1
|
Meng J, Tan JYT, Joseph CR, Ye J, Lim JCT, Goh D, Xue Y, Lim X, Koh VCY, Wee F, Tay TKY, Chan JY, Ng CCY, Iqbal J, Lau MC, Lim HE, Toh HC, Teh BT, Dent RA, Tan PH, Yeong JPS. The Prognostic Value of CD39 as a Marker of Tumor-Specific T Cells in Triple-Negative Breast Cancer in Asian Women. J Transl Med 2024; 104:100303. [PMID: 38103870 DOI: 10.1016/j.labinv.2023.100303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 11/09/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has a poor prognosis with limited therapeutic options available for affected patients. Efforts are ongoing to identify surrogate markers for tumor-specific CD8+ T cells that can predict the response to immune checkpoint inhibitor (ICI) therapies, such as programmed cell death protein 1 or programmed cell death ligand-1 blockade. We have previously identified tumor-specific CD39+CD8+ T cells in non-small cell lung cancer that might help predict patient responses to programmed cell death protein 1 or programmed cell death ligand-1 blockade. Based on this finding, we conducted a comparative interrogation of TNBC in an Asian cohort to evaluate the potential of CD39 as a surrogate marker of tumor-specific CD8+ T cells. Using ICI-treated TNBC mouse models (n = 24), flow cytometric analyses of peripheral blood mononuclear cells and tumor-infiltrating lymphocytes revealed that >99% of tumor-specific CD8+ T cells also expressed CD39. To investigate the relationship between CD39+CD8+ T-cell density and CD39 expression with disease prognosis, we performed multiplex immunohistochemistry staining on treatment-naive human TNBC tissues (n = 315). We saw that the proportion of CD39+CD8+ T cells in human TNBC tumors correlated with improved overall survival, as did the densities of other CD39+ immune cell infiltrates, such as CD39+CD68+ macrophages. Finally, increased CD39 expression on CD8+ T cells was also found to predict the response to ICI therapy (pembrolizumab) in a separate cohort of 11 TNBC patients. These findings support the potential of CD39+CD8+ T-cell density as a prognostic factor in Asian TNBC patients.
Collapse
Affiliation(s)
- Jia Meng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Jing Ying Tira Tan
- Duke-NUS Medical School, Singapore, Republic of Singapore; National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Craig Ryan Joseph
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Jiangfeng Ye
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Yuezhen Xue
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Xinru Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Valerie Cui Yun Koh
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Republic of Singapore
| | - Felicia Wee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Timothy Kwang Yong Tay
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Republic of Singapore
| | | | | | - Jabed Iqbal
- Duke-NUS Medical School, Singapore, Republic of Singapore; Department of Anatomical Pathology, Singapore General Hospital, Singapore, Republic of Singapore
| | - Mai Chan Lau
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Hsuen Elaine Lim
- National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Han Chong Toh
- National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Bin Tean Teh
- National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Rebecca Alexandra Dent
- Duke-NUS Medical School, Singapore, Republic of Singapore; National Cancer Centre Singapore, Singapore, Republic of Singapore.
| | - Puay Hoon Tan
- KK Women's and Children's Hospital, Singapore, Republic of Singapore; Luma Women's Imaging Centre/Medical Centre, Singapore, Republic of Singapore.
| | - Joe Poh Sheng Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore; Duke-NUS Medical School, Singapore, Republic of Singapore; National Cancer Centre Singapore, Singapore, Republic of Singapore; Department of Anatomical Pathology, Singapore General Hospital, Singapore, Republic of Singapore.
| |
Collapse
|
2
|
Ye K, Li F, Wang R, Cen T, Liu S, Zhao Z, Li R, Xu L, Zhang G, Xu Z, Deng L, Li L, Wang W, Stepanov A, Wan Y, Guo Y, Li Y, Wang Y, Tian Y, Gabibov AG, Yan Y, Zhang H. An armed oncolytic virus enhances the efficacy of tumor-infiltrating lymphocyte therapy by converting tumors to artificial antigen-presenting cells in situ. Mol Ther 2022; 30:3658-3676. [PMID: 35715953 PMCID: PMC9734027 DOI: 10.1016/j.ymthe.2022.06.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/20/2022] [Accepted: 06/13/2022] [Indexed: 12/14/2022] Open
Abstract
The full potential of tumor-infiltrating lymphocyte (TIL) therapy has been hampered by the inadequate activation and low persistence of TILs, as well as inefficient neoantigen presentation by tumors. We transformed tumor cells into artificial antigen-presenting cells (aAPCs) by infecting them with a herpes simplex virus 1 (HSV-1)-based oncolytic virus encoding OX40L and IL12 (OV-OX40L/IL12) to provide local signals for optimum T cell activation. The infected tumor cells displayed increased expression of antigen-presenting cell-related markers and induced enhanced T cell activation and killing in coculture with TILs. Combining OV-OX40L/IL12 and TIL therapy induced complete tumor regression in patient-derived xenograft and syngeneic mouse tumor models and elicited an antitumor immunological memory. In addition, the combination therapy produced aAPC properties in tumor cells, activated T cells, and reprogrammed macrophages to a more M1-like phenotype in the tumor microenvironment. This combination strategy unleashes the full potential of TIL therapy and warrants further evaluation in clinical studies.
Collapse
Affiliation(s)
- Kai Ye
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China; CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China
| | - Fan Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China; CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China
| | - Ruikun Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China; CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China
| | - Tianyi Cen
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China; CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China
| | - Shiyu Liu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China; CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China
| | - Zhuoqian Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China; CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China
| | - Ruonan Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China; CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China
| | - Lili Xu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China; CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China
| | - Guanmeng Zhang
- Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, Tianjin 300041, PR China
| | - Zhaoyuan Xu
- Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, Tianjin 300041, PR China
| | - Li Deng
- CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Beijing Institute of Biological Products, Beijing 100176, PR China
| | - Lili Li
- CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Beijing Institute of Biological Products, Beijing 100176, PR China
| | - Wei Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, PR China
| | - Alexey Stepanov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia
| | - Yajuan Wan
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China
| | - Yu Guo
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China; CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China
| | - Yuanke Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China; CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China
| | - Yuan Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China; CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China
| | - Yujie Tian
- CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China
| | - Alexander G Gabibov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia
| | - Yingbin Yan
- Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, Tianjin 300041, PR China.
| | - Hongkai Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, PR China; CNBG-Nankai University Joint Research and Development Center, Nankai University, Tianjin 300350, PR China; Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300350, PR China.
| |
Collapse
|
3
|
Potential Role of CXCL13/CXCR5 Signaling in Immune Checkpoint Inhibitor Treatment in Cancer. Cancers (Basel) 2022; 14:cancers14020294. [PMID: 35053457 PMCID: PMC8774093 DOI: 10.3390/cancers14020294] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Immunotherapy is currently the backbone of new drug treatments for many cancer patients. CXC chemokine ligand 13 (CXCL13) is an important factor involved in recruiting immune cells that express CXC chemokine receptor type 5 (CXCR5) in the tumor microenvironment and serves as a key molecular determinant of tertiary lymphoid structure (TLS) formation. An increasing number of studies have identified the influence of CXCL13 on prognosis in patients with cancer, regardless of the use of immunotherapy treatment. However, no comprehensive reviews of the role of CXCL13 in cancer immunotherapy have been published to date. This review aims to provide an overview of the CXCL13/CXCR5 signaling axis to summarize its mechanisms of action in cancer cells and lymphocytes, in addition to effects on immunity and cancer pathobiology, and its potential as a biomarker for the response to cancer immunotherapy. Abstract Immune checkpoint inhibitors (ICIs), including antibodies that target programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), or cytotoxic T lymphocyte antigen 4 (CTLA4), represent some of the most important breakthroughs in new drug development for oncology therapy from the past decade. CXC chemokine ligand 13 (CXCL13) exclusively binds CXC chemokine receptor type 5 (CXCR5), which plays a critical role in immune cell recruitment and activation and the regulation of the adaptive immune response. CXCL13 is a key molecular determinant of the formation of tertiary lymphoid structures (TLSs), which are organized aggregates of T, B, and dendritic cells that participate in the adaptive antitumor immune response. CXCL13 may also serve as a prognostic and predictive factor, and the role played by CXCL13 in some ICI-responsive tumor types has gained intense interest. This review discusses how CXCL13/CXCR5 signaling modulates cancer and immune cells to promote lymphocyte infiltration, activation by tumor antigens, and differentiation to increase the antitumor immune response. We also summarize recent preclinical and clinical evidence regarding the ICI-therapeutic implications of targeting the CXCL13/CXCR5 axis and discuss the potential role of this signaling pathway in cancer immunotherapy.
Collapse
|