
- Eva Bräunlein
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Angela M Krackhardt
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,German Cancer Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
299
|
Stevanović S, Pasetto A, Helman SR, Gartner JJ, Prickett TD, Howie B, Robins HS, Robbins PF, Klebanoff CA, Rosenberg SA, Hinrichs CS. Landscape of immunogenic tumor antigens in successful immunotherapy of virally induced epithelial cancer. Science 2017; 356:200-205. [PMID: 28408606 DOI: 10.1126/science.aak9510] [Citation(s) in RCA: 295] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 03/16/2017] [Indexed: 12/11/2022]
Abstract
Immunotherapy has clinical activity in certain virally associated cancers. However, the tumor antigens targeted in successful treatments remain poorly defined. We used a personalized immunogenomic approach to elucidate the global landscape of antitumor T cell responses in complete regression of human papillomavirus-associated metastatic cervical cancer after tumor-infiltrating adoptive T cell therapy. Remarkably, immunodominant T cell reactivities were directed against mutated neoantigens or a cancer germline antigen, rather than canonical viral antigens. T cells targeting viral tumor antigens did not display preferential in vivo expansion. Both viral and nonviral tumor antigen-specific T cells resided predominantly in the programmed cell death 1 (PD-1)-expressing T cell compartment, which suggests that PD-1 blockade may unleash diverse antitumor T cell reactivities. These findings suggest a new paradigm of targeting nonviral antigens in immunotherapy of virally associated cancers.
Collapse
Affiliation(s)
- Sanja Stevanović
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, MD 20892, USA.
| | - Anna Pasetto
- Surgery Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Sarah R Helman
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Jared J Gartner
- Surgery Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Todd D Prickett
- Surgery Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Bryan Howie
- Adaptive Biotechnologies, Seattle, WA 98102, USA
| | - Harlan S Robins
- Adaptive Biotechnologies, Seattle, WA 98102, USA.,Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Paul F Robbins
- Surgery Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Christopher A Klebanoff
- Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Parker Institute for Cancer Immunotherapy, New York, NY 10065, USA
| | | | - Christian S Hinrichs
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
300
|
Martincorena I, Raine KM, Gerstung M, Dawson KJ, Haase K, Van Loo P, Davies H, Stratton MR, Campbell PJ. Universal Patterns of Selection in Cancer and Somatic Tissues. Cell 2017; 171:1029-1041.e21. [PMID: 29056346 PMCID: PMC5720395 DOI: 10.1016/j.cell.2017.09.042] [Citation(s) in RCA: 894] [Impact Index Per Article: 111.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/09/2017] [Accepted: 09/22/2017] [Indexed: 01/17/2023]
Abstract
Cancer develops as a result of somatic mutation and clonal selection, but quantitative measures of selection in cancer evolution are lacking. We adapted methods from molecular evolution and applied them to 7,664 tumors across 29 cancer types. Unlike species evolution, positive selection outweighs negative selection during cancer development. On average, <1 coding base substitution/tumor is lost through negative selection, with purifying selection almost absent outside homozygous loss of essential genes. This allows exome-wide enumeration of all driver coding mutations, including outside known cancer genes. On average, tumors carry ∼4 coding substitutions under positive selection, ranging from <1/tumor in thyroid and testicular cancers to >10/tumor in endometrial and colorectal cancers. Half of driver substitutions occur in yet-to-be-discovered cancer genes. With increasing mutation burden, numbers of driver mutations increase, but not linearly. We systematically catalog cancer genes and show that genes vary extensively in what proportion of mutations are drivers versus passengers.
Collapse
Affiliation(s)
| | - Keiran M Raine
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridgeshire, UK
| | - Moritz Gerstung
- European Molecular Biology Laboratory, European Bioinformatics Institute EMBL-EBI, Hinxton CB10 1SD, UK
| | - Kevin J Dawson
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridgeshire, UK
| | | | - Peter Van Loo
- The Francis Crick Institute, London NW1 1AT, UK; Department of Human Genetics, University of Leuven, Leuven 3000, Belgium
| | - Helen Davies
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridgeshire, UK
| | | | - Peter J Campbell
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, Cambridgeshire, UK; Department of Haematology, University of Cambridge, Cambridge CB2 2XY, UK.
| |
Collapse
|