1
|
Okamura K, Wang L, Nagayama S, Yamashita M, Tate T, Matsumoto S, Takamatsu M, Kitano S, Kiyotani K, Nakamura Y. Characterization of double-negative T cells in colorectal cancers and their corresponding lymph nodes. Oncoimmunology 2024; 13:2373530. [PMID: 38979545 PMCID: PMC11229752 DOI: 10.1080/2162402x.2024.2373530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024] Open
Abstract
TCRαβ+ CD4- CD8- double-negative T (DNT) cells are minor populations in peripheral blood, and their roles have mostly been discussed in inflammation and autoimmunity. However, the functions of DNT cells in tumor microenvironment remain to be elucidated. We investigated their characteristics, possible origins and functions in colorectal cancer tissues as well as their corresponding tumor-draining lymph nodes. We found a significant enrichment of DNT cells in tumor tissues compared with their corresponding lymph nodes, especially in tumors with lower T cell infiltration. T cell receptor (TCR) sequence analysis of CD4+ T, CD8+ T and DNT cells indicated that TCR sequences detected in DNT cells were found in CD8+ T cells, but rarely in CD4+ T cells, suggesting that a part of DNT cells was likely to be originated from CD8+ T cells. Through a single-cell transcriptomic analysis of DNT cells, we found that a DNT cell cluster, which showed similar phenotypes to central memory CD8+ T cells with low expression of effector and exhaustion markers, revealed some specific gene expression patterns, including higher GZMK expression. Moreover, in flow cytometry analysis, we found that DNT cells lost production of cytotoxic mediators. These findings imply that DNT cells might function as negative regulators of anti-tumor immune responses in tumor microenvironment.
Collapse
Affiliation(s)
- Kazumi Okamura
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Lifang Wang
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satoshi Nagayama
- Department of Gastroenterological and Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Depart of Surgery, Uji-Tokusyukai Medical Center, Uji-shi, Kyoto, Japan
| | - Makiko Yamashita
- Division of Cancer Immunotherapy Development, Department of Advanced Medical Development, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomohiro Tate
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Saki Matsumoto
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Manabu Takamatsu
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shigehisa Kitano
- Division of Cancer Immunotherapy Development, Department of Advanced Medical Development, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kazuma Kiyotani
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
- Laboratory of Immunogenomics, Center for Intractable Diseases and ImmunoGenomics (CiDIG), National Institute of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki-shi, Osaka, Japan
| | - Yusuke Nakamura
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
- Laboratory of Immunogenomics, Center for Intractable Diseases and ImmunoGenomics (CiDIG), National Institute of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki-shi, Osaka, Japan
| |
Collapse
|
2
|
Fert I, Douguet L, Vesin B, Moncoq F, Noirat A, Authié P, Ciret S, Le Chevalier F, Blanc C, Vitrenko Y, Charneau P, Majlessi L, Anna F. T-cell immunity induced and reshaped by an anti-HPV immuno-oncotherapeutic lentiviral vector. NPJ Vaccines 2024; 9:102. [PMID: 38858404 PMCID: PMC11164992 DOI: 10.1038/s41541-024-00894-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 05/23/2024] [Indexed: 06/12/2024] Open
Abstract
We recently developed an immuno-oncotherapy against human papillomavirus (HPV)-induced tumors based on a lentiviral vector encoding the Early E6 and E7 oncoproteins of HPV16 and HPV18 genotypes, namely "Lenti-HPV-07". The robust and long-lasting anti-tumor efficacy of Lenti-HPV-07 is dependent on CD8+ T-cell induction and remodeling of the tumor microenvironment. Here, we first established that anti-vector immunity induced by Lenti-HPV-07 prime has no impact on the efficacy of a homologous boost to amplify anti-HPV T-cell immunity. To longitudinally monitor the evolution of the T-cell repertoire generated after the prime, homologous or heterologous boost with Lenti-HPV-07, we tracked T-cell clonotypes by deep sequencing of T-Cell Receptor (TCR) variable β and α chain mRNA, applied to whole peripheral blood cells (PBL) and a T cell population specific of an immunodominant E7HPV16 epitope. We observed a hyper-expansion of clonotypes post prime, accompanied by increased frequencies of HPV-07-specific T cells. Additionally, there was a notable diversification of clonotypes post boost in whole PBL, but not in the E7HPV16-specific T cells. We then demonstrated that the effector functions of such Lenti-HPV-07-induced T cells synergize with anti-checkpoint inhibitory treatments by systemic administration of anti-TIM3 or anti-NKG2A monoclonal antibodies. While Lenti-HPV-07 is about to enter a Phase I/IIa clinical trial, these results will help better elucidate its mode of action in immunotherapy against established HPV-mediated malignancies.
Collapse
Affiliation(s)
- Ingrid Fert
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Laëtitia Douguet
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Benjamin Vesin
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Fanny Moncoq
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Amandine Noirat
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Pierre Authié
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Sylvain Ciret
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Fabien Le Chevalier
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Catherine Blanc
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Yakov Vitrenko
- Institut Pasteur, Université Paris Cité, Biomics Technology Platform, F-75015, Paris, France
| | - Pierre Charneau
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Laleh Majlessi
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France.
| | - François Anna
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France.
| |
Collapse
|
3
|
FAN H, GE X, ZHOU X, LI Y, WANG A, HU Y. [Research Progress of Lung Cancer Vaccines]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2023; 26:692-700. [PMID: 37985155 PMCID: PMC10600751 DOI: 10.3779/j.issn.1009-3419.2023.106.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Indexed: 11/22/2023]
Abstract
With the development of medical technology, tumor vaccines as a novel precise immunotherapy approach have gradually received attention in clinical applications. Against the backdrop of the global corona virus disease 2019 (COVID-19) outbreak, vaccine technology has further advanced. Depending on the types of antigens, tumor vaccines can be divided into whole-cell vaccines, peptide vaccines, messenger ribonucleic acid (mRNA) vaccines, recombinant virus vaccines, etc. Although some tumor vaccines have been marketed and achieved certain therapeutic effects, the results of tumor vaccines in clinical trials have been unsatisfactory in the past period. With the maturation of next-generation sequencing (NGS) technology and the continuous development of bioinformatics, dynamic monitoring of the entire process of tumor subpopulation development has become a reality, which has laid a solid foundation for personalized, neoantigen-centered therapeutic tumor vaccines. This article reviews the recent developments of tumor vaccines of different types, starts with lung cancer and summarizes the achievements of tumor vaccines in clinical applications, and provides an outlook for the future development of antigen-centered tumor vaccines.
.
Collapse
|
4
|
Haraguchi M, Kiyotani K, Tate T, Sakata S, Sagawa R, Takagi S, Nagayama S, Takeuchi K, Takahashi K, Katayama R. Spatiotemporal commonality of the TCR repertoire in a T-cell memory murine model and in metastatic human colorectal cancer. Cancer Immunol Immunother 2023; 72:2971-2989. [PMID: 37270735 PMCID: PMC10992958 DOI: 10.1007/s00262-023-03473-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/22/2023] [Indexed: 06/05/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have shown superior clinical responses and significantly prolong overall survival (OS) for many types of cancer. However, some patients exhibit long-term OS, whereas others do not respond to ICI therapy at all. To develop more effective and long-lasting ICI therapy, understanding the host immune response to tumors and the development of biomarkers are imperative. In this study, we established an MC38 immunological memory mouse model by administering an anti-PD-L1 antibody and evaluating the detailed characteristics of the immune microenvironment including the T cell receptor (TCR) repertoire. In addition, we found that the memory mouse can be established by surgical resection of residual tumor following anti-PD-L1 antibody treatment with a success rate of > 40%. In this model, specific depletion of CD8 T cells revealed that they were responsible for the rejection of reinoculated MC38 cells. Analysis of the tumor microenvironment (TME) of memory mice using RNA-seq and flow cytometry revealed that memory mice had a quick and robust immune response to MC38 cells compared with naïve mice. A TCR repertoire analysis indicated that T cells with a specific TCR repertoire were expanded in the TME, systemically distributed, and preserved in the host for a long time period. We also identified shared TCR clonotypes between serially resected tumors in patients with colorectal cancer (CRC). Our results suggest that memory T cells are widely preserved in patients with CRC, and the MC38 memory model is potentially useful for the analysis of systemic memory T-cell behavior.
Collapse
Affiliation(s)
- Mizuki Haraguchi
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-Ku, Tokyo, 135-8550, Japan
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Kazuma Kiyotani
- Immunopharmacogenomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomohiro Tate
- Immunopharmacogenomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Seiji Sakata
- Pathology Project for Molecular Targets, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ray Sagawa
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Satoshi Takagi
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Satoshi Nagayama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Surgery, Uji-Tokushukai Medical Center, Kyoto, Japan
| | - Kengo Takeuchi
- Pathology Project for Molecular Targets, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-Ku, Tokyo, 135-8550, Japan.
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
5
|
Identification of T Cell Receptors Targeting a Neoantigen Derived from Recurrently Mutated FGFR3. Cancers (Basel) 2023; 15:cancers15041031. [PMID: 36831375 PMCID: PMC9953830 DOI: 10.3390/cancers15041031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Immunotherapies, including immune checkpoint blockades, play a critically important role in cancer treatments. For immunotherapies, neoantigens, which are generated by somatic mutations in cancer cells, are thought to be good targets due to their tumor specificity. Because neoantigens are unique in individual cancers, it is challenging to develop personalized immunotherapy targeting neoantigens. In this study, we screened "shared neoantigens", which are specific types of neoantigens derived from mutations observed commonly in a subset of cancer patients. Using exome sequencing data in the Cancer Genome Atlas (TCGA), we predicted shared neoantigen peptides and performed in vitro screening of shared neoantigen-reactive CD8+ T cells using peripheral blood from healthy donors. We examined the functional activity of neoantigen-specific T cell receptors (TCRs) by generating TCR-engineered T cells. Among the predicted shared neoantigens from TCGA data, we found that the mutated FGFR3Y373C peptide induced antigen-specific CD8+ T cells from the donor with HLA-A*02:06 via an ELISPOT assay. Subsequently, we obtained FGFR3Y373C-specific CD8+ T cell clones and identified two different sets of TCRs specifically reactive to FGFR3Y373C. We found that the TCR-engineered T cells expressing FGFR3Y373C-specific TCRs recognized the mutated FGFR3Y373C peptide but not the corresponding wild-type peptide. These two FGFR3Y373C-specific TCR-engineered T cells showed cytotoxic activity against mutated FGFR3Y373C-loaded cells. These results imply the possibility of strategies of immunotherapies targeting shared neoantigens, including cancer vaccines and TCR-engineered T cell therapies.
Collapse
|
6
|
Porciello N, Franzese O, D’Ambrosio L, Palermo B, Nisticò P. T-cell repertoire diversity: friend or foe for protective antitumor response? JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:356. [PMID: 36550555 PMCID: PMC9773533 DOI: 10.1186/s13046-022-02566-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Profiling the T-Cell Receptor (TCR) repertoire is establishing as a potent approach to investigate autologous and treatment-induced antitumor immune response. Technical and computational breakthroughs, including high throughput next-generation sequencing (NGS) approaches and spatial transcriptomics, are providing unprecedented insight into the mechanisms underlying antitumor immunity. A precise spatiotemporal variation of T-cell repertoire, which dynamically mirrors the functional state of the evolving host-cancer interaction, allows the tracking of the T-cell populations at play, and may identify the key cells responsible for tumor eradication, the evaluation of minimal residual disease and the identification of biomarkers of response to immunotherapy. In this review we will discuss the relationship between global metrics characterizing the TCR repertoire such as T-cell clonality and diversity and the resultant functional responses. In particular, we will explore how specific TCR repertoires in cancer patients can be predictive of prognosis or response to therapy and in particular how a given TCR re-arrangement, following immunotherapy, can predict a specific clinical outcome. Finally, we will examine current improvements in terms of T-cell sequencing, discussing advantages and challenges of current methodologies.
Collapse
Affiliation(s)
- Nicla Porciello
- grid.417520.50000 0004 1760 5276Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Ornella Franzese
- grid.6530.00000 0001 2300 0941Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Lorenzo D’Ambrosio
- grid.417520.50000 0004 1760 5276Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Belinda Palermo
- grid.417520.50000 0004 1760 5276Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Paola Nisticò
- grid.417520.50000 0004 1760 5276Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
7
|
Chen YL, Ho CL, Hung CY, Chen WL, Chang C, Hou YH, Chen JR, Chen PJ, Chow NH, Huang W, Hsu YT, Chen TY, Liu T. Enhancing diagnosis of T-cell lymphoma using non-recombined T-cell receptor sequences. Front Oncol 2022; 12:1014132. [PMID: 36568146 PMCID: PMC9772823 DOI: 10.3389/fonc.2022.1014132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
Clonality assessment, which can detect neoplastic T cells by identifying the uniquely recombined T-cell receptor (TCR) genes, provides important support in the diagnosis of T-cell lymphoma (TCL). BIOMED-2 is the gold standard clonality assay and has proven to be effective in European TCL patients. However, we failed to prove its sensitivity in Taiwanese TCL patients, especially based on the TCRβ gene. To explore potential impact of genetic background in the BIOMED-2 test, we analyzed TCRβ sequences of 21 healthy individuals and two TCL patients. This analysis suggests that genetic variations in the BIOMED-2 primer sites could not explain the difference in sensitivity. The BIOMED-2 test results of the two TCL patients were positive and negative, respectively. Interestingly, a higher percentage (>81%) of non-recombined TCRβ sequences was observed in the test-negative patient than those of the test-positive patient and all healthy individuals (13~66%). The result suggests a new TCR target for enhancing TCL diagnosis. To further explore the hypothesis, we proposed a cost-effective digital PCR assay that quantifies the relative abundance of non-recombined TCRβ sequences containing a J2-2P~J2-3 segment. With the digital PCR assay, bone marrow specimens from TCL patients (n=9) showed a positive outcome (i.e., the relative abundance of the J2-2P~J2-3 sequences ≧5%), whereas non-TCL patients (n=6) gave a negative result. As five of nine TCL patients had a negative BIOMED-2 test result, the J2-2P~J2-3 sequences may improve TCL detection. This is the first report showing the capability of characterizing non-recombined TCR sequences as a supplementary strategy for the BIOMED-2 clonality test.
Collapse
Affiliation(s)
- Yi-Lin Chen
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan,Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan,Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chung-Liang Ho
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan,Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan,Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chen-Yan Hung
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Wan-Li Chen
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan,Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chen Chang
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Yi-Hsin Hou
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan,Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Jian-Rong Chen
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan,Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Pin-Jun Chen
- Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Nan-Haw Chow
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan,Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan,Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wenya Huang
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan,Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Ting Hsu
- Section of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Tsai-Yun Chen
- Section of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Tsunglin Liu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan,*Correspondence: Tsunglin Liu,
| |
Collapse
|
8
|
Jiang J, Xia M, Zhang L, Chen X, Zhao Y, Zeng C, Yang H, Liang P, Li G, Li N, Qi H, Wei T, Ren L. Rapid generation of genetically engineered T cells for the treatment of virus-related cancers. Cancer Sci 2022; 113:3686-3697. [PMID: 35950597 DOI: 10.1111/cas.15528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/31/2022] [Accepted: 08/04/2022] [Indexed: 11/28/2022] Open
Abstract
Adoptive transfer of T cell receptor (TCR) engineered T cells targeting viral epitopes represents a promising approach for treating virus-related cancers. However, efficient identification of epitopes for T cells and corresponding TCR remains challenging. Here, we report a workflow permitting rapid generation of human papillomavirus (HPV)-specific TCR-T cells. Six epitopes of viral proteins belonged to HPV16 or HPV18 were predicted of high affinity to A11:01 according to bioinformatic analysis. Subsequently, cytotoxic T cells (CTLs) induction were performed with these six antigen peptides separately, and antigen-specific T cells were sorted by FACS. TCR clonotypes of these virus-specific T cells were determined by next-generation sequencing. To improve the efficiency of TCRαβ pairs validation, a lentiviral vector library containing 116 TCR constructs was generated, which was consisted of predominant TCRs according to TCR repoertire analysis. Later, TCR library transduced T cells were simulated with peptide pool-pulsed antigen presenting cells, then CD137-positive cells were sorted and subjected to TCR repoertire analysis. The top-hit TCRs and corresponding antigen peptides were deduced and validated. Through this workflow, a TCR targeting the E692-101 of HPV16 was identified. This HPV16-specific TCR-T cells showed high activities to HPV16-positive human cervical cancer cells in vitro and efficiently repressed tumor growth in murine model. This study provides a HPV16-specific TCR fitted to HLA-A11:01 population, and exemplifies an efficient approach which can be applied in large-scale screen of virus-specific TCRs, further encouraging researchers to exploit the therapeutic potential of TCR-T cell technique in treating virus-related cancers.
Collapse
Affiliation(s)
- Jinxing Jiang
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Ming Xia
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Lijie Zhang
- Department of Gynecology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Xi Chen
- RootPath, Inc. 65 Grove Street, Suite 203, 02472, Watertown, MA, USA
| | - Yue Zhao
- RootPath, Inc. 65 Grove Street, Suite 203, 02472, Watertown, MA, USA
| | - Chenquan Zeng
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Haiyan Yang
- RootPath, Inc. 65 Grove Street, Suite 203, 02472, Watertown, MA, USA
| | - Peng Liang
- RootPath, Inc. 65 Grove Street, Suite 203, 02472, Watertown, MA, USA
| | - Guanghe Li
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Ning Li
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Hui Qi
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Teng Wei
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Lili Ren
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| |
Collapse
|
9
|
Okamura K, Nagayama S, Tate T, Chan HT, Kiyotani K, Nakamura Y. Lymphocytes in tumor-draining lymph nodes co-cultured with autologous tumor cells for adoptive cell therapy. J Transl Med 2022; 20:241. [PMID: 35606862 PMCID: PMC9125345 DOI: 10.1186/s12967-022-03444-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/16/2022] [Indexed: 11/28/2022] Open
Abstract
Background Tumor-draining lymph nodes (TDLNs) are primary sites, where anti-tumor lymphocytes are primed to tumor-specific antigens and play pivotal roles in immune responses against tumors. Although adoptive cell therapy (ACT) using lymphocytes isolated from TDLNs were reported, characterization of immune activity of lymphocytes in TDLNs to tumor cells was not comprehensively performed. Here, we demonstrate TDLNs to have very high potential as cell sources for immunotherapy. Methods Lymphocytes from TDLNs resected during surgical operation were cultured with autologous-tumor cells for 2 weeks and evaluated tumor-reactivity by IFNγ ELISPOT assay. We investigated the commonality of T cell receptor (TCR) clonotypes expanded by the co-culture with tumor cells with those of tumor infiltrating lymphocytes (TILs). Results We found that that TCR clonotypes of PD-1-expressing CD8+ T cells in lymph nodes commonly shared with those of TILs in primary tumors and lymphocytes having tumor-reactivity and TCR clonotypes shared with TILs could be induced from non-metastatic lymph nodes when they were co-cultured with autologous tumor cells. Conclusion Our results imply that tumor-reactive effector T cells were present even in pathologically non-metastatic lymph nodes and could be expanded in vitro in the presence of autologous tumor cells and possibly be applied for ACT. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03444-1.
Collapse
|
10
|
Kockelbergh H, Evans S, Deng T, Clyne E, Kyriakidou A, Economou A, Luu Hoang KN, Woodmansey S, Foers A, Fowler A, Soilleux EJ. Utility of Bulk T-Cell Receptor Repertoire Sequencing Analysis in Understanding Immune Responses to COVID-19. Diagnostics (Basel) 2022; 12:1222. [PMID: 35626377 PMCID: PMC9140453 DOI: 10.3390/diagnostics12051222] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 01/27/2023] Open
Abstract
Measuring immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 19 (COVID-19), can rely on antibodies, reactive T cells and other factors, with T-cell-mediated responses appearing to have greater sensitivity and longevity. Because each T cell carries an essentially unique nucleic acid sequence for its T-cell receptor (TCR), we can interrogate sequence data derived from DNA or RNA to assess aspects of the immune response. This review deals with the utility of bulk, rather than single-cell, sequencing of TCR repertoires, considering the importance of study design, in terms of cohort selection, laboratory methods and analysis. The advances in understanding SARS-CoV-2 immunity that have resulted from bulk TCR repertoire sequencing are also be discussed. The complexity of sequencing data obtained by bulk repertoire sequencing makes analysis challenging, but simple descriptive analyses, clonal analysis, searches for specific sequences associated with immune responses to SARS-CoV-2, motif-based analyses, and machine learning approaches have all been applied. TCR repertoire sequencing has demonstrated early expansion followed by contraction of SARS-CoV-2-specific clonotypes, during active infection. Maintenance of TCR repertoire diversity, including the maintenance of diversity of anti-SARS-CoV-2 response, predicts a favourable outcome. TCR repertoire narrowing in severe COVID-19 is most likely a consequence of COVID-19-associated lymphopenia. It has been possible to follow clonotypic sequences longitudinally, which has been particularly valuable for clonotypes known to be associated with SARS-CoV-2 peptide/MHC tetramer binding or with SARS-CoV-2 peptide-induced cytokine responses. Closely related clonotypes to these previously identified sequences have been shown to respond with similar kinetics during infection. A possible superantigen-like effect of the SARS-CoV-2 spike protein has been identified, by means of observing V-segment skewing in patients with severe COVID-19, together with structural modelling. Such a superantigen-like activity, which is apparently absent from other coronaviruses, may be the basis of multisystem inflammatory syndrome and cytokine storms in COVID-19. Bulk TCR repertoire sequencing has proven to be a useful and cost-effective approach to understanding interactions between SARS-CoV-2 and the human host, with the potential to inform the design of therapeutics and vaccines, as well as to provide invaluable pathogenetic and epidemiological insights.
Collapse
Affiliation(s)
- Hannah Kockelbergh
- Department of Health Data Science, Institute of Population Health, University of Liverpool, Liverpool L69 3GF, UK;
| | - Shelley Evans
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (S.E.); (T.D.); (E.C.); (K.N.L.H.); (S.W.)
| | - Tong Deng
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (S.E.); (T.D.); (E.C.); (K.N.L.H.); (S.W.)
| | - Ella Clyne
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (S.E.); (T.D.); (E.C.); (K.N.L.H.); (S.W.)
| | - Anna Kyriakidou
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 1QP, UK; (A.K.); (A.E.)
| | - Andreas Economou
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 1QP, UK; (A.K.); (A.E.)
| | - Kim Ngan Luu Hoang
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (S.E.); (T.D.); (E.C.); (K.N.L.H.); (S.W.)
| | - Stephen Woodmansey
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (S.E.); (T.D.); (E.C.); (K.N.L.H.); (S.W.)
- Department of Respiratory Medicine, University Hospitals of Morecambe Bay, Kendal LA9 7RG, UK
| | - Andrew Foers
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7YF, UK;
| | - Anna Fowler
- Department of Health Data Science, Institute of Population Health, University of Liverpool, Liverpool L69 3GF, UK;
| | - Elizabeth J. Soilleux
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (S.E.); (T.D.); (E.C.); (K.N.L.H.); (S.W.)
| |
Collapse
|
11
|
Wei T, Leisegang M, Xia M, Kiyotani K, Li N, Zeng C, Deng C, Jiang J, Harada M, Agrawal N, Li L, Qi H, Nakamura Y, Ren L. Generation of neoantigen-specific T cells for adoptive cell transfer for treating head and neck squamous cell carcinoma. Oncoimmunology 2021; 10:1929726. [PMID: 34104546 PMCID: PMC8158031 DOI: 10.1080/2162402x.2021.1929726] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Adoptive cell therapy using TCR-engineered T cells (TCR-T cells) represents a promising strategy for treating relapsed and metastatic cancers. We previously established methods to identify neoantigen-specific TCRs based on patients’ PBMCs. However, in clinical practice isolation of PBMCs from advanced-stage cancer patients proves to be difficult. In this study, we substituted blood-derived T cells for tumor-infiltrating lymphocytes (TILs) and used an HLA-matched cell line of antigen-presenting cells (APCs) to replace autologous dendritic cells. Somatic mutations were determined in head and neck squamous cell carcinoma resected from two patients. HLA-A*02:01-restricted neoantigen libraries were constructed and transferred into HLA-matched APCs for stimulation of patient TILs. TCRs were isolated from reactive TIL cultures and functionality was tested using TCR- T cells in vitro and in vivo. To exemplify the screening approach, we identified the targeted neoantigen leading to recognition of the minigene construct that stimulated the strongest TIL response. Neoantigen peptides were used to load MHC-tetramers for T cell isolation and a TCR was identified targeting the KIAA1429D1358E mutation. TCR-T cells were activated, exhibited cytotoxicity, and secreted cytokines in a dose-dependent manner, and only when stimulated with the mutant peptide. Furthermore, comparable to a neoantigen-specific TCR that was isolated from the patient’s PBMCs, KIAA1429D1358E-specific TCR T cells destroyed human tumors in mice. The established protocol provides the required flexibility to methods striving to identify neoantigen-specific TCRs. By using an MHC-matched APC cell line and neoantigen-encoding minigene libraries, autologous TILs can be stimulated and screened when patient PBMCs and/or tumor material are not available anymore. Abbreviations: Head and neck squamous cell carcinoma (HNSCC); adoptive T cell therapy (ACT); T cell receptor (TCR); tumor-infiltrating lymphocytes (TIL); cytotoxic T lymphocyte (CTL); peripheral blood mononuclear cell (PBMC); dendritic cell (DC); antigen-presenting cells (APC)
Collapse
Affiliation(s)
- Teng Wei
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China.,Institute of Clinical Oncology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Matthias Leisegang
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,David and Etta Jonas Center for Cellular Therapy, the University of Chicago, Chicago, IL, USA.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ming Xia
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Kazuma Kiyotani
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ning Li
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Chenquan Zeng
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Chunyan Deng
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Jinxing Jiang
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Makiko Harada
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Nishant Agrawal
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Liangping Li
- Institute of Clinical Oncology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Hui Qi
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| | - Yusuke Nakamura
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Lili Ren
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Guangdong, China
| |
Collapse
|
12
|
Qian L, Zhaohui Z, Yaping X, Zhentian L, Zhentao L, Qiqi W, Yangchun G, Yan'e L, Wencheng Y, Fumei Y, Yanhong Y, Baoshan C, Li L. Blood T cell diversity associated with the prognosis of advanced non-small cell lung carcinoma treated with first-line pemetrexed based chemotherapy. Thorac Cancer 2021; 12:997-1005. [PMID: 33626215 PMCID: PMC8017263 DOI: 10.1111/1759-7714.13771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The tumor microenvironment is associated with prognosis in advanced non-small cell lung carcinoma (NSCLC). The aim of this study was to explore the relationship between blood T cell diversity and survival of patients treated with pemetrexed-based chemotherapy for nonsquamous NSCLC. METHODS This prospective clinical study enrolled 26 patients with advanced NSCLC treated with 4-6 cycles of first-line pemetrexed combined with platinum-based therapy. The complementarity-determining region 3 (CDR3) located in the T cell receptor beta chain (TCR β chain) was captured and deeply sequenced using next-generation sequencing (NGS) technology, and the correlation between TCR changes and efficacy after chemotherapy was analyzed. RESULTS Patients with an inferior quarter diversity index showed a significantly shorter progression-free survival (PFS) than the others (median, 5.0 months vs. 8.1 months, P = 0.014). After two cycles of chemotherapy, the TCR diversity was significantly higher than the baseline (P = 0.034). Just as with the baseline, patients with an inferior quarter diversity index at the endpoint of cycle 2 showed a shorter progression-free survival (PFS) than the others (median, 5.0 months vs. 8.4 months, P = 0.024). CONCLUSIONS In advanced NSCLC patients treated with first-line pemetrexed combined with platinum, the low level of blood TCR diversity at baseline with an endpoint of two cycles of chemotherapy was correlated with a poor prognosis.
Collapse
Affiliation(s)
- Li Qian
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Zhang Zhaohui
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Xu Yaping
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Liu Zhentian
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Liu Zhentao
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Wang Qiqi
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Gu Yangchun
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Liu Yan'e
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Yin Wencheng
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Yi Fumei
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Yao Yanhong
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Cao Baoshan
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Liang Li
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| |
Collapse
|
13
|
Impaired tumor immune response in metastatic tumors is a selective pressure for neutral evolution in CRC cases. PLoS Genet 2021; 17:e1009113. [PMID: 33476333 PMCID: PMC7864431 DOI: 10.1371/journal.pgen.1009113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/05/2021] [Accepted: 09/11/2020] [Indexed: 01/03/2023] Open
Abstract
A Darwinian evolutionary shift occurs early in the neutral evolution of advanced colorectal carcinoma (CRC), and copy number aberrations (CNA) are essential in the transition from adenoma to carcinoma. In light of this primary evolution, we investigated the evolutionary principles of the genome that foster postoperative recurrence of CRC. CNA and neoantigens (NAG) were compared between early primary tumors with recurrence (CRCR) and early primary tumors without recurrence (precancerous and early; PCRC). We compared CNA, single nucleotide variance (SNV), RNA sequences, and T-cell receptor (TCR) repertoire between 9 primary and 10 metastatic sites from 10 CRCR cases. We found that NAG in primary sites were fewer in CRCR than in PCRC, while the arm level CNA were significantly higher in primary sites in CRCR than in PCRC. Further, a comparison of genomic aberrations of primary and metastatic conditions revealed no significant differences in CNA. The driver mutations in recurrence were the trunk of the evolutionary phylogenic tree from primary sites to recurrence sites. Notably, PD-1 and TIM3, T cell exhaustion-related molecules of the tumor immune response, were abundantly expressed in metastatic sites compared to primary sites along with the increased number of CD8 expressing cells. The postoperative recurrence-free survival period was only significantly associated with the NAG levels and TCR repertoire diversity in metastatic sites. Therefore, CNA with diminished NAG and diverse TCR repertoire in pre-metastatic sites may determine postoperative recurrence of CRC. We found that copy number aberrations (CNAs) may be the most important selective pressure promoting cancer evolution from early-to-advanced tumors in primary sites. The diminished neoantigens (NAG) in cancer cells and the diverse TCR repertoire in cytotoxic T cells were crucial for the onset of postoperative recurrence during genomic neutral evolution, along with clonal CNA and several driver SNVs from primary to recurrent sites. Therefore, cancer metastasis could be prevented by activating CTL at the premetastatic sites before priming of the metastatic process.
Collapse
|
14
|
Kato T, Kiyotani K, Tomiyama E, Koh Y, Matsushita M, Hayashi Y, Nakano K, Ishizuya Y, Wang C, Hatano K, Kawashima A, Ujike T, Fujita K, Nonomura N, Uemura M. Peripheral T cell receptor repertoire features predict durable responses to anti-PD-1 inhibitor monotherapy in advanced renal cell carcinoma. Oncoimmunology 2021; 10:1862948. [PMID: 33537170 PMCID: PMC7833759 DOI: 10.1080/2162402x.2020.1862948] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) offer significant clinical benefits to a subset of cancer patients via the induction of a systemic T cell-mediated anti-cancer immune response. Thus, the dynamic characterization of T cell repertoires in the peripheral blood has the potential to demonstrate noninvasive predictive biomarkers for the clinical efficacy of ICIs. In this study, we collected tumor tissues and peripheral blood samples from 25 patients with advanced kidney cancer before anti-programmed cell death protein 1 (PD-1) treatment and 1, 3, and 6 months after treatment initiation. Furthermore, we applied a next-generation sequencing approach to characterize T cell receptor (TCR) alpha and beta repertoires. TCR repertoire analysis revealed that the responders to anti-PD-1 showed an expansion of certain T cell clones even in the blood, as evidenced by the significant decrease in the TCR diversity index and increase in the number of expanded TCR clonotypes 1 month after treatment. Interestingly, these expanded TCR clonotypes in the peripheral blood were significantly shared with tumor-infiltrating T cells in responders, indicating that they have many circulating T cells that may recognize cancer antigens. Expression analysis also revealed that 1 month after treatment, T cells from the peripheral blood of responders showed significantly elevated transcriptional levels of Granzyme B, Perforin, CD39, and PD-1, markers of cancer-associated antigen-specific T cells. Altogether, we propose that global TCR repertoire analysis may allow identifying early surrogate biomarkers in the peripheral blood for predicting clinical responses to anti-PD-1 monotherapy.
Collapse
Affiliation(s)
- Taigo Kato
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Urological Immuno-oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuma Kiyotani
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Eisuke Tomiyama
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoko Koh
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Makoto Matsushita
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yujiro Hayashi
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kosuke Nakano
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yu Ishizuya
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Cong Wang
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koji Hatano
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsunari Kawashima
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takeshi Ujike
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazutoshi Fujita
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Motohide Uemura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Urological Immuno-oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
15
|
Gotoh O, Kiyotani K, Chiba T, Sugiyama Y, Takazawa Y, Nemoto K, Kato K, Tanaka N, Nomura H, Hasegawa K, Fujiwara K, Takamatsu S, Matsumura N, Noda T, Mori S. Immunogenomic landscape of gynecologic carcinosarcoma. Gynecol Oncol 2020; 160:547-556. [PMID: 33298310 DOI: 10.1016/j.ygyno.2020.11.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Carcinosarcoma (CS) of the uterus or ovary is a rare, biphasic tumor comprising epithelial and mesenchymal elements, and exhibits more aggressive clinical features than its carcinoma counterpart. Four molecular subtypes of CS were recently established based on genomic aberration profiles (POLE, MSI, CNH, and CNL) and shown to be associated with multiple clinicopathological parameters, including patient outcomes. However, the role of the immune microenvironment in CS remains unclear. Here, we investigated the influence of the immune cells that infiltrate CS to better understand the immunological status of gynecological CS. METHODS Tumor immune microenvironmental analyses on CS samples were performed using immune cell profiling with RNA-seq, transcriptomic subtyping with microenvironmental genes, and T-cell receptor repertoire assay. Carcinoma and sarcoma elements from CS samples were also assessed separately. RESULTS Relying on estimations of tumor-infiltrating cell types from RNA-seq data, POLE and MSI (hypermutator) tumors showed an enrichment of M1 macrophages, plasma cells and CD8+ T cells, whereas CNH and CNL (non-hypermutator) tumors had high levels of M2 macrophages. Further subclassification by immune-related, non-cancer genes identified a fraction of tumors with distinct patient outcomes, particularly those with the CNH genomic aberration subtype. T-cell heterogeneity was independently correlated with prolonged progression-free survival. Differential analysis of carcinoma and sarcoma elements identified many shared mutations but there was little overlap in the T-cell receptor repertoire between the two elements. CONCLUSIONS Tumor immune microenvironmental analyses could offer potential clinical utility in the stratification of gynecological CS above classification by genomic aberration subtype alone.
Collapse
Affiliation(s)
- Osamu Gotoh
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, Japan
| | - Kazuma Kiyotani
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, Japan
| | - Tomohiro Chiba
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, Japan
| | - Yuko Sugiyama
- Department of Gynecology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, Japan
| | - Yutaka Takazawa
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, Japan
| | - Kensaku Nemoto
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, Japan
| | - Kazuyoshi Kato
- Department of Gynecology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, Japan
| | - Norio Tanaka
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, Japan
| | - Hidetaka Nomura
- Department of Gynecology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka-shi, Saitama, Japan
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka-shi, Saitama, Japan
| | - Shiro Takamatsu
- Department of Gynecologic Oncology, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osaka-Sayama, Osaka, Japan
| | - Tetsuo Noda
- Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, Japan
| | - Seiichi Mori
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, Japan.
| |
Collapse
|
16
|
Chang CM, Liao YM, Lan GY, Chang WC, Yen Y. Using T-cell repertoire profiles as predictor in a primary mucosal melanoma. Clin Transl Med 2020; 10:e136. [PMID: 32779855 PMCID: PMC7438817 DOI: 10.1002/ctm2.136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/11/2020] [Accepted: 07/11/2020] [Indexed: 02/03/2023] Open
Affiliation(s)
- Che-Mai Chang
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ming Liao
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - Gong-Yau Lan
- Section of Diagnostic Radiology, Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Integrative Therapy Center for Gastroenterologic Cancers, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yun Yen
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California
| |
Collapse
|
17
|
Dissecting efficiency of a 5' rapid amplification of cDNA ends (5'-RACE) approach for profiling T-cell receptor beta repertoire. PLoS One 2020; 15:e0236366. [PMID: 32702062 PMCID: PMC7377388 DOI: 10.1371/journal.pone.0236366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/02/2020] [Indexed: 01/21/2023] Open
Abstract
Deep sequencing of T-cell receptor (TCR) genes is powerful at profiling immune repertoire. To prepare a TCR sequencing library, multiplex polymerase chain reaction (mPCR) is widely applied and is highly efficient. That is, most mPCR products contain the region critical for antigen recognition, which also indicates regular V(D)J recombination. Multiplex PCR, however, may suffer from primer bias. A promising alternative is 5'-RACE, which avoids primer bias by applying only one primer pair. In 5'-RACE data, however, non-regular V(D)J recombination (e.g., TCR sequences without a V gene segment) has been observed and the frequency varies (30-80%) between studies. This suggests that the cause of or how to reduce non-regular TCR sequences is not yet well known by the science community. Although it is possible to speculate the cause by comparing the 5'-RACE protocols, careful experimental confirmation is needed and such a systematic study is still not available. Here, we examined the 5'-RACE protocol of a commercial kit and demonstrated how a modification increased the fraction of regular TCR-β sequences to >85%. We also found a strong linear correlation between the fraction of short DNA fragments and the percentage of non-regular TCR-β sequences, indicating that the presence of short DNA fragments in the library was the main cause of non-regular TCR-β sequences. Therefore, thorough removal of short DNA fragments from a 5'-RACE library is the key to high data efficiency. We highly recommend conducting a fragment length analysis before sequencing, and the fraction of short DNA fragments can be used to estimate the percentage of non-regular TCR sequences. As deep sequencing of TCR genes is still relatively expensive, good quality control should be valuable.
Collapse
|
18
|
Impact of T-cell receptor and B-cell receptor repertoire on the recurrence of early stage lung adenocarcinoma. Exp Cell Res 2020; 394:112134. [PMID: 32540399 DOI: 10.1016/j.yexcr.2020.112134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 12/25/2022]
Abstract
Surgical resection is the only curative treatment for patients with early stage non-small cell lung cancer. However, approximately 33% of non-small cell lung cancer patients recur with the stage I disease, which may be attributed to a deficiency in antitumor immunity. In the present study, for early stage lung adenocarcinoma patients with early recurrence and early non-recurrence, we investigated the quantity of tumor-infiltrating T and B cells by immunohistochemistry, as well as the genes in the complementarity determining region 3 of the T-cell receptor β chain and the B-cell receptor immunoglobulin heavy chain. A decreased number of tumor-infiltrating lymphocytes cells (CD3+, CD4+, CD8+ and CD20+) was present in early recurrence patients. A significant increase in oligoclones and a reduction in T-cell receptor diversity were observed in the early recurrence group. Furthermore, there was a preference for V, J gene, and VJ gene combinations in patients with early recurrence versus non-recurrence, suggesting that this may be a new biomarker for the recurrence of early stage lung adenocarcinoma. These data indicate that T and B cell receptor repertoires influence the depth of human adaptive immune responses, and in addition to the quantity of tumor infiltrating T and B cells, may contribute to the prevention of early stage lung adenocarcinoma recurrence after surgical resection. Our study illustrates the potential value of the immune repertoire for predicting clinical efficacy and patient outcomes.
Collapse
|
19
|
Schreiber K, Karrison TG, Wolf SP, Kiyotani K, Steiner M, Littmann ER, Pamer EG, Kammertoens T, Schreiber H, Leisegang M. Impact of TCR Diversity on the Development of Transplanted or Chemically Induced Tumors. Cancer Immunol Res 2019; 8:192-202. [PMID: 31831634 DOI: 10.1158/2326-6066.cir-19-0567] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/24/2019] [Accepted: 12/06/2019] [Indexed: 12/21/2022]
Abstract
Burnet postulated that the diversity of T-cell receptors (TCR) allows T cells to protect against the development of cancers that display antigens with a similar, seemingly endless diversity. To test this hypothesis, we developed a strategy in which a single breeding pair of mice gives rise to four groups of sibling mice. Three of the four groups had a similar number of CD8+ T cells, but TCR diversity was either broad, significantly reduced, or absent when expressing only one type of TCR. The fourth group had no T cells. All mice shared the same housing, and, therefore, their microbial environment was similar. Only slight differences in the intestinal flora were observed under these conditions. An undisturbed broad TCR repertoire was required for the rejection of inoculated cancers displaying the natural antigenic heterogeneity of primary tumors, whereas even one type of TCR was sufficient to protect against artificial cancers stably expressing cognate antigens. The three groups of mice with limited or no TCR repertoire showed an increased risk of developing primary tumors after chemical induction. However, the risk of early death or morbidity in these cohorts of mice was significantly higher than in mice with a diverse TCR repertoire, and it remains unknown whether mice with reduced TCR diversity, who died early without cancer, would have developed tumors with higher, lower, or equal probability after induction. Together, TCR diversity seems crucial to overcome the natural genetic instability of cancers and their antigenic heterogeneity, which impacts the design of cellular therapies.
Collapse
Affiliation(s)
- Karin Schreiber
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Theodore G Karrison
- Department of Public Health Sciences, The University of Chicago, Chicago, Illinois
| | - Steven P Wolf
- Department of Pathology, The University of Chicago, Chicago, Illinois.,Institute of Immunology, Charité - Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Kazuma Kiyotani
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Madeline Steiner
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Eric R Littmann
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric G Pamer
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Thomas Kammertoens
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Hans Schreiber
- Department of Pathology, The University of Chicago, Chicago, Illinois.
| | - Matthias Leisegang
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| |
Collapse
|
20
|
Akazawa Y, Hosono A, Yoshikawa T, Kaneda H, Nitani C, Hara J, Kinoshita Y, Kohashi K, Manabe A, Fukutani M, Wakabayashi M, Sato A, Shoda K, Shimomura M, Mizuno S, Nakamoto Y, Nakatsura T. Efficacy of the NCCV Cocktail-1 vaccine for refractory pediatric solid tumors: A phase I clinical trial. Cancer Sci 2019; 110:3650-3662. [PMID: 31571332 PMCID: PMC6890444 DOI: 10.1111/cas.14206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/18/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022] Open
Abstract
Pediatric refractory solid tumors are aggressive malignant diseases, resulting in an extremely poor prognosis. KOC1, FOXM1, and KIF20A are cancer antigens that could be ideal targets for anticancer immunotherapy against pediatric refractory solid tumors with positive expression for these antigens. This nonrandomized, open‐label, phase I clinical trial evaluated the safety and efficacy of the NCCV Cocktail‐1 vaccine, which is a cocktail of cancer peptides derived from KOC1, FOXM1, and KIF20A, in patients with pediatric refractory solid tumors. Twelve patients with refractory pediatric solid tumors underwent NCCV Cocktail‐1 vaccination weekly by intradermal injections. The primary endpoint was the safety of the NCCV Cocktail‐1 vaccination, and the secondary endpoints were the immune response, as measured by interferon‐r enzyme‐linked immunospot assay, and the clinical outcomes including tumor response and progression‐free survival. The NCCV Cocktail‐1 vaccine was well tolerated. The clinical response of this trial showed that 4 patients had stable disease after 8 weeks and 2 patients maintained remission for >11 months. In 4, 8, and 5 patients, the NCCV Cocktail‐1 vaccine induced the sufficient number of peptide‐specific CTLs for KOC1, FOXM1, and KIF20A, respectively. Patients with high peptide‐specific CTL frequencies for KOC1, FOXM1, and KIF20A had better progression‐free survival than those with low frequencies. The findings of this clinical trial showed that the NCCV Cocktail‐1 vaccine could be a novel therapeutic strategy, with adequate effects against pediatric refractory solid tumors. Future large‐scale trials should evaluate the efficacy of the NCCV Cocktail‐1 vaccination.
Collapse
Affiliation(s)
- Yu Akazawa
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Eiheiji-cho, Japan
| | - Ako Hosono
- Division of Pediatric Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Toshiaki Yoshikawa
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Hide Kaneda
- Division of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Chika Nitani
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Junichi Hara
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Yoshiaki Kinoshita
- Department of Pediatric Surgery, Kyushu University Hospital, Fukuoka, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University, Sapporo, Japan
| | - Miki Fukutani
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masashi Wakabayashi
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Akihiro Sato
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kayoko Shoda
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Manami Shimomura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Shoichi Mizuno
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Yasunari Nakamoto
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Eiheiji-cho, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
21
|
Quantitative characterization of T-cell repertoire alteration in Chinese patients with B-cell acute lymphocyte leukemia after CAR-T therapy. Bone Marrow Transplant 2019; 54:2072-2080. [PMID: 31383996 DOI: 10.1038/s41409-019-0625-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/27/2019] [Accepted: 05/18/2019] [Indexed: 01/08/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has displayed potent anti-leukemia activity in acute lymphocytic leukemia (ALL), acting as a new ray of hope to refractory/relapsed patients. However, the influence of CAR-T therapy on host immune system has not been well elucidated. Thus, We applied high-throughput T cell receptor β chain sequencing to track the dynamic change of T-cell repertoire induced by CAR-T therapy in B-cell ALL patients. Six Chinese patients achieving complete remission were under observation, whose blood samples, bone marrow samples and infused CAR-T samples were collected at serial time points before and after CAR-T therapy. We observed decreased TCR diversity and increased clonality of T-cell repertoire in both peripheral blood and bone marrow after CAR-T administration. The persistent T cell clones in blood and bone marrow expanded following leukemic cell destruction and were barely detected in CAR T-cell pool. For the first time, our results demonstrated CAR-T therapy could stimulate the clonal proliferation of CAR-negative T cells in patients. Considering other groups' animal results indicating that CAR-T therapy could facilitate the proliferation of tumor antigen-specific T cells and that the emergence of these T cell clones followed the destruction of leukemic cells, they are most likely tumor antigen-specific.
Collapse
|
22
|
Hasan MS, Wu X, Zhang L. Uncovering missed indels by leveraging unmapped reads. Sci Rep 2019; 9:11093. [PMID: 31366961 PMCID: PMC6668410 DOI: 10.1038/s41598-019-47405-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/12/2019] [Indexed: 02/08/2023] Open
Abstract
In current practice, Next Generation Sequencing (NGS) applications start with mapping/aligning short reads to the reference genome, with the aim of identifying genetic variants. Although existing alignment tools have shown great accuracy in mapping short reads to the reference genome, a significant number of short reads still remain unmapped and are often excluded from downstream analyses thereby causing nonnegligible information loss in the subsequent variant calling procedure. This paper describes Genesis-indel, a computational pipeline that explores the unmapped reads to identify novel indels that are initially missed in the original procedure. Genesis-indel is applied to the unmapped reads of 30 breast cancer patients from TCGA. Results show that the unmapped reads are conserved between the two subtypes of breast cancer investigated in this study and might contribute to the divergence between the subtypes. Genesis-indel identifies 72,997 novel high-quality indels previously not found, among which 16,141 have not been annotated in the widely used mutation database. Statistical analysis of these indels shows significant enrichment of indels residing in oncogenes and tumour suppressor genes. Functional annotation further reveals that these indels are strongly correlated with pathways of cancer and can have high to moderate impact on protein functions. Additionally, some of the indels overlap with the genes that do not have any indel mutations called from the originally mapped reads but have been shown to contribute to the tumorigenesis in multiple carcinomas, further emphasizing the importance of rescuing indels hidden in the unmapped reads in cancer and disease studies.
Collapse
Affiliation(s)
| | - Xiaowei Wu
- Department of Statistics, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Liqing Zhang
- Department of Computer Science, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
23
|
Nakagawa R, Muroyama R, Saeki C, Oikawa T, Kaise Y, Koike K, Arai J, Nakano M, Matsubara Y, Takano K, Hirata Y, Saruta M, Zeniya M, Kato N. CD4 + T cells from patients with primary biliary cholangitis show T cell activation and differentially expressed T-cell receptor repertoires. Hepatol Res 2019; 49:653-662. [PMID: 30690835 DOI: 10.1111/hepr.13318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 01/08/2019] [Accepted: 01/22/2019] [Indexed: 02/08/2023]
Abstract
AIM Primary biliary cholangitis (PBC) is an autoimmune liver disease with unknown pathogenesis. In PBC, activation of T-cell receptor (TCR) signaling is associated with inflammatory cytokine production through N-Ras upregulation. Although the CD4+ T cell TCR repertoire could be associated with PBC pathogenesis, it has not been evaluated. Thus, we analyzed the PBC-CD4+ T cell TCR repertoire using next generation sequencing (NGS). METHODS Four PBC patients (one treatment-naïve and three receiving ursodeoxycholic acid) and three healthy individuals were enrolled. NRAS expression in CD4+ T cells was assessed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). N-Ras dynamics in CD4+ T cells were assessed by qRT-PCR and GTP-N-Ras activation assay. The TCR α- (TRA) and β-chain (TRB) repertoires on CD4+ T cells were analyzed by NGS and profiled using hierarchical analysis. Motif analysis was undertaken to elucidate the structure of PBC-specific TCRs. RESULTS NRAS was upregulated in PBC relative to control CD4+ T cells (P < 0.05), and N-Ras enhanced T cell activation in CD4+ T cells. Among 2668 TRAs and 841 TRBs, 20 and 11, respectively, were differentially expressed in PBC compared to that in controls (P < 0.05, fold-change >2). Among them, TRAV29/J22, TRBV6-5/J2-6, and TRBV10-1/J2-1 were expressed in PBC but the expression was negligible in the controls, with more mature and longer forms observed in PBC-CD4+ T cells. CONCLUSIONS N-Ras was upregulated in PBC-CD4+ T cells, and it enhanced TCR activation, indicating that PBC-CD4+ T cells were activated by N-Ras upregulation with differentially expressed TCR repertoires on their surfaces.
Collapse
Affiliation(s)
- Ryo Nakagawa
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan.,Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryosuke Muroyama
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Chisato Saeki
- Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan
| | - Tsunekazu Oikawa
- Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan
| | - Yoshimi Kaise
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan
| | - Jun Arai
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Masanori Nakano
- Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan
| | - Yasuo Matsubara
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Keiko Takano
- Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan
| | - Yoshihiro Hirata
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Masayuki Saruta
- Department of Gastroenterology and Hepatology, Jikei University School of Medicine, Tokyo, Japan
| | - Mikio Zeniya
- Sanno Medical Center, International University of Health and Welfare, Tokyo, Japan
| | - Naoya Kato
- Division of Advanced Genome Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
24
|
Human urothelial bladder cancer generates a clonal immune response: The results of T-cell receptor sequencing. Urol Oncol 2019; 37:810.e1-810.e5. [PMID: 31103336 DOI: 10.1016/j.urolonc.2019.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/22/2019] [Accepted: 04/12/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND High T-cell receptor (TCR) repertoire clonality is associated with clinical response to immune checkpoint blockade in bladder cancer. OBJECTIVE To determine if TCR repertoire is more clonal in tumors than in benign inflammation. METHODS We prospectively identified 12 patients with bladder lesions undergoing transurethral resection. Specimens were collected at time of transurethral resection and stored at -80C. DNA was extracted and high throughput DNA sequencing of the CDR3 region of the TCR beta chain using the immunoSEQ assay (Adaptive Biotechnologies) was performed. T-cell fraction, clonal dominance, and maximum frequency of TCR clone were assessed. RESULTS Of the 12 bladder lesions resected, 3 of 12 were cT0, 3 of 12 were cTa, 3 of 12 were cT1, and 3 of 12 were cT2 or greater. The median number of T cells in urothelial carcinoma specimens (UC+) and benign (UC-) specimens was 5,569 and 25,872, respectively. The number of unique TCRs sequenced in UC+ and UC- specimens was 3,069 and 9,680, respectively. The median tumor infiltrating lymphocyte percentage in UC+ and UC- specimens was 2% and 12%, respectively. The UC+ specimens demonstrated clonality as evidenced by identification of a specific T-cell clone being present in up to 17% of the total tumor infiltrating lymphocyte pool, in contrast to 2% among UC- specimens. CONCLUSIONS Primary urothelial tumors contain clonally expanded T-cell populations. These data support the hypothesis that bladder tumors induce an antigen-driven immunogenic host response, in contrast to the benign inflammatory response, which does not appear to demonstrate any T-cell clonal dominance.
Collapse
|
25
|
Leduc C, Quoix E. [Vaccines for the treatment of non-small cell lung cancer]. Rev Mal Respir 2019; 36:415-425. [PMID: 30902445 DOI: 10.1016/j.rmr.2018.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 05/31/2018] [Indexed: 11/26/2022]
Abstract
Antigen-specific immunotherapy also known as cancer vaccination offers a novel approach for the treatment of non-small cell lung cancer patients. It relies on specific priming of the immune system in order to provoke or increase adaptive antitumor immune response against the vaccine component. Several molecules have been developed in lung cancer, based on whole-tumor cells, dendritic cells, peptides, recombinant proteins, or viral vectors. The aim of this review is to describe the mechanism of action of these vaccines and the results of the main clinical studies.
Collapse
Affiliation(s)
- C Leduc
- Service de pneumologie, CHRU de Strasbourg, Strasbourg, France
| | - E Quoix
- Service de pneumologie, CHRU de Strasbourg, Strasbourg, France.
| |
Collapse
|
26
|
Matsuda T, Miyauchi E, Hsu YW, Nagayama S, Kiyotani K, Zewde M, Park JH, Kato T, Harada M, Matsui S, Ueno M, Fukuda K, Suzuki N, Hazama S, Nagano H, Takeuchi H, Vigneswaran WT, Kitagawa Y, Nakamura Y. TCR sequencing analysis of cancer tissues and tumor draining lymph nodes in colorectal cancer patients. Oncoimmunology 2019; 8:e1588085. [PMID: 31069156 DOI: 10.1080/2162402x.2019.1588085] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/21/2019] [Accepted: 02/16/2019] [Indexed: 12/15/2022] Open
Abstract
Tumor draining lymph nodes (TDLNs) are located in the routes of lymphatic drainage from a primary tumor and have the highest risk of metastasis in various types of solid tumors. TDLNs are also considered as a tissue to activate the antitumor immunity, where antigen-specific effector T cells are generated. However, T cell receptor (TCR) repertoires in TDLNs have not been well characterized. We collected 23 colorectal cancer tumors with 203 lymph nodes with/without metastatic cancer cells (67 were metastasis-positive and the remaining 136 were metastasis-negative) and performed TCR sequencing. Metastasis-positive TDLNs showed a significantly lower TCR diversity and shared TCR clonotypes more frequently with primary tumor tissues compared to metastasis-negative TDLNs. Principal component analysis indicated that TDLNs with metastasis showed similar TCR repertoires. These findings suggest that cancer-reactive T cell clones could be enriched in the metastasis-positive TDLNs.
Collapse
Affiliation(s)
- Tatsuo Matsuda
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Department of Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Eisaku Miyauchi
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Yu-Wen Hsu
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - Satoshi Nagayama
- Department of Gastroenterological Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kazuma Kiyotani
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makda Zewde
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jae-Hyun Park
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Taigo Kato
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Makiko Harada
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Shimpei Matsui
- Department of Gastroenterological Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masashi Ueno
- Department of Gastroenterological Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kazumasa Fukuda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Nobuaki Suzuki
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Shoichi Hazama
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hiroya Takeuchi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Wickii T Vigneswaran
- Department of Thoracic and Cardiovascular Surgery, Loyola University Medical Center, Maywood, IL, USA
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yusuke Nakamura
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
27
|
Miyauchi E, Matsuda T, Kiyotani K, Low S, Hsu Y, Tsukita Y, Ichinose M, Sakurada A, Okada Y, Saito R, Nakamura Y. Significant differences in T cell receptor repertoires in lung adenocarcinomas with and without epidermal growth factor receptor mutations. Cancer Sci 2019; 110:867-874. [PMID: 30582659 PMCID: PMC6398877 DOI: 10.1111/cas.13919] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent clinical trials of non-small cell lung cancer with immune checkpoint inhibitors revealed that patients with epidermal growth factor receptor (EGFR) mutations had more unfavorable outcomes compared with those with wild-type EGFR. However, the underlying mechanism for the link between EGFR mutations and immune resistance remains unclear. We performed T cell receptor (TCR) repertoire analysis of resected lung adenocarcinoma tissues with and without EGFR mutations to investigate the characteristics of TCR repertoires. We collected a total of 39 paired (normal and tumor) lung tissue samples (20 had EGFR mutations) and conducted TCR repertoire analysis as well as whole-exome sequencing (WES) and transcriptome analysis. The TCR diversity index in EGFR-mutant tumors was significantly higher than that in EGFR-wild-type tumors (median [range] 552 [162-1,135] vs 230 [30-764]; P < .01), suggesting higher T cell clonal expansion in EGFR-wild-type tumors than in EGFR-mutant tumors. In WES, EGFR-mutant tumors showed lower numbers of non-synonymous mutations and predicted neoantigens than EGFR-wild-type tumors (P < .01, P = .03, respectively). The number of non-synonymous mutations revealed a positive correlation with the sum of frequencies of the TCRβ clonotypes of 1% or higher in tumors (r = .52, P = .04). The present study demonstrates significant differences in TCR repertoires and the number of predicted neoantigens between EGFR-mutant and wild-type lung tumors. Our findings provide important information for understanding the molecular mechanism behind EGFR-mutant patients showing unfavorable responses to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Eisaku Miyauchi
- Department of MedicineThe University of ChicagoChicagoIllinois
- Department of Respiratory MedicineTohoku UniversitySendaiJapan
| | - Tatsuo Matsuda
- Department of MedicineThe University of ChicagoChicagoIllinois
| | - Kazuma Kiyotani
- Cancer Precision Medicine CenterJapanese Foundation for Cancer ResearchTokyoJapan
| | - Siew‐Kee Low
- Cancer Precision Medicine CenterJapanese Foundation for Cancer ResearchTokyoJapan
| | - Yu‐Wen Hsu
- Department of MedicineThe University of ChicagoChicagoIllinois
- The Ph.D. Program for Translational MedicineTaipei Medical University and Academia SinicaTaipeiTaiwan
| | - Yoko Tsukita
- Department of Respiratory MedicineTohoku UniversitySendaiJapan
| | | | - Akira Sakurada
- Department of Thoracic SurgeryTohoku UniversitySendaiJapan
| | | | - Ryoko Saito
- Department of PathologyTohoku University HospitalSendaiJapan
| | - Yusuke Nakamura
- Department of MedicineThe University of ChicagoChicagoIllinois
- Cancer Precision Medicine CenterJapanese Foundation for Cancer ResearchTokyoJapan
- Department of SurgeryThe University of ChicagoChicagoIllinois
| |
Collapse
|
28
|
Li Y, Ma L, Dong X, Pan Y, Shi B, He X, Zhang T, Sun S, Yao X. Preliminary analysis of spatial-temporal homogeneity and heterogeneity of TCR β chain CDR3 repertoires in BALB/c mice. INTERNATIONAL JOURNAL OF MOLECULAR EPIDEMIOLOGY AND GENETICS 2019; 10:10-28. [PMID: 30911358 PMCID: PMC6420717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 01/21/2019] [Indexed: 06/09/2023]
Abstract
The T-cell response and tolerance in non-lymph tissues differs from those in lymph tissues such as the spleen and thymus. The distribution and composition of the TCR repertoires in non-lymph tissues and how they differ and associate with their counterparts in lymph tissue remain unclear. Thus, we studied the thymus, spleen, blood, liver and small intestine of BALB/c mice at the ages of one, three and five months to carry out a preliminary analysis of the spatial-temporal homogeneity and heterogeneity of the total TCR β chain CDR3 repertoire using high-throughput sequencing technology and immune bioinformatics approaches. The data show that the diversity of the CDR3 repertoires was decreased as the mouse age increased, except in the small intestine. The number of low-expanded clones in the CDR3 repertoires was greatest in the thymus, followed by the spleen, blood, liver and small intestine, and highly expanded clones had an opposite trend in the different mice ages. The thymus and the spleen showed the greatest overlap of CDR3 sequences with the other tissues across the different mice ages. The distribution of the CDR3 repertoire length was normal, with a median of 14 aa in all the mouse tissues, except the small intestine of the one-month-old mice had a median of 12 aa. In summary, the composition and characteristics of the CDR3 repertoires in the thymus were similar to those in the spleen, and repertoires in the blood were similar to those in the liver; only the small intestine showed a unique composition. These results offer a novel method to explore the source, differentiation, proliferation and response of distinct T cells in different tissues at different mice ages.
Collapse
Affiliation(s)
- Yuehong Li
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| | - Long Ma
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| | - Xiaoheng Dong
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| | - Yurong Pan
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| | - Bin Shi
- Department of Laboratory Medicine, Zunyi Medical UniversityZunyi 563003, China
| | - Xiaoyan He
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| | - Teng Zhang
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| | - Suhong Sun
- Department of Breast Surgery, The First Affiliated Hospital of Zunyi Medical CollegeZunyi 563003, China
| | - Xinsheng Yao
- Department of Immunology, Research Center for Medicine and Biology, Innovation and Practice Base for Graduate Students Education, Zunyi Medical UniversityZunyi 563003, China
| |
Collapse
|
29
|
Ren L, Leisegang M, Deng B, Matsuda T, Kiyotani K, Kato T, Harada M, Park JH, Saloura V, Seiwert T, Vokes E, Agrawal N, Nakamura Y. Identification of neoantigen-specific T cells and their targets: implications for immunotherapy of head and neck squamous cell carcinoma. Oncoimmunology 2019; 8:e1568813. [PMID: 30906664 PMCID: PMC6422382 DOI: 10.1080/2162402x.2019.1568813] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/23/2018] [Accepted: 11/02/2018] [Indexed: 01/04/2023] Open
Abstract
To develop a practically applicable method for T-cell receptor (TCR)-engineered T cell immunotherapy targeting neoantigens, we have been attempting to identify neoantigen-specific T cell receptors (TCRs) and establish TCR-engineered T cells in a 3-4-month period. In this study, we report the characterization of T cell repertoires in tumor microenvironment (TME) and identification of neoantigen-specific TCRs after stimulation of patient-derived T cells. We screened 15 potential neoantigen peptides and successfully identified two CD8+HLA-dextramer+ T cells, which recognized MAGOHBG17A and ZCCHC14P368L. All three dominant TCR clonotypes from MAGOHBG17A-HLA dextramer-sorted CD8+ T cells were also found in T cells in TME, while none of dominant TCR clonotypes from ZCCHC14P368L-HLA dextramer-sorted CD8+ T cells was found in the corresponding TME. The most dominant TCRA/TCRB pairs for these two neoantigens were cloned into HLA-matched healthy donors' T lymphocytes to generate TCR-engineered T cells. The functional assay showed MAGOHBG17A TCR-engineered T cells could be significantly activated in a mutation-specific, HLA-restricted and peptide-dose-dependent manner while ZCCHC14P368L TCR-engineered T cells could not. Our data showed neoantigen-reactive T cell clonotypes that were identified in the patient's peripheral blood could be present in the corresponding TME and might be good TCRs targeting neoantigens.
Collapse
Affiliation(s)
- Lili Ren
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Cytotherapy Laboratory, Shenzhen People's Hospital (The second Clinical Medical College of Jinan University), Shenzhen, China
| | - Matthias Leisegang
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Boya Deng
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Tatsuo Matsuda
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Kazuma Kiyotani
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Taigo Kato
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Makiko Harada
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jae-Hyun Park
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | | | - Tanguy Seiwert
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Everett Vokes
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Nishant Agrawal
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Yusuke Nakamura
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,Department of Surgery, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
30
|
Cui JH, Lin KR, Yuan SH, Jin YB, Chen XP, Su XK, Jiang J, Pan YM, Mao SL, Mao XF, Luo W. TCR Repertoire as a Novel Indicator for Immune Monitoring and Prognosis Assessment of Patients With Cervical Cancer. Front Immunol 2018; 9:2729. [PMID: 30524447 PMCID: PMC6262070 DOI: 10.3389/fimmu.2018.02729] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 11/05/2018] [Indexed: 01/22/2023] Open
Abstract
There is increasing evidence that deep sequencing-based T cell repertoire can sever as a biomarker of immune response in cancer patients; however, the characteristics of T cell repertoire including diversity and similarity, as well as its prognostic significance in patients with cervical cancer (CC) remain unknown. In this study, we applied a high throughput T cell receptor (TCR) sequencing method to characterize the T cell repertoires of peripheral blood samples from 25 CC patients, 30 cervical intraepithelial neoplasia (CIN) patients and 20 healthy women for understanding the immune alterations during the cervix carcinogenesis. In addition, we also explored the signatures of TCR repertoires in the cervical tumor tissues and paired sentinel lymph nodes from 16 CC patients and their potential value in predicting the prognosis of patients. Our results revealed that the diversity of circulating TCR repertoire gradually decreased during the cervix carcinogenesis and progression, but the circulating TCR repertoires in CC patients were more similar to CIN patients than healthy women. Interestingly, several clonotypes uniquely detected in CC patients tended to share similar CDR3 motifs, which differed from those observed in CIN patients. In addition, the TCR repertoire diversity in sentinel lymphatic nodes from CC patients was higher than in tumor tissues. More importantly, less clonotypes in TCR repertoire of sentinel lymphatic node was associated with the poor prognosis of the patients. Overall, our findings suggested that TCR repertoire might be a potential indicator of immune monitoring and a biomarker for predicting the prognosis of CC patients. Although functional studies of T cell populations are clearly required, this study have expanded our understanding of T cell immunity during the development of CC and provided an experimental basis for further studies on its pathogenesis and immunotherapy.
Collapse
Affiliation(s)
- Jin-Huan Cui
- Clinical Research Institute, The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan, China
| | - Kai-Rong Lin
- Clinical Research Institute, The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan, China
| | - Song-Hua Yuan
- Department of Gynecology, The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan, China
| | - Ya-Bin Jin
- Clinical Research Institute, The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan, China
| | - Xiang-Ping Chen
- Clinical Research Institute, The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan, China
| | - Xi-Kang Su
- Department of Clinical Laboratory, The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan, China
| | - Jun Jiang
- Department of Abdominothoracic Radiotherapy, The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan, China
| | - Ying-Ming Pan
- Clinical Research Institute, The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan, China
| | - Shao-Long Mao
- Clinical Research Institute, The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan, China
| | - Xiao-Fan Mao
- Clinical Research Institute, The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan, China
| | - Wei Luo
- Clinical Research Institute, The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan, China
| |
Collapse
|
31
|
Takeda K, Kitaura K, Suzuki R, Owada Y, Muto S, Okabe N, Hasegawa T, Osugi J, Hoshino M, Tsunoda T, Okumura K, Suzuki H. Quantitative T-cell repertoire analysis of peripheral blood mononuclear cells from lung cancer patients following long-term cancer peptide vaccination. Cancer Immunol Immunother 2018; 67:949-964. [PMID: 29568993 PMCID: PMC11028142 DOI: 10.1007/s00262-018-2152-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 03/13/2018] [Indexed: 12/14/2022]
Abstract
Therapeutic cancer peptide vaccination is an immunotherapy designed to elicit cytotoxic T-lymphocyte (CTL) responses in patients. A number of therapeutic vaccination trials have been performed, nevertheless there are only a few reports that have analyzed the T-cell receptors (TCRs) expressed on tumor antigen-specific CTLs. Here, we use next-generation sequencing (NGS) to analyze TCRs of vaccine-induced CTL clones and the TCR repertoire of bulk T cells in peripheral blood mononuclear cells (PBMCs) from two lung cancer patients over the course of long-term vaccine therapy. In both patients, vaccination with two epitope peptides derived from cancer/testis antigens (upregulated lung cancer 10 (URLC10) and cell division associated 1 (CDCA1)) induced specific CTLs expressing various TCRs. All URLC10-specific CTL clones tested showed Ca2+ influx, IFN-γ production, and cytotoxicity when co-cultured with URLC10-pulsed tumor cells. Moreover, in CTL clones that were not stained with the URLC10/MHC-multimer, the CD3 ζ chain was not phosphorylated. NGS of the TCR repertoire of bulk PBMCs demonstrated that the frequency of vaccine peptide-specific CTL clones was near the minimum detectable threshold level. These results demonstrate that vaccination induces antigen-specific CTLs expressing various TCRs at different time points in cancer patients, and that some CTL clones are maintained in PBMCs during long-term treatment, including some with TCRs that do not bind peptide/MHC-multimer.
Collapse
Affiliation(s)
- Kazuyoshi Takeda
- Division of Cell Biology, Biomedical Research Center, Graduate School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Department of Biofunctional Micribiota, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Kazutaka Kitaura
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, National Hospital Organization, Sagamihara, Kanagawa, 252-0392, Japan
| | - Ryuji Suzuki
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, National Hospital Organization, Sagamihara, Kanagawa, 252-0392, Japan
| | - Yuki Owada
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Satoshi Muto
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Naoyuki Okabe
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Takeo Hasegawa
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Jun Osugi
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Mika Hoshino
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Takuya Tsunoda
- Department of Clinical Immuno-oncology, Showa University, Setagaya-ku, Tokyo, 157-8577, Japan
| | - Ko Okumura
- Department of Biofunctional Micribiota, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, 113-8421, Japan
- Atopy (Allergy) Research Center, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hiroyuki Suzuki
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| |
Collapse
|
32
|
Zewde M, Kiyotani K, Park JH, Fang H, Yap KL, Yew PY, Alachkar H, Kato T, Mai TH, Ikeda Y, Matsuda T, Liu X, Ren L, Deng B, Harada M, Nakamura Y. The era of immunogenomics/immunopharmacogenomics. J Hum Genet 2018; 63:865-875. [PMID: 29785006 DOI: 10.1038/s10038-018-0468-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 11/09/2022]
Abstract
Although germline alterations and somatic mutations in disease cells have been extensively analyzed, molecular changes in immune cells associated with disease conditions have not been characterized in depth. It is clear that our immune system has a critical role in various biological and pathological conditions, such as infectious diseases, autoimmune diseases, drug-induced skin and liver toxicity, food allergy, and rejection of transplanted organs. The recent development of cancer immunotherapies, particularly drugs modulating the immune checkpoint molecules, has clearly demonstrated the importance of host immune cells in cancer treatments. However, the molecular mechanisms by which these new therapies kill tumor cells are still not fully understood. In this regard, we have begun to explore the role of newly developed tools such as next-generation sequencing in the genetic characterization of both cancer cells and host immune cells, a field that is called immunogenomics/ immunopharmacogenomics. This new field has enormous potential to help us better understand changes in our immune system during the course of various disease conditions. Here we report the potential of deep sequencing of T-cell and B-cell receptors in capturing the molecular contribution of the immune system, which we believe plays critical roles in the pathogenesis of various human diseases.
Collapse
Affiliation(s)
- Makda Zewde
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Kazuma Kiyotani
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA.,Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Jae-Hyun Park
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Hua Fang
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Kai Lee Yap
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Poh Yin Yew
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Houda Alachkar
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Taigo Kato
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Tu H Mai
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Yuji Ikeda
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Tatsuo Matsuda
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Xiao Liu
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Lili Ren
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Boya Deng
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Makiko Harada
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Yusuke Nakamura
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
33
|
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: 8.7] [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]
|
34
|
Santa-Maria CA, Kato T, Park JH, Kiyotani K, Rademaker A, Shah AN, Gross L, Blanco LZ, Jain S, Flaum L, Tellez C, Stein R, Uthe R, Gradishar WJ, Cristofanilli M, Nakamura Y, Giles FJ. A pilot study of durvalumab and tremelimumab and immunogenomic dynamics in metastatic breast cancer. Oncotarget 2018; 9:18985-18996. [PMID: 29721177 PMCID: PMC5922371 DOI: 10.18632/oncotarget.24867] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/06/2018] [Indexed: 12/31/2022] Open
Abstract
Immune checkpoint inhibitors produce modest responses in metastatic breast cancer, however, combination approaches may improve responses. A single arm pilot study was designed to determine the overall response rate (ORR) of durvalumab and tremelimumab, and evaluate immunogenomic dynamics in metastatic endocrine receptor (ER) positive or triple negative breast cancer (TNBC). Simon two-stage design indicated at least four responses from the first 18 patients were needed to proceed with the second stage. T-cell receptor (TCR) sequencing and immune-gene expression profiling were conducted at baseline and two months, whole exome sequencing was conducted at baseline. Eighteen evaluable patients were accrued (11 ER-positive; seven TNBC). Only three patients had a response (ORR = 17%), thus the study did not proceed to the second stage. Responses were only observed in patients with TNBC (ORR = 43%). Responders versus non-responders had upregulation of CD8, granzyme A, and perforin 1 gene expression, and higher mutational and neoantigen burden. Patients with TNBC had an oligoclonal shift of the most abundant TCR-beta clonotypes compared to those with ER-positive disease, p = 0.004. We conclude responses are low in unselected metastatic breast cancer, however, higher rates of clinical benefit were observed in TNBC. Immunogenomic dynamics may help identify phenotypes most likely to respond to immunotherapy.
Collapse
Affiliation(s)
| | - Taigo Kato
- The University of Chicago, Department of Medicine, Chicago, Illinois, USA
| | - Jae-Hyun Park
- The University of Chicago, Department of Medicine, Chicago, Illinois, USA
| | - Kazuma Kiyotani
- The University of Chicago, Department of Medicine, Chicago, Illinois, USA
| | - Alfred Rademaker
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Ami N. Shah
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Leeaht Gross
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Luis Z. Blanco
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Sarika Jain
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Lisa Flaum
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Claudia Tellez
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Regina Stein
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Regina Uthe
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| | - William J. Gradishar
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Massimo Cristofanilli
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Yusuke Nakamura
- The University of Chicago, Department of Medicine, Chicago, Illinois, USA
- The University of Chicago, Department of Surgery, Chicago, Illinois, USA
| | - Francis J. Giles
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| |
Collapse
|
35
|
Durgeau A, Virk Y, Corgnac S, Mami-Chouaib F. Recent Advances in Targeting CD8 T-Cell Immunity for More Effective Cancer Immunotherapy. Front Immunol 2018; 9:14. [PMID: 29403496 PMCID: PMC5786548 DOI: 10.3389/fimmu.2018.00014] [Citation(s) in RCA: 322] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/04/2018] [Indexed: 12/18/2022] Open
Abstract
Recent advances in cancer treatment have emerged from new immunotherapies targeting T-cell inhibitory receptors, including cytotoxic T-lymphocyte associated antigen (CTLA)-4 and programmed cell death (PD)-1. In this context, anti-CTLA-4 and anti-PD-1 monoclonal antibodies have demonstrated survival benefits in numerous cancers, including melanoma and non-small-cell lung carcinoma. PD-1-expressing CD8+ T lymphocytes appear to play a major role in the response to these immune checkpoint inhibitors (ICI). Cytotoxic T lymphocytes (CTL) eliminate malignant cells through recognition by the T-cell receptor (TCR) of specific antigenic peptides presented on the surface of cancer cells by major histocompatibility complex class I/beta-2-microglobulin complexes, and through killing of target cells, mainly by releasing the content of secretory lysosomes containing perforin and granzyme B. T-cell adhesion molecules and, in particular, lymphocyte-function-associated antigen-1 and CD103 integrins, and their cognate ligands, respectively, intercellular adhesion molecule 1 and E-cadherin, on target cells, are involved in strengthening the interaction between CTL and tumor cells. Tumor-specific CTL have been isolated from tumor-infiltrating lymphocytes and peripheral blood lymphocytes (PBL) of patients with varied cancers. TCRβ-chain gene usage indicated that CTL identified in vitro selectively expanded in vivo at the tumor site compared to autologous PBL. Moreover, functional studies indicated that these CTL mediate human leukocyte antigen class I-restricted cytotoxic activity toward autologous tumor cells. Several of them recognize truly tumor-specific antigens encoded by mutated genes, also known as neoantigens, which likely play a key role in antitumor CD8 T-cell immunity. Accordingly, it has been shown that the presence of T lymphocytes directed toward tumor neoantigens is associated with patient response to immunotherapies, including ICI, adoptive cell transfer, and dendritic cell-based vaccines. These tumor-specific mutation-derived antigens open up new perspectives for development of effective second-generation therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Aurélie Durgeau
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France.,ElyssaMed, Paris Biotech Santé, Paris, France
| | - Yasemin Virk
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
36
|
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: 36] [Impact Index Per Article: 6.0] [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
|
37
|
Tsuruta M, Ueda S, Yew PY, Fukuda I, Yoshimura S, Kishi H, Hamana H, Hirayama M, Yatsuda J, Irie A, Senju S, Yuba E, Kamba T, Eto M, Nakayama H, Nishimura Y. Bladder cancer-associated cancer-testis antigen-derived long peptides encompassing both CTL and promiscuous HLA class II-restricted Th cell epitopes induced CD4 + T cells expressing converged T-cell receptor genes in vitro. Oncoimmunology 2018; 7:e1415687. [PMID: 29632734 DOI: 10.1080/2162402x.2017.1415687] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/02/2017] [Accepted: 12/04/2017] [Indexed: 10/18/2022] Open
Abstract
DEP domain containing 1 (DEPDC1) and M-phase phosphoprotein 1 (MPHOSPH1) are human cancer testis antigens that are frequently overexpressed in urinary bladder cancer. In a phase I/II clinical trial, a DEPDC1- and MPHOSPH1-derived short peptide vaccine demonstrated promising efficacy in preventing bladder cancer recurrence. Here, we aimed to identify long peptides (LPs) derived from DEPDC1 and MPHOSPH1 that induced both T-helper (Th) cells and tumor-reactive cytotoxic T lymphocytes (CTLs). Stimulation of peripheral blood mononuclear cells (PBMCs) from healthy donors with the synthetic DEPDC1- and MPHOSPH1-LPs predicted to bind to promiscuous human leukocyte antigen (HLA) class II molecules by a computer algorithm induced specific CD4+ T cells as revealed by interferon-γ enzyme-linked immunospot assays. Three of six LPs encompassed HLA-A2- or -A24-restricted CTL epitopes or both, and all six LPs stimulated DEPDC1- or MPHOSPH1-specific Th cells restricted by promiscuous and frequently observed HLA class II molecules in the Japanese population. Some LPs are naturally processed from the proteins in DCs, and the capacity of these LPs to cross-prime CTLs was confirmed in vivo using HLA-A2 or -A24 transgenic mice. The LP-specific and HLA class II-restricted T-cell responses were also observed in PBMCs from patients with bladder cancer. Repeated stimulation of PBMCs with DEPDC1-LPs and MPHOSPH1-LPs yielded clonal Th cells expressing specific T-cell receptor (TCR)-α and β genes. These DEPDC1- or MPHOSPH1-derived LPs may have applications in immunotherapy in patients with bladder cancer, and the TCR genes identified may be useful for monitoring of Th cells specific to LPs in vivo.
Collapse
Affiliation(s)
- Miki Tsuruta
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan.,Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan
| | - Shohei Ueda
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan.,Department of Urology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Poh Yin Yew
- Tumor Immunoanalysis Department, OncoTherapy Science, Inc., Sakado, Takatsu-ku, Kawasaki, Kanagawa, Japan
| | - Isao Fukuda
- Tumor Immunoanalysis Department, OncoTherapy Science, Inc., Sakado, Takatsu-ku, Kawasaki, Kanagawa, Japan
| | - Sachiko Yoshimura
- Tumor Immunoanalysis Department, OncoTherapy Science, Inc., Sakado, Takatsu-ku, Kawasaki, Kanagawa, Japan
| | - Hiroyuki Kishi
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences (Medicine), University of Toyama, Sugitani, Toyama, Toyama, Japan
| | - Hiroshi Hamana
- Department of Innovative Cancer Immunotherapy, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Toyama, Japan
| | - Masatoshi Hirayama
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan.,Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan
| | - Junji Yatsuda
- Department of Urology, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan
| | - Atsushi Irie
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan
| | - Satoru Senju
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan
| | - Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, Gakuen-cho, Naka-ku, Sakai, Osaka, Japan
| | - Tomomi Kamba
- Department of Urology, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan.,Department of Urology, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan
| | - Hideki Nakayama
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan
| | - Yasuharu Nishimura
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan.,Nishimura Project Laboratory, Center for Resource Development and Analysis, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan
| |
Collapse
|
38
|
Ren L, Matsuda T, Deng B, Kiyotani K, Kato T, Park JH, Seiwert TY, Vokes EE, Agrawal N, Nakamura Y. Similarity and difference in tumor-infiltrating lymphocytes in original tumor tissues and those of in vitro expanded populations in head and neck cancer. Oncotarget 2017; 9:3805-3814. [PMID: 29423084 PMCID: PMC5790501 DOI: 10.18632/oncotarget.23454] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 12/10/2017] [Indexed: 01/04/2023] Open
Abstract
Though adoptive tumor-infiltrating lymphocyte (TIL) therapy has been explored in clinical trials for many years, there is little information for the clonotype composition between TILs in original tumor tissues and TILs that were in vitro expanded and infused to cancer patients. To investigate the similarity/difference in TILs in original tumor tissues and those of in vitro expanded populations in squamous cell carcinoma of head and neck (SCCHN) as well as their correlation with somatic mutations in cancer cells, we performed whole exome analysis, expression profile analysis of immune-related genes, and T cell receptor (TCR) analysis of original TILs and in vitro expanded TILs in 8 surgically-resected HPV-negative fresh tumors with SCCHN. We found an unusually high number of non-synonymous somatic mutations (4290, 1779 and 901 mutations) in three SCCHN tumors, in which we identified mutations in mismatch repair genes, MSH2 or MSH4, or a DNA polymerase gene, POLE. Interestingly, dominant TCR clonotypes of expanded CD8+ TILs derived from these three tumors revealed high similarity to those in original tumors while for remaining tumors with the lower mutational load, we found that T cell clonotypes between TILs in original tumor tissues and those expanded in vitro were almost entirely different. Our findings might provide clinically useful information for identification of tumor-antigen-specific T cell clones that may lead to further improvement of adoptive TIL therapy for SCCHN patients.
Collapse
Affiliation(s)
- Lili Ren
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Cytotherapy Laboratory, Shenzhen People's Hospital (The second Clinical Medical College of Jinan University), Shenzhen, China
| | - Tatsuo Matsuda
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Boya Deng
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Kazuma Kiyotani
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Taigo Kato
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jae-Hyun Park
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Tanguy Y Seiwert
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Everett E Vokes
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Nishant Agrawal
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Yusuke Nakamura
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Department of Surgery, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
39
|
Characterization of the B-cell receptor repertoires in peanut allergic subjects undergoing oral immunotherapy. J Hum Genet 2017; 63:239-248. [PMID: 29192240 DOI: 10.1038/s10038-017-0364-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/09/2017] [Accepted: 09/09/2017] [Indexed: 12/12/2022]
Abstract
B-cell receptors (BCRs) play a critical role in adaptive immunity as they generate highly diverse immunoglobulin repertoires to recognize a wide variety of antigens. To better understand immune responses, it is critically important to establish a quantitative and rapid method to analyze BCR repertoire comprehensively. Here, we developed "Bcrip", a novel approach to characterize BCR repertoire by sequencing millions of BCR cDNA using next-generation sequencer. Using this method and quantitative real-time PCR, we analyzed expression levels and repertoires of BCRs in a total of 17 peanut allergic subjects' peripheral blood samples before and after receiving oral immunotherapy (OIT) or placebo. By our methods, we successfully identified all of variable (V), joining (J), and constant (C) regions, in an average of 79.1% of total reads and 99.6% of these VJC-mapped reads contained the C region corresponding to the isotypes that we aimed to analyze. In the 17 peanut allergic subjects' peripheral blood samples, we observed an oligoclonal enrichment of certain immunoglobulin heavy chain alpha (IGHA) and IGH gamma (IGHG) clones (P = 0.034 and P = 0.027, respectively) in peanut allergic subjects after OIT. This newly developed BCR sequencing and analysis method can be applied to investigate B-cell repertoires in various research areas, including food allergies as well as autoimmune and infectious diseases.
Collapse
|
40
|
Rosati E, Dowds CM, Liaskou E, Henriksen EKK, Karlsen TH, Franke A. Overview of methodologies for T-cell receptor repertoire analysis. BMC Biotechnol 2017; 17:61. [PMID: 28693542 PMCID: PMC5504616 DOI: 10.1186/s12896-017-0379-9] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 06/26/2017] [Indexed: 12/13/2022] Open
Abstract
Background The T-cell receptor (TCR), located on the surface of T cells, is responsible for the recognition of the antigen-major histocompatibility complex, leading to the initiation of an inflammatory response. Analysing the TCR repertoire may help to gain a better understanding of the immune system features and of the aetiology and progression of diseases, in particular those with unknown antigenic triggers. The extreme diversity of the TCR repertoire represents a major analytical challenge; this has led to the development of specialized methods which aim to characterize the TCR repertoire in-depth. Currently, next generation sequencing based technologies are most widely employed for the high-throughput analysis of the immune cell repertoire. Results Here, we report on the latest methodological advancements in the field by describing and comparing the available tools; from the choice of the starting material and library preparation method, to the sequencing technologies and data analysis. Finally, we provide a practical example and our own experience by reporting some exemplary results from a small internal benchmark study, where current approaches from the literature and the market are employed and compared. Conclusions Several valid methods for clonotype identification and TCR repertoire analysis exist, however, a gold standard method for the field has not yet been identified. Depending on the purpose of the scientific study, some approaches may be more suitable than others. Finally, due to possible method specific biases, scientists must be careful when comparing results obtained using different methods. Electronic supplementary material The online version of this article (doi:10.1186/s12896-017-0379-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elisa Rosati
- Institute of Clinical Molecular Biology, Kiel University, Rosalind-Franklin-Str. 12, 24105, Kiel, Germany
| | - C Marie Dowds
- Institute of Clinical Molecular Biology, Kiel University, Rosalind-Franklin-Str. 12, 24105, Kiel, Germany
| | - Evaggelia Liaskou
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research Unit, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Eva Kristine Klemsdal Henriksen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Tom H Karlsen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Section of Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Rosalind-Franklin-Str. 12, 24105, Kiel, Germany.
| |
Collapse
|
41
|
Integrated analysis of somatic mutations and immune microenvironment of multiple regions in breast cancers. Oncotarget 2017; 8:62029-62038. [PMID: 28977923 PMCID: PMC5617483 DOI: 10.18632/oncotarget.18790] [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: 04/28/2017] [Accepted: 05/20/2017] [Indexed: 12/21/2022] Open
Abstract
Next-generation sequencing technology enables us to analyze the complexity of intra- and inter-tumoral heterogeneity, which may influence to prognosis of cancer patients. In this study, we collected surgically-resected tumor tissues from five breast cancer patients and characterized three different portions of individual tumors through somatic mutation analysis by whole exome sequencing, T cell receptor beta (TCRB) repertoire analysis of tumor-infiltrating lymphocytes (TILs), and the expression analysis of immune-related genes at 15 different sites. This integrated analysis revealed distinguished patterns of somatic mutations and TIL clonotypes in the three portions of each tumor, implying that the tumor heterogeneity is comprised by spatially different somatic mutations as well as the presence of diverse T cell clones. Furthermore, higher numbers of the non-synonymous somatic mutations were significantly correlated with the higher ratio of GZMA/TCRB expression (P = 0.0004), implying that high somatic mutation load in tumor might be correlated to the number of immunogenic antigens and then functionally activate TILs with higher cytolytic activity. Our findings suggest that breast cancers comprise with very complex tumor heterogeneity by the spatially different mutational landscape and immune microenvironment, and that mutation/neoantigen load may be strongly correlated with induction of cancer-specific TILs and affect the immune microenvironment in breast tumors.
Collapse
|
42
|
Kato T, Iwasaki T, Uemura M, Nagahara A, Higashihara H, Osuga K, Ikeda Y, Kiyotani K, Park JH, Nonomura N, Nakamura Y. Characterization of the cryoablation-induced immune response in kidney cancer patients. Oncoimmunology 2017; 6:e1326441. [PMID: 28811963 DOI: 10.1080/2162402x.2017.1326441] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/26/2017] [Accepted: 04/29/2017] [Indexed: 10/19/2022] Open
Abstract
Cryoablation is one of treatment modalities for kidney cancer and is expected to induce strong local immune responses as well as systemic T-cell-mediated immune reactions that may lead to the regression of distant metastatic lesions. Thus, the characterization of T cell repertoire and immune environment in tumors before and after treatment should contribute to the better understanding of the cryoablation-induced anticancer immune responses. In this study, we collected tumor tissues from 22 kidney cancer patients, before cryoablation and at 3 mo after cryoablation. In addition, blood samples were collected from 14 patients at the same time points. We applied a next generation sequencing approach to characterize T cell receptor β (TCRB) repertoires using RNAs isolated from tumor tissues and peripheral blood mononuclear cells. TCRB repertoire analysis revealed the expansion of certain T cell clones in tumor tissues by cryoablation. We also found that proportions of abundant TCRB clonotypes (defined as clonotypes with ≥ 1% frequency among total TCRB reads) were significantly increased in the post-cryoablation tissue samples than those of pre-cryoablation tumor samples. Some of these TCRB clonotypes were found to be increased in peripheral blood. Expression analysis of immune-related genes in the tissues of pre- and post-cryoablation showed significantly elevated transcriptional levels of CD8+ , CD4+ , Granzyme A (GZMA), and CD11c along with a high CD8/FOXP3 ratio in the post-cryoablation tissue samples. Our findings revealed that cryoablation could induce strong immune reactions in tumors with oligoclonal expansion of antitumor T cells, which circulate systemically.
Collapse
Affiliation(s)
- Taigo Kato
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Tomoyuki Iwasaki
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Motohide Uemura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akira Nagahara
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroki Higashihara
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keigo Osuga
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuji Ikeda
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Kazuma Kiyotani
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jae-Hyun Park
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Nakamura
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Department of Surgery, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
43
|
Mai T, Takano A, Suzuki H, Hirose T, Mori T, Teramoto K, Kiyotani K, Nakamura Y, Daigo Y. Quantitative analysis and clonal characterization of T-cell receptor β repertoires in patients with advanced non-small cell lung cancer treated with cancer vaccine. Oncol Lett 2017; 14:283-292. [PMID: 28693166 DOI: 10.3892/ol.2017.6125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 03/03/2017] [Indexed: 12/31/2022] Open
Abstract
With the development of cancer immunotherapy that may activate T cells, a practical and quantitative method to improve monitoring and/or prediction of immunological response of patients as a predictive biomarker is of importance. To examine possible biomarkers for a therapeutic cancer vaccine containing a mixture of three epitope peptides derived from cell division-associated 1, lymphocyte antigen 6 complex locus K and insulin-like growth factor-II mRNA-binding protein 3, T-cell receptor β (TCRβ) repertoires of blood samples from 24 patients with human leukocyte antigen-A*2402-positive non-small cell lung cancer were characterized prior to and following 8 weeks of the cancer vaccine treatment, by applying a next-generation sequencing method. It was identified that 14 patients with overall survival (OS) times of ≥12 months had significantly lower TCRβ diversity indexes in samples prior to treatment, compared with 10 patients who succumbed within 1 year (P=0.03). In addition, patients with a high level of activated CD8+ T cells that are defined by a high granzyme A/CD8 ratio had favorable OS rates (log-rank test, P=0.04). The TCRβ diversity index and immunogenic gene markers following vaccine administration may serve as predictive or monitoring biomarkers for cancer vaccine treatment.
Collapse
Affiliation(s)
- Tu Mai
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, IL 60637, USA.,Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Atsushi Takano
- Department of Medical Oncology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.,Cancer Center, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Hiroyuki Suzuki
- Department of Regenerative Surgery, Fukushima Medical University, School of Medicine, Fukushima 960-1295, Japan
| | - Takashi Hirose
- Division of Respiratory Medicine and Allergology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Takahiro Mori
- Tohoku Community Cancer Services Program, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8574, Japan
| | - Koji Teramoto
- Department of Medical Oncology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.,Cancer Center, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Kazuma Kiyotani
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Yusuke Nakamura
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, IL 60637, USA.,Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.,Department of Surgery, University of Chicago, Chicago, IL 60637, USA
| | - Yataro Daigo
- Department of Medical Oncology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.,Cancer Center, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| |
Collapse
|
44
|
Saloura V, Fatima A, Zewde M, Kiyotani K, Brisson R, Park JH, Ikeda Y, Vougiouklakis T, Bao R, Khattri A, Seiwert T, Cipriani N, Lingen M, Vokes E, Nakamura Y. Characterization of the T-Cell Receptor Repertoire and Immune Microenvironment in Patients with Locoregionally Advanced Squamous Cell Carcinoma of the Head and Neck. Clin Cancer Res 2017; 23:4897-4907. [DOI: 10.1158/1078-0432.ccr-17-0103] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/15/2017] [Accepted: 04/20/2017] [Indexed: 11/16/2022]
|
45
|
Kimura T, Fukushima S, Miyashita A, Aoi J, Jinnin M, Kosaka T, Ando Y, Matsukawa M, Inoue H, Kiyotani K, Park JH, Nakamura Y, Ihn H. Myasthenic crisis and polymyositis induced by one dose of nivolumab. Cancer Sci 2017; 107:1055-8. [PMID: 27420474 PMCID: PMC4946722 DOI: 10.1111/cas.12961] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 04/21/2016] [Accepted: 04/23/2016] [Indexed: 11/30/2022] Open
Abstract
An 80-year-old man, who developed multiple lymph node and skin metastasis of malignant melanoma, received nivolumab monotherapy. Two weeks after the first dose, he experienced anorexia and fatigue, and suffered from progressive, severe dyspnea and muscle weakness. We diagnosed him with myocarditis, myositis, and myasthenic crisis induced by nivolumab. We commenced steroid therapy, immune absorption therapy, plasma exchange therapy, and i.v. immunoglobulin therapy, and succeeded in saving his life. Because his serum level of anti-acetylcholine receptor antibodies in a sample collected before nivolumab treatment were positive and were elevated significantly after nivolumab, we suspected that nivolumab triggered a severe autoimmune response, which progressed subclinical myasthenia gravis to myasthenic crisis. We carried out T cell receptor repertoire analysis using next-generation sequencing technologies and identified infiltration of clonally expanded T cell populations in the skeletal muscle after nivolumab treatment, implying a very strong T cell immune response against muscular cells. To avoid severe immune-related adverse events, the exclusion of patients with subclinical autoimmune disease is very important for treatment with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Toshihiro Kimura
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Azusa Miyashita
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Jun Aoi
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Masatoshi Jinnin
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takayuki Kosaka
- Department of Neurology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yukio Ando
- Department of Neurology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Masakazu Matsukawa
- Cardiovascular Center, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Hiroyuki Inoue
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Kazuma Kiyotani
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Jae-Hyun Park
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Yusuke Nakamura
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
46
|
Kiyotani K, Park JH, Inoue H, Husain A, Olugbile S, Zewde M, Nakamura Y, Vigneswaran WT. Integrated analysis of somatic mutations and immune microenvironment in malignant pleural mesothelioma. Oncoimmunology 2017; 6:e1278330. [PMID: 28344893 DOI: 10.1080/2162402x.2016.1278330] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/27/2016] [Accepted: 12/28/2016] [Indexed: 12/29/2022] Open
Abstract
To investigate the link between the genomic landscape of cancer cells and immune microenvironment in tumor tissues, we characterized somatic mutations and tumor-infiltrating lymphocytes (TILs) in malignant pleural mesothelioma (MPM), including mutation/neoantigen load, spatial heterogeneity of somatic mutations of cancer cells and TILs (T-cell receptor β (TCRβ) repertoire), and expression profiles of immune-related genes using specimens of three different tumor sites (anterior, posterior, and diaphragm) obtained from six MPM patients. Integrated analysis identified the distinct patterns of somatic mutations and the immune microenvironment signatures both intratumorally and interindividually. MPM cases showed intratumoral heterogeneity in somatic mutations with unique TCRβ clonotypes of TILs that were restricted to each tumor site, suggesting the presence of a neoantigen-related immune response. Correlation analyses showed that higher neoantigen load was significantly correlated with stronger clonal expansion of TILs (p = 0.048) and a higher expression level of an immune-associated cytolytic factor (PRF1 (p = 0.0041) in tumor tissues), suggesting that high neoantigen loads in tumor cells might promote expansion of functional tumor-specific T cells in the tumor bed. Our results collectively indicate that MPM tumors constitute a diverse heterogeneity in both the genomic landscape and immune microenvironment, and that mutation/neoantigen load may affect the immune microenvironment in MPM tissues.
Collapse
Affiliation(s)
- Kazuma Kiyotani
- Department of Medicine, The University of Chicago , Chicago, IL, USA
| | - Jae-Hyun Park
- Department of Medicine, The University of Chicago , Chicago, IL, USA
| | - Hiroyuki Inoue
- Department of Medicine, The University of Chicago , Chicago, IL, USA
| | - Aliya Husain
- Department of Pathology, The University of Chicago , Chicago, IL, USA
| | - Sope Olugbile
- Department of Medicine, The University of Chicago , Chicago, IL, USA
| | - Makda Zewde
- Department of Medicine, The University of Chicago , Chicago, IL, USA
| | - Yusuke Nakamura
- Department of Medicine, The University of Chicago, Chicago, IL, USA; Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Wickii T Vigneswaran
- Department of Surgery, The University of Chicago, Chicago, IL, USA; Department of Thoracic and Cardiovascular Surgery, Loyola University Medical Center, Maywood, IL, USA
| |
Collapse
|
47
|
High Expression of KIF20A Is Associated with Poor Overall Survival and Tumor Progression in Early-Stage Cervical Squamous Cell Carcinoma. PLoS One 2016; 11:e0167449. [PMID: 27941992 PMCID: PMC5152822 DOI: 10.1371/journal.pone.0167449] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/14/2016] [Indexed: 12/05/2022] Open
Abstract
Background The kinesin family member 20a (KIF20A) protein has been implicated in the development and progression of many human cancers; however, its precise function and role in cervical cancer remain largely unclear. This study aimed to investigate the expression profile and prognostic value of KIF20A in patients with early-stage cervical squamous cell carcinoma. Methods We examined the mRNA and protein levels of KIF20A in eight cervical cancer cell lines and eight paired cervical cancer samples, compared with normal cervical epithelial cells and adjacent normal cervical tissues, respectively. Immunohistochemistry was performed to detect the expression of KIF20A in paraffin-embedded specimens from 169 early-stage cervical squamous cell carcinoma patients. Statistical analyses were applied to analyze the association between KIF20A expression and clinical variables, as well with patient survival. Results The mRNA and protein expression levels of KIF20A were significantly elevated in cervical cancer cell lines and lesions compared with normal cells and corresponding normal tissues (P < 0.05). Immunohistochemistry analysis in 169 cervical cancer cases revealed that increased KIF20A expression was strongly associated with human papillomavirus (HPV) infection (P = 0.008), clinical stage (P = 0.001), tumor recurrence (P = 0.016), vital status (P < 0.001), the property of the surgical margin (P = 0.032), the lymphovascular space involvement (P = 0.014), and pelvic lymph node metastasis (P = 0.001). The overall survival and disease-free survival of patients with high levels of KIF20A expression were significantly poorer than those with low KIF20A expression. KIF20A was an independent survival prognostic factor, as evidenced by univariate and multivariate analysis. Conclusions Our results illustrate that elevated KIF20A expression correlates with HPV infection, clinical stage, tumor recurrence, lymphovascular space involvement, pelvic lymph node metastasis, and poor outcome in early-stage cervical squamous cell carcinoma patients. KIF20A aberrant expression is a novel independent unfavorable prognostic factor and may present a potential therapeutic target for cervical cancer.
Collapse
|
48
|
Alachkar H, Mutonga M, Kato T, Kalluri S, Kakuta Y, Uemura M, Imamura R, Nonomura N, Vujjini V, Alasfar S, Rabb H, Nakamura Y, Alachkar N. Quantitative characterization of T-cell repertoire and biomarkers in kidney transplant rejection. BMC Nephrol 2016; 17:181. [PMID: 27871261 PMCID: PMC5117555 DOI: 10.1186/s12882-016-0395-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 11/09/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND T-cell-mediated rejection (TCMR) remains a major cause of kidney allograft failure. The characterization of T-cell repertoire in different immunological disorders has emerged recently as a novel tool with significant implications. We herein sought to characterize T-cell repertoire using next generation sequencing to diagnose TCMR. METHODS In this prospective study, we analyzed samples from 50 kidney transplant recipients. We collected blood and kidney transplant biopsy samples at sequential time points before and post transplant. We used next generation sequencing to characterize T-cell receptor (TCR) repertoire by using illumina miSeq on cDNA synthesized from RNA extracted from six patients' samples. We also measured RNA expression levels of FOXP3, CD8, CD4, granzyme and perforin in blood samples from all 50 patients. RESULTS Seven patients developed TCMR during the first three months of the study. Out of six patients who had complete sets of blood and biopsy samples two had TCMR. We found an expansion of the TCR repertoire in blood at time of rejection when compared to that at pre-transplant or one-month post transplant. Patients with TCMR (n = 7) had significantly higher RNA expression levels of FOXP3, Perforin, Granzyme, CD4 and CD8 in blood samples than those with no TCMR (n = 43) (P = 0.02, P = 0.003, P = 0.002, P = 0.017, and P = 0.01, respectively). CONCLUSIONS Our study provides a potential utilization of TCR clone kinetics analysis in the diagnosis of TCMR. This approach may allow for the identification of the expanded T-cell clones associated with the rejection and lead to potential noninvasive diagnosis and targeted therapies of TCMR.
Collapse
Affiliation(s)
- Houda Alachkar
- School of Pharmacy, University of Southern California, Los Angeles, CA, 90089, USA.
| | - Martin Mutonga
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Taigo Kato
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Sowjanya Kalluri
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Kendall regional medical center, Miami, FL, 33175, USA
| | - Yoichi Kakuta
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Motohide Uemura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryoichi Imamura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Vikas Vujjini
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Johns Hopkins Hospital, Baltimore, MD, 21287, USA
| | - Sami Alasfar
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Johns Hopkins Hospital, Baltimore, MD, 21287, USA
| | - Hamid Rabb
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Johns Hopkins Hospital, Baltimore, MD, 21287, USA
| | - Yusuke Nakamura
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Nada Alachkar
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Johns Hopkins Hospital, Baltimore, MD, 21287, USA
| |
Collapse
|
49
|
Xu L, You X, Zheng P, Zhang BM, Gupta PK, Lavori P, Meyer E, Zehnder JL. Methodologic Considerations in the Application of Next-Generation Sequencing of Human TRB Repertoires for Clinical Use. J Mol Diagn 2016; 19:72-83. [PMID: 27815002 DOI: 10.1016/j.jmoldx.2016.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/24/2016] [Accepted: 07/28/2016] [Indexed: 01/08/2023] Open
Abstract
Next-generation sequencing (NGS) of immune receptors has become a standard tool to assess minimal residual disease (MRD) in patients treated for lymphoid malignancy, and it is being used to study the T-cell repertoire in many clinical settings. To better understanding the potential clinical utility and limitations of this application outside of MRD, we developed a BIOMED-2 primer-based NGS method and characterized its performance in controls and patients with graft-versus-host disease (GVHD) after allogeneic hematopoietic transplant. For controls and patients with GVHD, replicate sequencing of the same T-cell receptor β (TRB) libraries was highly reproducible. Higher variability was observed in sequencing of different TRB libraries made from the same DNA stock. Variability was increased in patients with GVHD compared with controls; patients with GVHD also had lower diversity than controls. In the T-cell repertoire of a healthy person, approximately 99.6% of the CDR3 clones were in low abundance, with frequency <10-3. A single library could identify >93% of the clones with frequency ≥10-3 in the repertoire. Sequencing in duplicate increased the average detection rate to >97%. This work demonstrates that NGS reliably and robustly characterizes TRB populations in healthy individuals and patients with GVHD with frequency ≥10-3 and provides a methodologic framework for applying NGS immune repertoire methods to clinical testing applications beyond MRD.
Collapse
Affiliation(s)
- Liwen Xu
- Department of Pathology, Stanford School of Medicine, Stanford University, Stanford, California
| | - Xiaoqing You
- Department of Pathology, Stanford School of Medicine, Stanford University, Stanford, California
| | - PingPing Zheng
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford School of Medicine, Stanford University, Stanford, California
| | - Bing M Zhang
- Department of Pathology, Stanford School of Medicine, Stanford University, Stanford, California
| | - Puja K Gupta
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford School of Medicine, Stanford University, Stanford, California
| | - Philip Lavori
- Department of Biomedical Data Science, Stanford School of Medicine, Stanford University, Stanford, California
| | - Everett Meyer
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford School of Medicine, Stanford University, Stanford, California
| | - James L Zehnder
- Department of Pathology, Stanford School of Medicine, Stanford University, Stanford, California.
| |
Collapse
|
50
|
Hung SJ, Chen YL, Chu CH, Lee CC, Chen WL, Lin YL, Lin MC, Ho CL, Liu T. TRIg: a robust alignment pipeline for non-regular T-cell receptor and immunoglobulin sequences. BMC Bioinformatics 2016; 17:433. [PMID: 27782801 PMCID: PMC5080739 DOI: 10.1186/s12859-016-1304-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 10/21/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND T cells and B cells are essential in the adaptive immunity via expressing T cell receptors and immunoglogulins respectively for recognizing antigens. To recognize a wide variety of antigens, a highly diverse repertoire of receptors is generated via complex recombination of the receptor genes. Reasonably, frequencies of the recombination events have been shown to predict immune diseases and provide insights into the development of immunity. The field is further boosted by high-throughput sequencing and several computational tools have been released to analyze the recombined sequences. However, all current tools assume regular recombination of the receptor genes, which is not always valid in data prepared using a RACE approach. Compared to the traditional multiplex PCR approach, RACE is free of primer bias, therefore can provide accurate estimation of recombination frequencies. To handle the non-regular recombination events, a new computational program is needed. RESULTS We propose TRIg to handle non-regular T cell receptor and immunoglobulin sequences. Unlike all current programs, TRIg does alignments to the whole receptor gene instead of only to the coding regions. This brings new computational challenges, e.g., ambiguous alignments due to multiple hits to repetitive regions. To reduce ambiguity, TRIg applies a heuristic strategy and incorporates gene annotation to identify authentic alignments. On our own and public RACE datasets, TRIg correctly identified non-regularly recombined sequences, which could not be achieved by current programs. TRIg also works well for regularly recombined sequences. CONCLUSIONS TRIg takes into account non-regular recombination of T cell receptor and immunoglobulin genes, therefore is suitable for analyzing RACE data. Such analysis will provide accurate estimation of recombination events, which will benefit various immune studies directly. In addition, TRIg is suitable for studying aberrant recombination in immune diseases. TRIg is freely available at https://github.com/TLlab/trig .
Collapse
Affiliation(s)
- Sheng-Jou Hung
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan City, Taiwan
| | - Yi-Lin Chen
- Molecular Diagnostic Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan City, Taiwan.,Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan City, Taiwan
| | - Chia-Hung Chu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan City, Taiwan
| | - Chuan-Chun Lee
- Molecular Diagnostic Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan City, Taiwan.,Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan City, Taiwan
| | - Wan-Li Chen
- Molecular Diagnostic Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan City, Taiwan.,Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan City, Taiwan
| | - Ya-Lan Lin
- Molecular Diagnostic Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan City, Taiwan.,Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan City, Taiwan
| | - Ming-Ching Lin
- Molecular Diagnostic Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan City, Taiwan.,Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan City, Taiwan
| | - Chung-Liang Ho
- Molecular Diagnostic Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan City, Taiwan.,Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan City, Taiwan
| | - Tsunglin Liu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan City, Taiwan.
| |
Collapse
|