201
|
Zhang J, Wang M, He D, Zhang L, Liu T, Wang K. Synergetic regulation of cancer cells and exhausted T cells to fight cold tumors with a fluorinated EGCG-based nanocomplex. J Nanobiotechnology 2023; 21:420. [PMID: 37957632 PMCID: PMC10644671 DOI: 10.1186/s12951-023-02205-6] [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: 06/15/2023] [Accepted: 11/07/2023] [Indexed: 11/15/2023] Open
Abstract
Immune therapy that targets PD-L1 (programmed cell death-ligand 1) is attractive to augment immune response by breaking the programmed cell death-1 (PD-1)/PD-L1 axis. However, T cell exhaustion associated with insufficient T cells infiltration may diminish the efficacy of cancer therapy. Here, we report a novel delivery system of FEGCG/FPEI@siTOX composed of fluorinated EGCG (FEGCG) and fluorinated polyethyleneimine (FPEI) for delivery of small interfering RNA anti-TOX (thymus high mobility group box protein, TOX) to treat tumor and metastasis. In this way, the reduction in PD-L1 expression by FEGCG can promote T-cell function, while inhibition of TOX expression with siTOX can alleviate T-cell exhaustion. FPEI are designed to deliver siRNA with high efficiency and low toxicity compared to classical PEI. Integrating FEGCG, FPEI and siTOX into such a novel system resulted in excellent anti-tumor and antimetastatic effects. It is a promising delivery system and potential strategy for the treatment of "cold" tumors.
Collapse
Affiliation(s)
- Jinlin Zhang
- Department of Pharmacy, Affiliated Cancer Hospital of Nantong University, Nantong University, Nantong, 226001, China
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Mingyue Wang
- Department of Pharmacy, Affiliated Cancer Hospital of Nantong University, Nantong University, Nantong, 226001, China
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Doudou He
- Department of Pharmacy, Affiliated Cancer Hospital of Nantong University, Nantong University, Nantong, 226001, China
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Liang Zhang
- Department of Pharmacy, Affiliated Cancer Hospital of Nantong University, Nantong University, Nantong, 226001, China
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, 2145, Australia.
| | - Kaikai Wang
- Department of Pharmacy, Affiliated Cancer Hospital of Nantong University, Nantong University, Nantong, 226001, China.
- School of Pharmacy, Nantong University, Nantong, 226001, China.
| |
Collapse
|
202
|
Kuah CY, Monfries R, Quartagno M, Seckl MJ, Ghorani E. What is the optimal duration, dose and frequency for anti-PD1 therapy of non-small cell lung cancer? Ther Adv Med Oncol 2023; 15:17588359231210271. [PMID: 37954230 PMCID: PMC10638879 DOI: 10.1177/17588359231210271] [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: 03/02/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Over the past decade, immune checkpoint inhibitors (ICIs) have transformed the management of multiple malignancies including lung cancer. However, the optimal use of these agents in terms of duration, dose and administration frequency remains unknown. Focusing on anti-PD1 agents nivolumab and pembrolizumab in the context of non-small cell lung cancer, we argue that several lines of evidence suggest current administration regimens of these drugs may result in overtreatment with potentially important implications for cost, quality of life and toxicity. This review summarizes evidence for the scope to optimize anti-PD1 regimens, the limitations of existing data and potential approaches to solve these problems including with a novel multi-arm clinical trial design implemented in the recently opened REFINE-Lung study.
Collapse
Affiliation(s)
- Chii Yang Kuah
- Department of Medical Oncology, Charing Cross Hospital Campus of Imperial College London, UK
| | - Robert Monfries
- Department of Medical Oncology, Charing Cross Hospital Campus of Imperial College London, UK
| | - Matteo Quartagno
- Institute for Clinical Trials and Methodology, University College London, London, UK
| | - Michael J. Seckl
- Department of Medical Oncology, Charing Cross Hospital Campus of Imperial College London W6 8RF, UK
| | - Ehsan Ghorani
- Department of Medical Oncology, Charing Cross Hospital Campus of Imperial College London W6 8RF, UK
| |
Collapse
|
203
|
Zhou R, Tong F, Zhang Y, Zhang R, Bin Y, Zhang S, Yang N, Dong X. Genomic alterations associated with pseudoprogression and hyperprogressive disease during anti-PD1 treatment for advanced non-small-cell lung cancer. Front Oncol 2023; 13:1231094. [PMID: 38023206 PMCID: PMC10667039 DOI: 10.3389/fonc.2023.1231094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction This study aimed to elucidate the relationship between dynamic genomic mutation alteration and pseudoprogression (PsPD)/hyperprogressive disease (HPD) in immunotherapy-treated advanced non-small-cell lung cancer (NSCLC), to provide clinical evidence for identifying and distinguishing between PsPD and HPD. Method Patients with advanced NSCLC who were treated with anti-PD1 were enrolled. Whole blood was collected at baseline and post image progression. Serum was separated and sequenced using 425-panel next-generation sequencing analysis (NGS). Results NGS revealed that not only single gene mutations were associated with PsPD/HPD before treatment, dynamic monitoring of the whole-blood genome mutation spectrum also varied greatly. Mutational burden, allele frequency%, and relative circulating tumor DNA abundance indicated that the fold change after image progression was much higher in the HPD group. Discussion The gene mutation profiles of PsPD and HPD not only differed before treatment, but higher genome mutation spectrum post image progression indicated true disease progression in patients with HPD. This suggests that dynamic whole-genome mutation profile monitoring as NGS can distinguish PsPD from HPD more effectively than single gene detection, providing a novel method for guiding clinical immune treatment.
Collapse
Affiliation(s)
- Rui Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Tong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongchang Zhang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruigang Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yawen Bin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nong Yang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
204
|
Kaiho T, Suzuki H, Hata A, Matsumoto H, Tanaka K, Sakairi Y, Motohashi S, Yoshino I. Targeting PD-1/PD-L1 inhibits rejection in a heterotopic tracheal allograft model of lung transplantation. Front Pharmacol 2023; 14:1298085. [PMID: 38026994 PMCID: PMC10657857 DOI: 10.3389/fphar.2023.1298085] [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: 09/21/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Immune checkpoint molecules such as programmed death-1 (PD-1) and programmed death ligand-1 (PD-L1) have revolutionized the field of lung cancer treatment. As part of our study, we examined the role of these proteins in acute rejection in a mouse model of heterotopic tracheal transplantation. Recipient mice were untreated (Allo group) or treated with anti-PD-L1 (aPDL1 group) or PD-L1 Fc recombinant protein (PD-L1 Fc group). A further group of C57BL/6 mice received isografts (Iso group). The occlusion rate was significantly higher in the Allo group than in the Iso group (p = 0.0075), and also higher in the aPD-L1 group (p = 0.0066) and lower in the PD-L1 Fc group (p = 0.030) than in the Allo group. PD-L1 Fc recombinant protein treatment significantly decreased interleukin-6 and interferon-γ levels and reduced the CD4+/CD8+ T cell ratio, without increasing PD-1 and T-cell immunoglobulin mucin 3 expression in CD4+ T cells. These data suggest that PD-L1 Fc recombinant protein decreases the levels of inflammatory cytokines and the proportion of CD4+ T cells without exhaustion. The PD-L1-mediated immune checkpoint mechanism was associated with rejection in the murine tracheal transplant model, suggesting a potential novel target for immunotherapy in lung transplantation.
Collapse
Affiliation(s)
- Taisuke Kaiho
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hidemi Suzuki
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Atsushi Hata
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hiroki Matsumoto
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kazuhisa Tanaka
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yuichi Sakairi
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shinichiro Motohashi
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Medical Immunology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Ichiro Yoshino
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
205
|
Niederlova V, Tsyklauri O, Kovar M, Stepanek O. IL-2-driven CD8 + T cell phenotypes: implications for immunotherapy. Trends Immunol 2023; 44:890-901. [PMID: 37827864 PMCID: PMC7615502 DOI: 10.1016/j.it.2023.09.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
The therapeutic potential of interleukin (IL)-2 in cancer treatment has been known for decades, yet its widespread adoption in clinical practice remains limited. Recently, chimeric proteins of an anti-PD-1 antibody and suboptimal IL-2 variants were shown to stimulate potent antitumor and antiviral immunity by inducing unique effector CD8+ T cells in mice. A similar subset of cytotoxic T cells is induced by depletion of regulatory T cells (Tregs), suggesting IL-2 sequestration as a major mechanism through which regulatory T cells suppress activated CD8+ T cells. Here, we present our view of how IL-2-based biologicals can boost the antitumor response at a cellular level, and propose that the role of Tregs following such treatments may have been previously overestimated.
Collapse
Affiliation(s)
- Veronika Niederlova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Oksana Tsyklauri
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Marek Kovar
- Laboratory of Tumor Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Ondrej Stepanek
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
206
|
Wu Q, Pan C, Zhou Y, Wang S, Xie L, Zhou W, Ding L, Chen T, Qian J, Su R, Gao X, Mei Z, Qiao Y, Yin S, Wu Y, Wang J, Zhou L, Zheng S. Targeting neuropilin-1 abolishes anti-PD-1-upregulated regulatory T cells and synergizes with 4-1BB agonist for liver cancer treatment. Hepatology 2023; 78:1402-1417. [PMID: 36811396 DOI: 10.1097/hep.0000000000000320] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/09/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND AIMS Regulatory T cells (Tregs) are an obstacle to PD-1 blockade-mediated antitumor efficacy. However, the behaviors of Tregs response to anti-PD-1 in HCC and the characteristics of Tregs tissue adaptation from peripheral lymphoid tissues to the tumor are still unclear. APPROACH RESULTS Here, we determine that PD-1 monotherapy potentially augments the accumulation of tumor CD4 + Tregs. Mechanistically, anti-PD-1 mediates Tregs proliferation in lymphoid tissues rather than in the tumor. Increased peripheral Tregs burden replenishes intratumoral Tregs, raising the ratio of intratumoral CD4 + Tregs to CD8 + T cells. Subsequently, single-cell transcriptomics revealed that neuropilin-1 (Nrp-1) supports Tregs migration behavior, and the genes of Crem and Tnfrsf9 regulate the behaviors of the terminal suppressive Tregs. Nrp-1 + 4-1BB - Tregs stepwise develop to the Nrp-1 - 4-1BB + Tregs from lymphoid tissues into the tumor. Moreover, Treg-restricted Nrp1 depletion abolishes anti-PD-1-upregulated intratumoral Tregs burden and synergizes with the 4-1BB agonist to enhance the antitumor response. Finally, a combination of the Nrp-1 inhibitor and the 4-1BB agonist in humanized HCC models showed a favorable and safe outcome and evoked the antitumor effect of the PD-1 blockade. CONCLUSION Our findings elucidate the potential mechanism of anti-PD-1-mediated intratumoral Tregs accumulation in HCC and uncover the tissue adaptation characteristics of Tregs and identify the therapeutic potential of targeting Nrp-1 and 4-1BB for reprogramming the HCC microenvironment.
Collapse
Affiliation(s)
- Qinchuan Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Caixu Pan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Yuan Zhou
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuai Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Liver Transplantation, Shulan Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, China
| | - Liting Xie
- Department of Ultrasound, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wuhua Zhou
- Department of Hepatobiliary Pancreatic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Limin Ding
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Tianchi Chen
- Department of vascular surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junjie Qian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Rong Su
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Xingxing Gao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Zhibin Mei
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Yiting Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Shengyong Yin
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Yi Wu
- Lyvgen Biopharma, Shanghai, China
| | | | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of Liver Transplantation, Shulan Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, China
| |
Collapse
|
207
|
Chen M, Bie L, Ying J. Cancer cell-intrinsic PD-1: Its role in malignant progression and immunotherapy. Biomed Pharmacother 2023; 167:115514. [PMID: 37716115 DOI: 10.1016/j.biopha.2023.115514] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023] Open
Abstract
Programmed cell death protein-1 (PD-1), also called CD279, is coded by the PDCD1 gene and is constitutively expressed on the surface of immune cells. As a receptor and immune checkpoint, PD-1 can bind to programmed death ligand-1/programmed death ligand-2 (PD-L1/PD-L2) in tumor cells, leading to tumor immune evasion. Anti-PD-1 and anti-PD-L1 are important components in tumor immune therapy. PD-1 is also expressed as an intrinsic variant (iPD-1) in cancer cells where it plays important roles in malignant progression as proposed by recent studies. However, iPD-1 has received much less attention compared to PD-1 expressed on immune cells although there is an unmet medical need for fully elucidating the mechanisms of actions to achieve the best response in tumor immunotherapy. iPD-1 suppresses tumorigenesis in non-small cell lung cancer (NSCLC) and colon cancer, whereas it promotes tumorigenesis in melanoma, hepatocellular carcinoma (HCC), pancreatic ductal adenocarcinoma (PDAC), thyroid cancer (TC), glioblastoma (GBM), and triple-negative breast cancer (TNBC). In this review, we focus on the role of iPD-1 in tumorigenesis and development and its molecular mechanisms. We also deeply discuss nivolumab-based combined therapy in common tumor therapy. iPD-1 may explain the different therapeutic effects of anti-PD-1 treatment and provide critical information for use in combined anti-tumor approaches.
Collapse
Affiliation(s)
- Muhua Chen
- Department of Hepato-Pancreato-Biliary & Gastric Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Lei Bie
- Department of Thoracic Surgery, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jieer Ying
- Department of Hepato-Pancreato-Biliary & Gastric Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
208
|
Todd KL, Lai J, Sek K, Huang YK, Newman DM, Derrick EB, Koay HF, Nguyen D, Hoang TX, Petley EV, Chan CW, Munoz I, House IG, Lee JN, Kim JS, Li J, Tong J, N de Menezes M, Scheffler CM, Yap KM, Chen AXY, Dunbar PA, Haugen B, Parish IA, Johnstone RW, Darcy PK, Beavis PA. A 2AR eGFP reporter mouse enables elucidation of A 2AR expression dynamics during anti-tumor immune responses. Nat Commun 2023; 14:6990. [PMID: 37914685 PMCID: PMC10620403 DOI: 10.1038/s41467-023-42734-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023] Open
Abstract
There is significant clinical interest in targeting adenosine-mediated immunosuppression, with several small molecule inhibitors having been developed for targeting the A2AR receptor. Understanding of the mechanism by which A2AR is regulated has been hindered by difficulty in identifying the cell types that express A2AR due to a lack of robust antibodies for these receptors. To overcome this limitation, here an A2AR eGFP reporter mouse is developed, enabling the expression of A2AR during ongoing anti-tumor immune responses to be assessed. This reveals that A2AR is highly expressed on all tumor-infiltrating lymphocyte subsets including Natural Killer (NK) cells, NKT cells, γδ T cells, conventional CD4+ and CD8+ T lymphocytes and on a MHCIIhiCD86hi subset of type 2 conventional dendritic cells. In response to PD-L1 blockade, the emergence of PD-1+A2AR- cells correlates with successful therapeutic responses, whilst IL-18 is identified as a cytokine that potently upregulates A2AR and synergizes with A2AR deficiency to improve anti-tumor immunity. These studies provide insight into the biology of A2AR in the context of anti-tumor immunity and reveals potential combination immunotherapy approaches.
Collapse
Affiliation(s)
- Kirsten L Todd
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia.
| | - Junyun Lai
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Kevin Sek
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Yu-Kuan Huang
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Dane M Newman
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
- Translational Hematology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Emily B Derrick
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Hui-Fern Koay
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Dat Nguyen
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Thang X Hoang
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Emma V Petley
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Cheok Weng Chan
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Isabelle Munoz
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Imran G House
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Joel N Lee
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Joelle S Kim
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Jasmine Li
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Junming Tong
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Maria N de Menezes
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Christina M Scheffler
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Kah Min Yap
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Amanda X Y Chen
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Phoebe A Dunbar
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Brandon Haugen
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Ian A Parish
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Ricky W Johnstone
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
- Translational Hematology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
- Department of Immunology, Monash University, Clayton, Australia
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia.
| |
Collapse
|
209
|
Park J, Kim JC, Lee M, Lee J, Kim YN, Lee YJ, Kim S, Kim SW, Park SH, Lee JY. Frequency of peripheral PD-1 +regulatory T cells is associated with treatment responses to PARP inhibitor maintenance in patients with epithelial ovarian cancer. Br J Cancer 2023; 129:1841-1851. [PMID: 37821637 PMCID: PMC10667217 DOI: 10.1038/s41416-023-02455-z] [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: 01/16/2023] [Revised: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Poly (adenosine diphosphate [ADP]-ribose) polymerase inhibitors (PARPis) are becoming the standard of care for epithelial ovarian cancer (EOC). Recently, clinical trials of triple maintenance therapy (PARPi+anti-angiogenic agent+anti-PD-1/L1) are actively ongoing. Here, we investigated the immunological effects of PARPi or triple maintenance therapy on T cells and their impact on clinical responses. METHODS We collected serial blood from EOC patients receiving PARPi therapy (cohort 1: PARPi, n = 49; cohort 2: olaparib+bevacizumab+pembrolizumab, n = 31). Peripheral T cells were analyzed using flow cytometry and compared according to the PARPi response. Progression-free survival (PFS) was assessed according to prognostic biomarkers identified in a comparative analysis. RESULTS Regulatory T cells (Tregs) were suppressed by PARPi therapy, whereas PD-1 was not significantly changed. Short PFS group exhibited a higher percentage of baseline PD-1+Tregs than long PFS group, and the patients with high percentage of PD-1+Tregs before treatment showed poor PFS in cohort 1. However, the expression of PD-1 on Tregs significantly decreased after receiving triple maintenance therapy, and the reduction in PD-1+Tregs was associated with superior PFS in cohort 2 (P = 0.0078). CONCLUSION PARPi suppresses Tregs, but does not affect PD-1 expression. Adding anti-PD-1 to PARPi decreases PD-1+Tregs, which have negative prognostic value for PARPi monotherapy.
Collapse
Affiliation(s)
- Junsik Park
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Chul Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Miran Lee
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - JooHyang Lee
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoo-Na Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Jae Lee
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sunghoon Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Wun Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Su-Hyung Park
- Laboratory of Translational Immunology and Vaccinology, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
210
|
Proschinger S, Schenk A, Weßels I, Donath L, Rappelt L, Metcalfe AJ, Zimmer P. Intensity- and time-matched acute interval and continuous endurance exercise similarly induce an anti-inflammatory environment in recreationally active runners: focus on PD-1 expression in T regs and the IL-6/IL-10 axis. Eur J Appl Physiol 2023; 123:2575-2584. [PMID: 37336816 PMCID: PMC10615943 DOI: 10.1007/s00421-023-05251-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 06/02/2023] [Indexed: 06/21/2023]
Abstract
PURPOSE Acute exercise elicits a transient anti-inflammatory state during the early recovery period. Since recent studies reported on regimen-specific effects on immune-related humoral factors and cellular subsets, this study compared the effects of intensity- and time-matched acute interval and continuous exercise on peripheral anti-inflammatory cellular and humoral immune parameters with a particular focus on the PD-1 expression in CD4+ regulatory T cells (Tregs). METHODS Twenty-four recreationally active runners (age: 29.7 ± 4.3 years, BMI: 22.2 ± 2.4, VO2peak: 56.6 ± 6.4 ml × kg-1 × min-1) participated in this crossover RCT. Each subject conducted a moderate continuous (MCE) and a high-intensity interval exercise (HIIE) session in a counterbalanced design. Blood was drawn before, immediately after, and 1 h after exercise. Treg subsets and levels of PD-1 and Foxp3 were assessed by flow cytometry. Serum levels of IL-10 and IL-6 were quantified by ELISA. RESULTS PD-1 levels on Tregs increased within the recovery period after HIIE (p < .001) and MCE (p < 0.001). Total counts of Tregs (HIIE: p = 0.044; MCE: p = .021), naïve Tregs (HIIE: p < 0.001; MCE: p < 0.001), and PD-1+ effector Tregs (eTregs) (HIIE: p = .002) decreased 1 h after exercise. IL-10 increased 1 h after HIIE (p < 0.001) and MCE (p = 0.018), while IL-6 increased immediately after both HIIE (p = 0.031) and MCE (p = 0.021). Correlations between changes in IL-6 and IL-10 (p = 0.017, r = 0.379) and baseline VO2peak and Treg frequency (p = 0.002, r = 0.660) were identified. CONCLUSION This is the first study that investigates PD-1 expression in circulating Tregs after acute exercise, revealing an increase in PD-1 levels on eTregs during the early recovery period after intensity- and time-matched HIIE and MCE. Future studies are needed to investigate the PD-1 signalosome in eTregs, together with the expression of key effector molecules (i.e., IL-10, TGF-β, IL-35, CTLA-4) to elucidate PD-1-dependent changes in cellular function. Based on changes in serum cytokines, this study further reveals a regimen-independent establishment of an anti-inflammatory milieu and underpins the role of the IL-6/IL-10 axis.
Collapse
Affiliation(s)
- Sebastian Proschinger
- Division of Performance and Health (Sports Medicine), TU Dortmund University, Institute for Sport and Sport Science, 44227, Dortmund, Germany
| | - Alexander Schenk
- Division of Performance and Health (Sports Medicine), TU Dortmund University, Institute for Sport and Sport Science, 44227, Dortmund, Germany
| | - Inga Weßels
- Faculty of Medicine, RWTH Aachen University, Institute of Immunology, 52074, Aachen, Germany
| | - Lars Donath
- Department of Intervention Research in Exercise Training, German Sport University Cologne, Cologne, Germany
| | - Ludwig Rappelt
- Department of Intervention Research in Exercise Training, German Sport University Cologne, Cologne, Germany
| | - Alan J Metcalfe
- Department for Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Philipp Zimmer
- Division of Performance and Health (Sports Medicine), TU Dortmund University, Institute for Sport and Sport Science, 44227, Dortmund, Germany.
| |
Collapse
|
211
|
Kang JH, Zappasodi R. Modulating Treg stability to improve cancer immunotherapy. Trends Cancer 2023; 9:911-927. [PMID: 37598003 DOI: 10.1016/j.trecan.2023.07.015] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/21/2023]
Abstract
Immunosuppressive regulatory T cells (Tregs) provide a main mechanism of tumor immune evasion. Targeting Tregs, especially in the tumor microenvironment (TME), continues to be investigated to improve cancer immunotherapy. Recent studies have unveiled intratumoral Treg heterogeneity and plasticity, furthering the complexity of the role of Tregs in tumor immunity and immunotherapy response. The phenotypic and functional diversity of intratumoral Tregs can impact their response to therapy and may offer new targets to modulate specific Treg subsets. In this review we provide a unifying framework of critical factors contributing to Treg heterogeneity and plasticity in the TME, and we discuss how this information can guide the development of more specific Treg-targeting therapies for cancer immunotherapy.
Collapse
Affiliation(s)
- Jee Hye Kang
- Weill Cornell Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY, USA
| | - Roberta Zappasodi
- Weill Cornell Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY, USA.
| |
Collapse
|
212
|
Xu X, Chen J, Li W, Feng C, Liu Q, Gao W, He M. Immunology and immunotherapy in gastric cancer. Clin Exp Med 2023; 23:3189-3204. [PMID: 37322134 DOI: 10.1007/s10238-023-01104-2] [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: 01/16/2023] [Accepted: 05/24/2023] [Indexed: 06/17/2023]
Abstract
Gastric cancer is the fifth leading cause of cancer-related deaths worldwide. As the diagnosis of early gastric cancer is difficult, most patients are at a late stage of cancer progression when diagnosed. The current therapeutic approaches based on surgical or endoscopic resection and chemotherapy indeed improve patients' outcomes. Immunotherapy based on immune checkpoint inhibitors has opened a new era for cancer treatment, and the immune system of the host is reshaped to combat tumor cells and the strategy differs according to the patient's immune system. Thus, an in-depth understanding of the roles of various immune cells in the progression of gastric cancer is beneficial to application for immunotherapy and the discovery of new therapeutic targets. This review describes the functions of different immune cells in gastric cancer development, mainly focusing on T cells, B cells, macrophages, natural killer cells, dendritic cells, neutrophils as well as chemokines or cytokines secreted by tumor cells. And this review also discusses the latest advances in immune-related therapeutic approaches such as immune checkpoint inhibitors, CAR-T or vaccine, to reveal potential and promising strategies for gastric cancer treatment.
Collapse
Affiliation(s)
- Xiaqing Xu
- Department of Pharmacy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, People's Republic of China
| | - Jiaxing Chen
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450002, Henan, People's Republic of China
| | - Wenxing Li
- Department of Pharmacy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, People's Republic of China
| | - Chenlu Feng
- Department of Cancer Center, Nanyang First People's Hospital, Nanyang, 473000, Henan, People's Republic of China
| | - Qian Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450002, Henan, People's Republic of China
| | - Wenfang Gao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450002, Henan, People's Republic of China
| | - Meng He
- Department of Pharmacy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, People's Republic of China.
| |
Collapse
|
213
|
Zhang P, Wang Y, Miao Q, Chen Y. The therapeutic potential of PD-1/PD-L1 pathway on immune-related diseases: Based on the innate and adaptive immune components. Biomed Pharmacother 2023; 167:115569. [PMID: 37769390 DOI: 10.1016/j.biopha.2023.115569] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/30/2023] Open
Abstract
Currently, immunotherapy targeting programmed cell death 1 (PD-1) or programmed death ligand 1 (PD-L1) has revolutionized the treatment strategy of human cancer patients. Meanwhile, PD-1/PD-L1 pathway has also been implicated in the pathogenesis of many immune-related diseases, such as autoimmune diseases, chronic infection diseases and adverse pregnancy outcomes, by regulating components of the innate and adaptive immune systems. Given the power of the new therapy, a better understanding of the regulatory effects of PD-1/PD-L1 pathway on innate and adaptive immune responses in immune-related diseases will facilitate the discovery of novel biomarkers and therapeutic drug targets. Targeting this pathway may successfully halt or potentially even reverse these pathological processes. In this review, we discuss recent major advances in PD-1/PD-L1 axis regulating innate and adaptive immune components in immune-related diseases. We reveal that the impact of PD-1/PD-L1 axis on the immune system is complex and manifold and multi-strategies on the targeted PD-1/PD-L1 axis are taken in the treatment of immune-related diseases. Consequently, targeting PD-1/PD-L1 pathway, alone or in combination with other treatments, may represent a novel strategy for future therapeutic intervention on immune-related diseases.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Yuting Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Qianru Miao
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Ying Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
214
|
Parks OB, Antos D, Eddens T, Walters S, Johnson M, Oury TD, Gottschalk RA, Erickson JJ, Williams JV. PD-1 Impairs CD8+ T Cell Granzyme B Production in Aged Mice during Acute Viral Respiratory Infection. Immunohorizons 2023; 7:771-787. [PMID: 38015461 PMCID: PMC10696419 DOI: 10.4049/immunohorizons.2300094] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 11/29/2023] Open
Abstract
CD8+ T cell dysfunction contributes to severe respiratory viral infection outcomes in older adults. CD8+ T cells are the primary cell type responsible for viral clearance. With increasing age, CD8+ T cell function declines in conjunction with an accumulation of cytotoxic tissue-resident memory (TRM) CD8+ T cells. We sought to elucidate the role of PD-1 signaling on aged CD8+ T cell function and accumulation of CD8+ TRM cells during acute viral respiratory tract infection, given the importance of PD-1 regulating CD8+ T cells during acute and chronic infections. PD-1 blockade or genetic ablation in aged mice yielded improved CD8+ T cell granzyme B production comparable to that in young mice during human metapneumovirus and influenza viral infections. Syngeneic transplant and adoptive transfer strategies revealed that improved granzyme B production in aged Pdcd1-/- CD8+ T cells was primarily cell intrinsic because aged wild-type CD8+ T cells did not have increased granzyme B production when transplanted into a young host. PD-1 signaling promoted accumulation of cytotoxic CD8+ TRM cells in aged mice. PD-1 blockade of aged mice during rechallenge infection resulted in improved clinical outcomes that paralleled reduced accumulation of CD8+ TRM cells. These findings suggest that PD-1 signaling impaired CD8+ T cell granzyme B production and contributed to CD8+ TRM cell accumulation in the aged lung. These findings have implications for future research investigating PD-1 checkpoint inhibitors as a potential therapeutic option for elderly patients with severe respiratory viral infections.
Collapse
Affiliation(s)
- Olivia B. Parks
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Danielle Antos
- Division of Pulmonology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Taylor Eddens
- Division of Allergy/Immunology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Sara Walters
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Monika Johnson
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Tim D. Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Rachel A. Gottschalk
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - John J. Erickson
- Division of Neonatology and Pulmonary Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH
| | - John V. Williams
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Institute for Infection, Inflammation, and Immunity in Children (i4Kids), Pittsburgh, PA
| |
Collapse
|
215
|
Chen W, Zou F, Song T, Xia Y, Xing J, Rao T, Zhou X, Ning J, Zhao S, Yu W, Cheng F. Comprehensive analysis reveals XCL2 as a cancer prognosis and immune infiltration-related biomarker. Aging (Albany NY) 2023; 15:11891-11917. [PMID: 37905956 PMCID: PMC10683633 DOI: 10.18632/aging.205156] [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: 05/15/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND X-C Motif Chemokine Ligand 2 (XCL2) is a 114 amino acid, structurally conserved chemokine involved in activating cytotoxic T cells. However, the pathophysiological mechanisms of XCL2 protein in various disease conditions, particularly cancer, remain poorly understood. METHODS Bioinformatics was used to detect the expression of XCL2, the relationship between survival time and XCL2 in BLCA patients, the mutational status of XCL2, the role of XCL2 in the tumor immune microenvironment, and the sensitivity of XCL2-targeted drugs in 33 cancers. In vitro experiments were conducted to investigate the chemotactic effects of XCL2 expression on M1-type macrophages in human specimens and in isolated cancer cells. RESULTS XCL2 expression was downregulated in tumor tissues and closely associated with the prognosis of human cancers. Furthermore, XCL2 affects DNA methylation, tumor mutation burden (TMB), microsatellite instability (MSI), and mismatch repair (MMR) in human cancers. The expression level of XCL2 significantly correlated with infiltrated immune cells, immunological pathways, and other immune markers. More importantly, we found that XCL2 was positively associated with T lymphocytes and macrophages in the transcriptome and single-cell sequencing data. Using multiple immunofluorescence staining, we found that the expression level of XCL2 was upregulated in many cells in pan-cancer samples, and the number of M1 macrophage marker CD68 and INOS-positive cells increased. 786O, U251, and MDA-MB-231 cells could recruit more M1 macrophages in vitro after overexpressing XCL2. CONCLUSIONS Our results reveal that XCL2 could act as a vital chemokine in pan-cancer and provide new targets and concepts for cancer treatment.
Collapse
Affiliation(s)
- Wu Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fan Zou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tianbao Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuqi Xia
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ji Xing
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ting Rao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jinzhuo Ning
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Sheng Zhao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
216
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593'"] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
217
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593'||'] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
218
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593%' and 2*3*8=6*8 and 'eho8'!='eho8%] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
219
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593' and 2*3*8=6*8 and 'x7c8'='x7c8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
220
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593����%2527%2522\'\"] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
221
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593" and 2*3*8=6*8 and "d5lf"="d5lf] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
222
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593j4dtuwxg] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
223
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 PMCID: PMC10642438 DOI: 10.3748/wjg.v29.i40.5593] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| |
Collapse
|
224
|
Mahdi HS, Woodall-Jappe M, Singh P, Czuczman MS. Targeting regulatory T cells by E7777 enhances CD8 T-cell-mediated anti-tumor activity and extends survival benefit of anti-PD-1 in solid tumor models. Front Immunol 2023; 14:1268979. [PMID: 38022532 PMCID: PMC10646188 DOI: 10.3389/fimmu.2023.1268979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Regulatory T cell (Treg)-targeting cancer immunotherapy aims to transiently deplete Treg cells in the tumor microenvironment, without affecting effector T cells (Teff), thus both enhancing anti-tumor activity and avoiding autoimmunity. This study evaluated whether adding E7777 (a new formulation of denileukin diftitox [DD]) improved the efficacy of anti-PD-1 antibody therapy. DD is a recombinant protein containing the hydrophobic and catalytic portions of diphtheria toxin fused to full-length human IL-2. E7777 has the same amino acid sequence and brief circulatory half-life as DD, but with greater purity and potency. Methods Subcutaneous syngeneic murine solid tumor models (colon cancer CT-26 and liver cancer H22) were used to evaluate safety, efficacy, and overall survival with E7777 and anti-PD-1 antibodies, each administered as monotherapy or in concurrent or sequential combination. In Experiment 1, treatments were compared to assess anti-tumor activity at various time points, with tumors excised and dissociated and tumor leukocytes characterized. In Experiment 2, tumor growth, response, and overall survival were characterized for 100 days following a 3-week treatment. Results E7777 administered in combination with anti-PD-1 led to significantly increased anti-tumor activity and durable, extended overall survival compared to either treatment alone. In both tumor models, the Treg cell infiltration induced by anti-PD-1 treatment was counterbalanced by co-treatment with E7777, suggesting potential synergistic activity. Combination therapy showed the most favorable results. Treatment with E7777 was safe and well-tolerated. Discussion Combined E7777 and anti-PD-1 therapy was well tolerated and more effective than monotherapy with either drug.
Collapse
Affiliation(s)
- Haider S. Mahdi
- Department of Obstetrics, Gynecology & Reproductive Sciences, Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Preeti Singh
- Clinical Development and Medical Affairs, Citius Pharmaceuticals, Inc., Cranford, NJ, United States
| | - Myron S. Czuczman
- Clinical Development and Medical Affairs, Citius Pharmaceuticals, Inc., Cranford, NJ, United States
| |
Collapse
|
225
|
Roy D, Gilmour C, Patnaik S, Wang LL. Combinatorial blockade for cancer immunotherapy: targeting emerging immune checkpoint receptors. Front Immunol 2023; 14:1264327. [PMID: 37928556 PMCID: PMC10620683 DOI: 10.3389/fimmu.2023.1264327] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023] Open
Abstract
The differentiation, survival, and effector function of tumor-specific CD8+ cytotoxic T cells lie at the center of antitumor immunity. Due to the lack of proper costimulation and the abundant immunosuppressive mechanisms, tumor-specific T cells show a lack of persistence and exhausted and dysfunctional phenotypes. Multiple coinhibitory receptors, such as PD-1, CTLA-4, VISTA, TIGIT, TIM-3, and LAG-3, contribute to dysfunctional CTLs and failed antitumor immunity. These coinhibitory receptors are collectively called immune checkpoint receptors (ICRs). Immune checkpoint inhibitors (ICIs) targeting these ICRs have become the cornerstone for cancer immunotherapy as they have established new clinical paradigms for an expanding range of previously untreatable cancers. Given the nonredundant yet convergent molecular pathways mediated by various ICRs, combinatorial immunotherapies are being tested to bring synergistic benefits to patients. In this review, we summarize the mechanisms of several emerging ICRs, including VISTA, TIGIT, TIM-3, and LAG-3, and the preclinical and clinical data supporting combinatorial strategies to improve existing ICI therapies.
Collapse
Affiliation(s)
- Dia Roy
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Cassandra Gilmour
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, United States
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Sachin Patnaik
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Li Lily Wang
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, United States
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
226
|
Flati I, Di Vito Nolfi M, Dall’Aglio F, Vecchiotti D, Verzella D, Alesse E, Capece D, Zazzeroni F. Molecular Mechanisms Underpinning Immunometabolic Reprogramming: How the Wind Changes during Cancer Progression. Genes (Basel) 2023; 14:1953. [PMID: 37895302 PMCID: PMC10606647 DOI: 10.3390/genes14101953] [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: 09/25/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Metabolism and the immunological state are intimately intertwined, as defense responses are bioenergetically expensive. Metabolic homeostasis is a key requirement for the proper function of immune cell subsets, and the perturbation of the immune-metabolic balance is a recurrent event in many human diseases, including cancer, due to nutrient fluctuation, hypoxia and additional metabolic changes occurring in the tumor microenvironment (TME). Although much remains to be understood in the field of immunometabolism, here, we report the current knowledge on both physiological and cancer-associated metabolic profiles of immune cells, and the main molecular circuits involved in their regulation, highlighting similarities and differences, and emphasizing immune metabolic liabilities that could be exploited in cancer therapy to overcome immune resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, Via Vetoio, Coppito 2, 67100 L’Aquila, Italy; (I.F.); (M.D.V.N.); (F.D.); (D.V.); (D.V.); (E.A.); (F.Z.)
| | | |
Collapse
|
227
|
Liu S, Wang W, Hu S, Jia B, Tuo B, Sun H, Wang Q, Liu Y, Sun Z. Radiotherapy remodels the tumor microenvironment for enhancing immunotherapeutic sensitivity. Cell Death Dis 2023; 14:679. [PMID: 37833255 PMCID: PMC10575861 DOI: 10.1038/s41419-023-06211-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/22/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023]
Abstract
Cancer immunotherapy has transformed traditional treatments, with immune checkpoint blockade being particularly prominent. However, immunotherapy has minimal benefit for patients in most types of cancer and is largely ineffective in some cancers (such as pancreatic cancer and glioma). A synergistic anti-tumor response may be produced through the combined application with traditional tumor treatment methods. Radiotherapy (RT) not only kills tumor cells but also triggers the pro-inflammatory molecules' release and immune cell infiltration, which remodel the tumor microenvironment (TME). Therefore, the combination of RT and immunotherapy is expected to achieve improved efficacy. In this review, we summarize the effects of RT on cellular components of the TME, including T cell receptor repertoires, different T cell subsets, metabolism, tumor-associated macrophages and other myeloid cells (dendritic cells, myeloid-derived suppressor cells, neutrophils and eosinophils). Meanwhile, non-cellular components such as lactate and extracellular vesicles are also elaborated. In addition, we discuss the impact of different RT modalities on tumor immunity and issues related to the clinical practice of combination therapy.
Collapse
Affiliation(s)
- Senbo Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Bin Jia
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Baojing Tuo
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Haifeng Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 450001, Zhengzhou, China.
| | - Yang Liu
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 450001, Zhengzhou, China.
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
228
|
Kubiak AM, Claessen L, Zhang Y, Khazaie K, Bailey TS. Refined control of CRISPR-Cas9 gene editing in Clostridium sporogenes: the creation of recombinant strains for therapeutic applications. Front Immunol 2023; 14:1241632. [PMID: 37869009 PMCID: PMC10585264 DOI: 10.3389/fimmu.2023.1241632] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/01/2023] [Indexed: 10/24/2023] Open
Abstract
Despite considerable clinical success, the potential of cancer immunotherapy is restricted by a lack of tumour-targeting strategies. Treatment requires systemic delivery of cytokines or antibodies at high levels to achieve clinically effective doses at malignant sites. This is exacerbated by poor penetration of tumour tissue by therapeutic antibodies. High-grade immune-related adverse events (irAEs) occur in a significant number of patients (5-15%, cancer- and therapeutic-dependent) that can lead to lifelong issues and can exclude from treatment patients with pre-existing autoimmune diseases. Tumour-homing bacteria, genetically engineered to produce therapeutics, is one of the approaches that seeks to mitigate these drawbacks. The ability of Clostridium sporogenes to form spores that are unable to germinate in the presence of oxygen (typical of healthy tissue) offers a unique advantage over other vectors. However, the limited utility of existing gene editing tools hinders the development of therapeutic strains. To overcome the limitations of previous systems, expression of the Cas9 protein and the gRNA was controlled using tetracycline inducible promoters. Furthermore, the components of the system were divided across two plasmids, improving the efficiency of cloning and conjugation. Genome integrated therapeutic genes were assayed biochemically and in cell-based functional assays. The potency of these strains was further improved through rationally-conceived gene knock-outs. The new system was validated by demonstrating the efficient addition and deletion of large sequences from the genome. This included the creation of recombinant strains expressing two pro-inflammatory cytokines, interleukin-2 (IL-2) and granulocyte macrophage-colony stimulating factor (GM-CSF), and a pro-drug converting enzyme (PCE). A comparative, temporal in vitro analysis of the integrant strains and their plasmid-based equivalents revealed a substantial reduction of cytokine activity in chromosome-based constructs. To compensate for this loss, a 7.6 kb operon of proteolytic genes was deleted from the genome. The resultant knock-out strains showed an 8- to 10-fold increase in cytokine activity compared to parental strains.
Collapse
Affiliation(s)
- Aleksandra M. Kubiak
- Exomnis Biotech BV, Maastricht, Netherlands
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| | - Luuk Claessen
- Exomnis Biotech BV, Maastricht, Netherlands
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| | - Yanchao Zhang
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| | - Khashayarsha Khazaie
- Department of Immunology and Cancer Biology, Mayo Clinic, Phoenix, AZ, United States
| | - Tom S. Bailey
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
229
|
Chamoto K, Yaguchi T, Tajima M, Honjo T. Insights from a 30-year journey: function, regulation and therapeutic modulation of PD1. Nat Rev Immunol 2023; 23:682-695. [PMID: 37185300 DOI: 10.1038/s41577-023-00867-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 05/17/2023]
Abstract
PD1 was originally discovered in 1992 as a molecule associated with activation-induced cell death in T cells. Over the past 30 years, it was found that PD1 has a critical role in avoiding overactivation-induced cell death and autoimmunity, whereas its inhibition unleashes anticancer immunity. Here, we outline the journey from the discovery of PD1 to its role as a breakthrough target in cancer immunotherapy. We describe its regulation and function and examine how a mechanistic understanding of PD1 signalling suggests a central function in setting the T cell activation threshold, thereby controlling T cell proliferation, differentiation, exhaustion and metabolic status. This threshold theory, in combination with new insights into T cell metabolism and a better understanding of immune cell modulation by the microbiota, can provide guidance for the development of efficient combination therapies. Moreover, we discuss the mechanisms underlying immune-related adverse events after PD1-targeted therapy and their possible treatment.
Collapse
Affiliation(s)
- Kenji Chamoto
- Division of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomonori Yaguchi
- Division of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masaki Tajima
- Division of Integrated High-Order Regulatory Systems, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tasuku Honjo
- Division of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
230
|
Lin X, Zong C, Zhang Z, Fang W, Xu P. Progresses in biomarkers for cancer immunotherapy. MedComm (Beijing) 2023; 4:e387. [PMID: 37799808 PMCID: PMC10547938 DOI: 10.1002/mco2.387] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 10/07/2023] Open
Abstract
Currently, checkpoint inhibitor-based immunotherapy has emerged as prevailing treatment modality for diverse cancers. However, immunotherapy as a first-line therapy has not consistently yielded durable responses. Moreover, the risk of immune-related adverse events increases with combination regimens. Thus, the development of predictive biomarkers is needed to optimize individuals benefit, minimize risk of toxicities, and guide combination approaches. The greatest focus has been on tumor programmed cell death-ligand 1 (PD-L1), microsatellite instability (MSI), and tumor mutational burden (TMB). However, there remains a subject of debate due to thresholds variability and significant heterogeneity. Major unmet challenges in immunotherapy are the discovery and validation of predictive biomarkers. Here, we show the status of tumor PD-L1, MSI, TMB, and emerging data on novel biomarker strategies with oncogenic signaling and epigenetic regulation. Considering the exploration of peripheral and intestinal immunity has served as noninvasive alternative in predicting immunotherapy, this review also summarizes current data in systemic immunity, encompassing solute PD-L1 and TMB, circulating tumor DNA and infiltrating lymphocytes, routine emerging inflammatory markers and cytokines, as well as gut microbiota. This review provides up-to-date information on the evolving field of currently available biomarkers in predicting immunotherapy. Future exploration of novel biomarkers is warranted.
Collapse
Affiliation(s)
- Xuwen Lin
- Department of Pulmonary and Critical Care MedicinePeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
- Department of Internal MedicineShantou University Medical CollegeShantouGuangdong ProvinceChina
| | - Chenyu Zong
- Department of Pulmonary and Critical Care MedicinePeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
- Department of Internal MedicineZunyi Medical UniversityZunyiGuizhou ProvinceChina
| | - Zhihan Zhang
- Department of Pulmonary and Critical Care MedicinePeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
| | - Weiyi Fang
- Cancer Research InstituteSchool of Basic Medical ScienceSouthern Medical UniversityGuangzhouGuangdong ProvinceChina
- Cancer CenterIntegrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouGuangdong ProvinceChina
| | - Ping Xu
- Department of Pulmonary and Critical Care MedicinePeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
- Department of Internal MedicineZunyi Medical UniversityZunyiGuizhou ProvinceChina
| |
Collapse
|
231
|
Massa C, Seliger B. Combination of multiple omics techniques for a personalized therapy or treatment selection. Front Immunol 2023; 14:1258013. [PMID: 37828984 PMCID: PMC10565668 DOI: 10.3389/fimmu.2023.1258013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/05/2023] [Indexed: 10/14/2023] Open
Abstract
Despite targeted therapies and immunotherapies have revolutionized the treatment of cancer patients, only a limited number of patients have long-term responses. Moreover, due to differences within cancer patients in the tumor mutational burden, composition of the tumor microenvironment as well as of the peripheral immune system and microbiome, and in the development of immune escape mechanisms, there is no "one fit all" therapy. Thus, the treatment of patients must be personalized based on the specific molecular, immunologic and/or metabolic landscape of their tumor. In order to identify for each patient the best possible therapy, different approaches should be employed and combined. These include (i) the use of predictive biomarkers identified on large cohorts of patients with the same tumor type and (ii) the evaluation of the individual tumor with "omics"-based analyses as well as its ex vivo characterization for susceptibility to different therapies.
Collapse
Affiliation(s)
- Chiara Massa
- Institute for Translational Immunology, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Barbara Seliger
- Institute for Translational Immunology, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| |
Collapse
|
232
|
Cao H, Wu T, Zhou X, Xie S, Sun H, Sun Y, Li Y. Progress of research on PD-1/PD-L1 in leukemia. Front Immunol 2023; 14:1265299. [PMID: 37822924 PMCID: PMC10562551 DOI: 10.3389/fimmu.2023.1265299] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 09/13/2023] [Indexed: 10/13/2023] Open
Abstract
Leukemia cells prevent immune system from clearing tumor cells by inducing the immunosuppression of the bone marrow (BM) microenvironment. In recent years, further understanding of the BM microenvironment and immune landscape of leukemia has resulted in the introduction of several immunotherapies, including checkpoint inhibitors, T-cell engager, antibody drug conjugates, and cellular therapies in clinical trials. Among them, the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) axis is a significant checkpoint for controlling immune responses, the PD-1 receptor on tumor-infiltrating T cells is bound by PD-L1 on leukemia cells. Consequently, the activation of tumor reactive T cells is inhibited and their apoptosis is promoted, preventing the rejection of the tumor by immune system and thus resulting in the occurrence of immune tolerance. The PD-1/PD-L1 axis serves as a significant mechanism by which tumor cells evade immune surveillance, and PD-1/PD-L1 checkpoint inhibitors have been approved for the treatment of lymphomas and varieties of solid tumors. However, the development of drugs targeting PD-1/PD-L1 in leukemia remains in the clinical-trial stage. In this review, we tally up the basic research and clinical trials on PD-1/PD-L1 inhibitors in leukemia, as well as discuss the relevant toxicity and impacts of PD-1/PD-L1 on other immunotherapies such as hematopoietic stem cell transplantation, bi-specific T-cell engager, chimeric antigen receptor T-cell immunotherapy.
Collapse
Affiliation(s)
- Huizhen Cao
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Tianyu Wu
- Department of Gastrointestinal Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Xue Zhou
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Shuyang Xie
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| | - Hongfang Sun
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| | - Yunxiao Sun
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| |
Collapse
|
233
|
Motoo I, Ando T, Hamashima T, Kajiura S, Sakumura M, Ueda Y, Murayama A, Ogawa K, Tsukada K, Ueda A, Suzuki N, Nakada N, Nakashima K, Hosokawa A, Yasuda I. Liver metastasis affects progression pattern during immune checkpoint inhibitors monotherapy in gastric cancer. Front Oncol 2023; 13:1193533. [PMID: 37790758 PMCID: PMC10542891 DOI: 10.3389/fonc.2023.1193533] [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: 03/25/2023] [Accepted: 08/29/2023] [Indexed: 10/05/2023] Open
Abstract
Introduction The efficacy of immune checkpoint inhibitors (ICIs) is heterogeneous at each metastatic site, and tumor progression pattern is associated with survival; however, it remains unclear in gastric cancer (GC). Therefore, we aimed to clarify the progression pattern in response to ICIs in patients with GC, and we analyzed its mechanism focusing on the intratumoral immune cells. Methods Patients who received ICIs were retrospectively classified into non-systemic and systemic progression groups based on their radiological assessments. Moreover, the best percentage change in target lesions from each organ was compared. Results Among 148 patients, the non-systemic progression group showed a significant improvement in overall survival (OS) compared with the systemic progression group (median, 5.6 months vs. 3.3 months; HR, 0.53; 95%CI, 0.32-0.89; p = 0.012). Poor performance status (HR, 1.73, 95%CI, 1.00-2.87) and systemic progression (HR, 3.09, 95%CI, 1.95-4.82) were associated with OS. Of all metastatic sites, the liver showed the poorest percentage change, and liver metastasis (OR, 2.99, 95%CI, 1.04-8.58) was associated with systemic progression. Hence, intratumoral CD8+ T-cell density was lower in patients with liver metastasis than in those without liver metastasis after ICIs, although the density of CD4+ T-cells (Th1, Th17, and Treg) and CD163+ cells (TAM) were not significantly different. Conclusion The new progression pattern was associated with OS in GC. Liver metastasis may be a predictive factor of systemic progression during ICIs by regulating intratumoral CD8+ T-cells.
Collapse
Affiliation(s)
- Iori Motoo
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Takayuki Ando
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | | | - Shinya Kajiura
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Miho Sakumura
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Yuko Ueda
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Aiko Murayama
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Kohei Ogawa
- Department of Gastroenterology, Toyama Prefectural Central Hospital, Toyama, Japan
| | - Kenichiro Tsukada
- Department of Gastroenterology, Kouseiren Takaoka Hospital, Takaoka, Japan
| | - Akira Ueda
- Department of Medical Oncology, Toyama Red Cross Hospital, Toyama, Japan
| | - Nobuhiro Suzuki
- Department of Gastroenterology, Jouetsu Sogo Hospital, Jouetsu, Japan
| | - Naokatsu Nakada
- Department of Gastroenterology, Itoigawa Sogo Hospital, Itoigawa, Japan
| | - Koji Nakashima
- Department of Clinical Oncology, University of Miyazaki Hospital, Miyazaki, Japan
| | - Ayumu Hosokawa
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
- Department of Clinical Oncology, University of Miyazaki Hospital, Miyazaki, Japan
| | - Ichiro Yasuda
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
234
|
Nagira Y, Nagira M, Nagai R, Nogami W, Hirata M, Ueyama A, Yoshida T, Yoshikawa M, Shinonome S, Yoshida H, Haruna M, Miwa H, Chatani N, Ohkura N, Wada H, Tanaka H. S-531011, a Novel Anti-Human CCR8 Antibody, Induces Potent Antitumor Responses through Depletion of Tumor-Infiltrating CCR8-Expressing Regulatory T Cells. Mol Cancer Ther 2023; 22:1063-1072. [PMID: 37420296 PMCID: PMC10477828 DOI: 10.1158/1535-7163.mct-22-0570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 05/17/2023] [Accepted: 06/30/2023] [Indexed: 07/09/2023]
Abstract
Although regulatory T cells (Treg) are inhibitory immune cells that are essential for maintaining immune homeostasis, Tregs that infiltrate tumor tissue promote tumor growth by suppressing antitumor immunity. Selective reduction of tumor-infiltrating Tregs is, therefore, expected to activate antitumor immunity without affecting immune homeostasis. We previously reported that selective Treg depletion targeted by a C-C motif chemokine receptor 8 (CCR8) resulted in induction of strong antitumor immunity without any obvious autoimmunity in mouse models. Thus, herein, we developed a novel humanized anti-CCR8 monoclonal antibody, S-531011, aimed as a cancer immunotherapy strategy for patients with cancer. S-531011 exclusively recognized human CCR8 among all chemokine receptors and showed potent antibody-dependent cell-mediated cytotoxicity activity toward CCR8+ cells and neutralization activity against CCR8-mediated signaling. We observed that S-531011 reduced tumor-infiltrating CCR8+ Tregs and induced potent antitumor activity in a tumor-bearing human-CCR8 knock-in mouse model. Moreover, combination therapy with S-531011 and anti-mouse programmed cell death 1 (PD-1) antibody strongly suppressed tumor growth compared with anti-PD-1 antibody alone with no observable adverse effects. S-531011 also depleted human tumor-infiltrating Tregs, but not Tregs derived from human peripheral blood mononuclear cells. These results suggest that S-531011 is a promising drug for inducing antitumor immunity without severe side effects in the clinical setting.
Collapse
Affiliation(s)
- Yoji Nagira
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Morio Nagira
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Ryohei Nagai
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Wataru Nogami
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Michinari Hirata
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Azumi Ueyama
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tetsuya Yoshida
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
- Department of Basic Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mai Yoshikawa
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Satomi Shinonome
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Hiroshi Yoshida
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Miya Haruna
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroto Miwa
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Natsumi Chatani
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Naganari Ohkura
- Department of Basic Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidekazu Tanaka
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| |
Collapse
|
235
|
Wang YK, Lv XX, Wang ZQ, Zhou YM, Jiang B, Wang SN, Chen XD. The significance of the microlymphangiogenesis, microangiogenesis, and combined detection of programmed cell death-1 protein (PD-1)/ki67 in gastric cancer tissues. J Cancer Res Clin Oncol 2023; 149:9129-9137. [PMID: 37179266 DOI: 10.1007/s00432-023-04709-y] [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: 01/30/2023] [Accepted: 03/17/2023] [Indexed: 05/15/2023]
Abstract
PURPOSE To investigate the relationship between the microlymphangiogenesis, microangiogenesis, and combined detection of the programmed cell death-1 protein (PD-1)/ki67 in patients with gastric cancer as well as the disease prognosis. METHODS Immunohistochemistry was used to detect the microlymphatic density (MLD) and microvessel density (MVD) in the central and peripheral zones in 92 cases of gastric cancer, along with the number of PD-1- and ki67-positive tumor cells. RESULTS The central zone of the gastric cancer tissue contained fewer atretic cord-like lymphatic vessels than the peripheral zone, while the peripheral zone contained an increased number of lymphatic vessels compared with the central zone. In most cases, the lumen was also dilated. Compared with the MLD in the peripheral zone, the MLD in central zone was significantly decreased. Compared with the number of PD-1-positive cells in the peripheral zone, the number of PD-1-positive cells in the central zone was significantly decreased, and compared with the number of ki67-positive cells in the peripheral zone. The differences in the microlymphangiogenesis, microangiogenesis, and the number of PD-1- and ki67-positive cells among the different histological types were not statistically significant. The microlymphangiogenesis, microangiogenesis, and PD-1- and ki67-positive cells were significantly decreased in the gastric cancer tissues from the patients in stages T1 and T2 compared with the gastric cancer tissues from the patients in stages T3 and T4. CONCLUSIONS The detection of the MLD and MVD as well as the positive expression of PD-1 and ki67 in gastric cancer tissue are important reference indicators for judging the prognosis of gastric cancer.
Collapse
Affiliation(s)
- Yang-Kun Wang
- Department of Pathology, Shenzhen Longgang District Fourth People's Hospital, Shenzhen, 518123, China
| | - Xue-Xia Lv
- Department of Pathology, 989th Hospital of the Joint Logistic Support Force of the PLA, Luoyang, 471031, China
| | - Zhi-Qiang Wang
- Department of Pathology, Foresea Life Insurance Guangzhou General Hospital, No. 703 Xincheng Avenue, Zengcheng District, Guangzhou, 511300, China
| | - Yong-Mei Zhou
- Department of Pathology, Foresea Life Insurance Guangzhou General Hospital, No. 703 Xincheng Avenue, Zengcheng District, Guangzhou, 511300, China
| | - Bo Jiang
- Department of Pathology, No. 990 Hospital of the PLA Joint Logistics Support Force, Zhumadian, 463000, China
| | - Su-Nan Wang
- Shenzhen Polytechnic, Xili Lake, Xilihu Town, Nanshan District, Shenzhen, 518055, China.
| | - Xiao-Dong Chen
- Department of Pathology, Foresea Life Insurance Guangzhou General Hospital, No. 703 Xincheng Avenue, Zengcheng District, Guangzhou, 511300, China.
| |
Collapse
|
236
|
You Y, Jin F, Du Y, Zhu L, Liu D, Zhu M, Du Y, Lang J, Li W, Ji JS, Du YZ. A photo-activable nano-agonist for the two-signal model of T cell in vivo activation. J Control Release 2023; 361:681-693. [PMID: 37595667 DOI: 10.1016/j.jconrel.2023.08.033] [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: 04/10/2023] [Revised: 07/24/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
The two-signal model of T cell activation has helped shape our understanding of the adaptive immune response for over four decades. According to the model, activation of T cells requires a stimulus through the T cell receptor/CD3 complex (signal 1) and a costimulatory signal 2. Stimulation of activatory signals via T cell agonists has thus emerged. However, for a robust T cell activation, it necessitates not only the presence of both signal 1 and signal 2, but also a high signaling strength. Herein, we report a photo-activable nano-agonist for the two-signal model of T cell in vivo activation. A UV-crosslinkable polymer is coated onto upconversion nanoparticles with satisfactory NIR-to-UV light conversion efficiency. Then dual signal molecules, i.e., signal 1 and signal 2, are conjugated to the polymer end to yield the photo-activable T cell nano-agonist. In melanoma and breast cancer models, photo-activable nano-agonist could bind onto corresponding activatory receptors on the surface of T cells, but has limited activity without the application of NIR light (absence of photo-crosslinking of receptors and consequently a poor signaling strength). While when the NIR light is switched on locally, T cells in tumor are remarkably activated and kill tumor cells effectively. Moreover, we do not observe any detectable toxicities related to the photo-activable nano-agonist. We believe with two activatory signals being simultaneously strengthened by local photo-switched crosslinking, T cells realize a robust and selective activation in tumor and, consequently contribute to an enhanced and safe tumor immunotherapy.
Collapse
Affiliation(s)
- Yuchan You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, PR China
| | - Feiyang Jin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, PR China
| | - Yan Du
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, PR China
| | - Luwen Zhu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, PR China
| | - Di Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, PR China
| | - Minxia Zhu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, PR China
| | - Yuyin Du
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, PR China
| | - Jialu Lang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, PR China
| | - Weishuo Li
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, PR China.
| | - Jian-Song Ji
- Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Radiology, 289 Kuocang Road, Lishui 323000, PR China.
| | - Yong-Zhong Du
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, PR China; Innovation Center of Transformational Pharmacy, Jinhua Institute of Zhejiang University, Jinhua 321299, PR China.
| |
Collapse
|
237
|
Gadwa J, Amann M, Bickett TE, Knitz MW, Darragh LB, Piper M, Van Court B, Bukkapatnam S, Pham TT, Wang XJ, Saviola AJ, Deak LC, Umaña P, Klein C, D'Alessandro A, Karam SD. Selective targeting of IL2Rβγ combined with radiotherapy triggers CD8- and NK-mediated immunity, abrogating metastasis in HNSCC. Cell Rep Med 2023; 4:101150. [PMID: 37586327 PMCID: PMC10439274 DOI: 10.1016/j.xcrm.2023.101150] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 04/21/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
The implementation of cancer immunotherapies has seen limited clinical success in head and neck squamous cell carcinoma (HNSCC). Interleukin-2 (IL-2), which modulates the survival and functionality of lymphocytes, is an attractive target for new immunotherapies but one that is limited by presence of regulatory T cells (Tregs) expressing the high-affinity IL-2Rα. The bispecific immunocytokine PD1-IL2v preferentially delivers IL-2 signaling through IL-2Rβγ on PD-1-expressing cells. Selectively targeting the intermediate-affinity IL-2Rβγ can be leveraged to induce anti-tumor immune responses in effector T cells and natural killer (NK) cells while limiting the negative regulation of IL-2Rα activation on Tregs. Using radiation therapy (RT) in combination with PD1-IL2v improves local tumor control and survival, and controls metastatic spread in orthotopic HNSCC tumor models. PD1-IL2v drives systemic activation and expansion of circulating and tumor-infiltrating cytotoxic T cells and NK cells while limiting Treg-mediated immunosuppression. These data show that PD1-L2v induces durable systemic tumor control in HNSCC.
Collapse
Affiliation(s)
- Jacob Gadwa
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology & Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Maria Amann
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), 8952 Schlieren, Switzerland
| | - Thomas E Bickett
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael W Knitz
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Laurel B Darragh
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology & Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Miles Piper
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Benjamin Van Court
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sanjana Bukkapatnam
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tiffany T Pham
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Xiao-Jing Wang
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Laura Codarri Deak
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), 8952 Schlieren, Switzerland
| | - Pablo Umaña
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), 8952 Schlieren, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), 8952 Schlieren, Switzerland
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology & Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
238
|
Wei J, Lu X, Liu Q, Fu Y, Liu S, Zhao Y, Zhou J, Chen H, Wang M, Li L, Yang J, Liu F, Zheng L, Yin H, Yang Y, Zhou C, Zeng P, Zhou X, Ding N, Chen S, Zhao X, Yan J, Fan X, Guan W, Liu B. Neoadjuvant sintilimab in combination with concurrent chemoradiotherapy for locally advanced gastric or gastroesophageal junction adenocarcinoma: a single-arm phase 2 trial. Nat Commun 2023; 14:4904. [PMID: 37580320 PMCID: PMC10425436 DOI: 10.1038/s41467-023-40480-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 07/31/2023] [Indexed: 08/16/2023] Open
Abstract
In this multicenter, single-arm phase 2 trial (ChiCTR1900024428), patients with locally advanced gastric/gastroesophageal junction cancers receive one cycle of sintilimab (anti-PD1) and chemotherapy (S-1 and nab-paclitaxel), followed by 5 weeks of concurrent chemoradiotherapy and sintilimab, and another cycle of sintilimab and chemotherapy thereafter. Surgery is preferably scheduled within one to three weeks, and three cycles of adjuvant sintilimab and chemotherapy are administrated. The primary endpoint is the pathological complete response. Our results meet the pre-specified primary endpoint. Thirteen of 34 (38.2%) enrolled patients achieve pathological complete response (95% CI: 22.2-56.4). The secondary objectives include disease-free survival (DFS), major pathological response, R0 resection rate, overall survival (OS), event-free survival (EFS), and safety profile. The median DFS and EFS were 17.0 (95%CI: 11.1-20.9) and 21.1 (95%CI: 14.7-26.1) months, respectively, while the median OS was not reached, and the 1-year OS rate was 92.6% (95%CI: 50.1-99.5%). Seventeen patients (50.0%) have grade ≥3 adverse events during preoperative therapy. In prespecified exploratory biomarker analysis, CD3+ T cells, CD56+ NK cells, and the M1/M1 + M2-like macrophage infiltration at baseline are associated with pathological complete response. Here, we show the promising efficacy and manageable safety profile of sintilimab in combination with concurrent chemoradiotherapy for the perioperative treatment of locally advanced gastric/gastroesophageal junction adenocarcinoma.
Collapse
Affiliation(s)
- Jia Wei
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Xiaofeng Lu
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Qin Liu
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Yao Fu
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Song Liu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yang Zhao
- Department of Biostatistics, Nanjing Medical University, Nanjing, 210029, China
| | - Jiawei Zhou
- Department of Biostatistics, Nanjing Medical University, Nanjing, 210029, China
| | - Hui Chen
- Medical Affairs, 3D Medicines Inc, Shanghai, 201114, China
| | - Meng Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Lin Li
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Ju Yang
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Fangcen Liu
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Liming Zheng
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Haitao Yin
- Department of Radiotherapy, Xuzhou Central Hospital, Xuzhou, 221009, China
| | - Yang Yang
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Chong Zhou
- Department of Radiotherapy, Xuzhou Central Hospital, Xuzhou, 221009, China
| | - Ping Zeng
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xiaoyu Zhou
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Naiqing Ding
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Shiqing Chen
- Medical Affairs, 3D Medicines Inc, Shanghai, 201114, China
| | - Xiaochen Zhao
- Medical Affairs, 3D Medicines Inc, Shanghai, 201114, China
| | - Jing Yan
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Xiangshan Fan
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Wenxian Guan
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Baorui Liu
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
- Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
239
|
Cheng Z, Zeng T, Yang G, Liu D, Zheng Z, Yuan Z. Genomic and Immune Features in an Intrahepatic Cholangiocarcinoma Patient with Microsatellite Instability-High Suffered Rapid Acquired Resistance to PD-1 Inhibitor. Liver Cancer 2023; 12:281-288. [PMID: 37593364 PMCID: PMC10427924 DOI: 10.1159/000530273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 03/14/2023] [Indexed: 08/19/2023] Open
Abstract
Introduction Intrahepatic cholangiocarcinoma (ICC) is a highly aggressive liver malignancy with poor prognosis. Recently, the development of immune checkpoint inhibitors (ICIs), such as programmed cell death 1 (PD-1) inhibitors, has emerged as a promising strategy in multiple tumor types, including ICC. Microsatellite instability-high (MSI-H) is an important biomarker for ICIs in solid tumors. The response rate in patients with MSI-H is significantly higher than in those with microsatellite stability/microsatellite instability-low. And approximately 80-90% of the patients with MSI-H could maintain sustained clinical benefits once they had an initial response. However, some patients could have primary resistance at the beginning, and some might have acquired resistance after long-term treatment. Case Presentation We present the case of an ICC patient with MSI-H who suffered rapid progression after a short-term remission with camrelizumab, a kind of PD-1 inhibitor, as second-line treatment. The patient's genomic and immune features were analyzed by next-generation sequencing and multiplex immunofluorescence staining to explore the possible mechanisms of the rapidly acquired resistance of ICIs in this MSI-H case. Conclusion The genomic and immunohistochemical analysis showed that TGFBR2 mutation, loss of HLA B44 supertype, carrying B62 supertype, and increased PD-L1+ cells, macrophages, and Tregs in the tumor microenvironment might be related to the nonsustain benefit of ICIs in this MSI-H patient.
Collapse
Affiliation(s)
- Zhuo Cheng
- Department of Oncology, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Tianmei Zeng
- Department of Oncology, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Guang Yang
- Department of Oncology, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Di Liu
- Genecast Biotechnology Co., Ltd, Wuxi City, China
| | - Zhi Zheng
- Department of General Surgery, Shanghai General Hospital Jiading Branch, Shanghai, China
| | - Zhengang Yuan
- Department of Oncology, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| |
Collapse
|
240
|
Zhang Z, Wang G, Shao X, Wu H, Su X, Zhu L, Ji Z. A Novel Prognostic Biomarker CCR8 for Gastric Cancer and Anti-CCR8 Blockade Attenuate the Immunosuppressive Capacity of Tregs In Vitro. Cancer Biother Radiopharm 2023; 38:415-424. [PMID: 37102694 DOI: 10.1089/cbr.2022.0095] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023] Open
Abstract
Objective: To investigate the immunotherapeutic roles and functions of C-C Motif Chemokine Receptor 8 (CCR8) molecule in gastric cancer (GC). Materials and Methods: Clinicopathological features of 95 GC cases were collected by a follow-up survey. The expression level of CCR8 was measured by immunohistochemistry (IHC) staining and analyzed with the cancer genome atlas database. The relationship between CCR8 expression and Clinicopathological features of GC cases was evaluated by univariate and multivariate analysis. Flow cytometry was used to determine the expression of cytokines and the proliferation of CD4+ regulator T cells (Tregs) and CD8+ T cells. Results: An upregulated expression of CCR8 in GC tissues was associated with tumor grade, nodal metastasis, and overall survival (OS). Tumor-infiltrated Tregs with higher expression of CCR8 produced more IL10 molecules in vitro. In addition, anti-CCR8 blocking downregulated IL10 expression produced by CD4+ Tregs, and reversed the suppression by Tregs on the secretion and proliferation of CD8+ T cells. Conclusion: CCR8 molecule could be a prognostic biomarker for GC cases and a therapeutic target for immune treatments.
Collapse
Affiliation(s)
- Zhigang Zhang
- Medical School of Southeast University, Nanjing, China
- Department of General Surgery, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Guoqing Wang
- Department of Pathology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Xiangyu Shao
- Medical School of Southeast University, Nanjing, China
- Department of General Surgery, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Hailu Wu
- Medical School of Southeast University, Nanjing, China
| | - Xiangyu Su
- Medical School of Southeast University, Nanjing, China
| | - Long Zhu
- Medical School of Southeast University, Nanjing, China
| | - Zhenling Ji
- Department of General Surgery, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| |
Collapse
|
241
|
Huang Y, Wu X, Tang S, Wu H, Nasri U, Qin Q, Song Q, Wang B, Tao H, Chong AS, Riggs AD, Zeng D. Donor programmed cell death 1 ligand 1 is required for organ transplant tolerance in major histocompatibility complex-mismatched mixed chimeras although programmed cell death 1 ligand 1 and major histocompatibility complex class II are not required for inducing chimerism. Am J Transplant 2023; 23:1116-1129. [PMID: 37105316 DOI: 10.1016/j.ajt.2023.04.022] [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: 11/16/2022] [Revised: 04/19/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023]
Abstract
Induction of major histocompatibility complex (MHC) human leukocyte antigen (HLA)-mismatched mixed chimerism is a promising approach for organ transplantation tolerance; however, human leukocyte antigen-mismatched stable mixed chimerism has not been achieved in the clinic. Tolerogenic dendritic cell (DC) expression of MHC class II (MHC II) and programmed cell death 1 ligand 1 (PD-L1) is important for immune tolerance, but whether donor-MHC II or PD-L1 is required for the induction of stable MHC-mismatched mixed chimerism and transplant tolerance is unclear. Here, we show that a clinically applicable radiation-free regimen can establish stable MHC-mismatched mixed chimerism and organ transplant tolerance in murine models. Induction of MHC-mismatched mixed chimerism does not require donor cell expression of MHC II or PD-L1, but donor-type organ transplant tolerance in the mixed chimeras (MC) requires the donor hematopoietic cells and the organ transplants to express PD-L1. The PD-L1 expressed by donor hematopoietic cells and the programmed cell death 1 expressed by host cells augment host-type donor-reactive CD4+ and CD8+ T cell anergy/exhaustion and differentiation into peripheral regulatory T (pTreg) cells in association with the organ transplant tolerance in the MC. Conversely, host-type Treg cells augment the expansion of donor-type tolerogenic CD8+ DCs that express PD-L1. These results indicate that PD-L1 expressed by donor-type tolerogenic DCs and expansion of host-type pTreg cells in MHC-mismatched MCs play critical roles in mediating organ transplant tolerance.
Collapse
Affiliation(s)
- Yaxun Huang
- Department of Liver Transplantation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA; Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Xiwei Wu
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Shanshan Tang
- Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA; Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Huiqing Wu
- Department of Pathology, City of Hope National Medical Center, Duarte, California, USA
| | - Ubaydah Nasri
- Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA; Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Qi Qin
- Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA; Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA; Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingxiao Song
- Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA; Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Bixin Wang
- Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA; Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA; Fujian Medical University Center of Translational Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hansen Tao
- Arthur Riggs Diabetes and Metabolism Research Institute, Summer Student Academy of City of Hope, Duarte, California, USA
| | - Anita S Chong
- The section of Transplantation, Department of Surgery, the University of Chicago, Chicago, Illinois, USA
| | - Arthur D Riggs
- Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Defu Zeng
- Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA; Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA.
| |
Collapse
|
242
|
Sun XX, Nosrati Z, Ko J, Lee CM, Bennewith KL, Bally MB. Induced Vascular Normalization-Can One Force Tumors to Surrender to a Better Microenvironment? Pharmaceutics 2023; 15:2022. [PMID: 37631236 PMCID: PMC10458586 DOI: 10.3390/pharmaceutics15082022] [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: 05/03/2023] [Revised: 06/19/2023] [Accepted: 07/14/2023] [Indexed: 08/27/2023] Open
Abstract
Immunotherapy has changed the way many cancers are being treated. Researchers in the field of immunotherapy and tumor immunology are investigating similar questions: How can the positive benefits achieved with immunotherapies be enhanced? Can this be achieved through combinations with other agents and if so, which ones? In our view, there is an urgent need to improve immunotherapy to make further gains in the overall survival for those patients that should benefit from immunotherapy. While numerous different approaches are being considered, our team believes that drug delivery methods along with appropriately selected small-molecule drugs and drug candidates could help reach the goal of doubling the overall survival rate that is seen in some patients that are given immunotherapeutics. This review article is prepared to address how immunotherapies should be combined with a second treatment using an approach that could realize therapeutic gains 10 years from now. For context, an overview of immunotherapy and cancer angiogenesis is provided. The major targets in angiogenesis that have modulatory effects on the tumor microenvironment and immune cells are highlighted. A combination approach that, for us, has the greatest potential for success involves treatments that will normalize the tumor's blood vessel structure and alter the immune microenvironment to support the action of immunotherapeutics. So, this is reviewed as well. Our focus is to provide an insight into some strategies that will engender vascular normalization that may be better than previously described approaches. The potential for drug delivery systems to promote tumor blood vessel normalization is considered.
Collapse
Affiliation(s)
- Xu Xin Sun
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- NanoMedicines Innovation Network, Vancouver, BC V6T 1Z3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
| | - Zeynab Nosrati
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
| | - Janell Ko
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
| | - Che-Min Lee
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Kevin L. Bennewith
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Marcel B. Bally
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- NanoMedicines Innovation Network, Vancouver, BC V6T 1Z3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
243
|
Zou W, Green DR. Beggars banquet: Metabolism in the tumor immune microenvironment and cancer therapy. Cell Metab 2023; 35:1101-1113. [PMID: 37390822 PMCID: PMC10527949 DOI: 10.1016/j.cmet.2023.06.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/23/2023] [Accepted: 06/05/2023] [Indexed: 07/02/2023]
Abstract
Metabolic programming in the tumor microenvironment (TME) alters tumor immunity and immunotherapeutic response in tumor-bearing mice and patients with cancer. Here, we review immune-related functions of core metabolic pathways, key metabolites, and crucial nutrient transporters in the TME, discuss their metabolic, signaling, and epigenetic impact on tumor immunity and immunotherapy, and explore how these insights can be applied to the development of more effective modalities to potentiate the function of T cells and sensitize tumor cell receptivity to immune attack, thereby overcoming therapeutic resistance.
Collapse
Affiliation(s)
- Weiping Zou
- Departments of Surgery and Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Graduate Programs in Immunology and Cancer Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
244
|
Moreno Ayala MA, Campbell TF, Zhang C, Dahan N, Bockman A, Prakash V, Feng L, Sher T, DuPage M. CXCR3 expression in regulatory T cells drives interactions with type I dendritic cells in tumors to restrict CD8 + T cell antitumor immunity. Immunity 2023; 56:1613-1630.e5. [PMID: 37392735 PMCID: PMC10752240 DOI: 10.1016/j.immuni.2023.06.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 03/07/2023] [Accepted: 06/06/2023] [Indexed: 07/03/2023]
Abstract
Infiltration of regulatory T (Treg) cells, an immunosuppressive population of CD4+ T cells, into solid cancers represents a barrier to cancer immunotherapy. Chemokine receptors are critical for Treg cell recruitment and cell-cell interactions in inflamed tissues, including cancer, and thus are an ideal therapeutic target. Here, we show in multiple cancer models that CXCR3+ Treg cells were increased in tumors compared with lymphoid tissues, exhibited an activated phenotype, and interacted preferentially with CXCL9-producing BATF3+ dendritic cells (DCs). Genetic ablation of CXCR3 in Treg cells disrupted DC1-Treg cell interactions and concomitantly increased DC-CD8+ T cell interactions. Mechanistically, CXCR3 ablation in Treg cells increased tumor antigen-specific cross-presentation by DC1s, increasing CD8+ T cell priming and reactivation in tumors. This ultimately impaired tumor progression, especially in combination with anti-PD-1 checkpoint blockade immunotherapy. Overall, CXCR3 is shown to be a critical chemokine receptor for Treg cell accumulation and immune suppression in tumors.
Collapse
Affiliation(s)
- Mariela A Moreno Ayala
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Timothy F Campbell
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chenyu Zhang
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Noa Dahan
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Alissa Bockman
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Varsha Prakash
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Lawrence Feng
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Theo Sher
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michel DuPage
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
245
|
Valentini N, Requejo Cier CJ, Lamarche C. Regulatory T-cell dysfunction and its implication for cell therapy. Clin Exp Immunol 2023; 213:40-49. [PMID: 37158407 PMCID: PMC10324551 DOI: 10.1093/cei/uxad051] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/04/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023] Open
Abstract
Regulatory T cells (Tregs) are a subtype of CD4+ T cells that can mediate immune tolerance by a multitude of immunomodulatory mechanisms. Treg-based adoptive immunotherapy is currently being tested in multiple phases I and II clinical trials in transplantation and autoimmune diseases. We have learned from the work done on conventional T cells that distinct mechanistic states can define their dysfunctions, such as exhaustion, senescence, and anergy. All three can negatively impact the therapeutic effectiveness of T-cell-based therapies. However, whether Tregs are susceptible to such dysfunctional states is not well studied, and results are sometimes found to be controversial. In addition, Treg instability and loss of FOXP3 expression is another Treg-specific dysfunction that can decreasein their suppressive potential. A better understanding of Treg biology and pathological states will be needed to compare and interpret the results of the different clinical and preclinical trials. We will review herein Tregs' mechanisms of action, describe different T-cell dysfunction subtypes and how and if they apply to Tregs (exhaustion, senescence, anergy, and instability), and finally how this knowledge should be taken into consideration when designing and interpreting Treg adoptive immunotherapy trials.
Collapse
Affiliation(s)
- Nicolas Valentini
- Medicine Department, Hôpital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada
- Microbiology, Infectiology and Immunology Department, Université de Montréal, Montreal, QC, Canada
| | - Christopher J Requejo Cier
- Medicine Department, Hôpital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada
- Microbiology, Infectiology and Immunology Department, Université de Montréal, Montreal, QC, Canada
| | - Caroline Lamarche
- Medicine Department, Hôpital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada
- Medicine Department, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
246
|
Liu S, Wang H, Shao X, Chen H, Chao S, Zhang Y, Gao Z, Yao Q, Zhang P. Advances in PD-1 signaling inhibition-based nano-delivery systems for tumor therapy. J Nanobiotechnology 2023; 21:207. [PMID: 37403095 PMCID: PMC10318732 DOI: 10.1186/s12951-023-01966-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/22/2023] [Indexed: 07/06/2023] Open
Abstract
In recent years, cancer immunotherapy has emerged as an exciting cancer treatment. Immune checkpoint blockade brings new opportunities for more researchers and clinicians. Programmed cell death receptor-1 (PD-1) is a widely studied immune checkpoint, and PD-1 blockade therapy has shown promising results in a variety of tumors, including melanoma, non-small cell lung cancer and renal cell carcinoma, which greatly improves patient overall survival and becomes a promising tool for the eradication of metastatic or inoperable tumors. However, low responsiveness and immune-related adverse effects currently limit its clinical application. Overcoming these difficulties is a major challenge to improve PD-1 blockade therapies. Nanomaterials have unique properties that enable targeted drug delivery, combination therapy through multidrug co-delivery strategies, and controlled drug release through sensitive bonds construction. In recent years, combining nanomaterials with PD-1 blockade therapy to construct novel single-drug-based or combination therapy-based nano-delivery systems has become an effective mean to address the limitations of PD-1 blockade therapy. In this study, the application of nanomaterial carriers in individual delivery of PD-1 inhibitors, combined delivery of PD-1 inhibitors and other immunomodulators, chemotherapeutic drugs, photothermal reagents were reviewed, which provides effective references for designing new PD-1 blockade therapeutic strategies.
Collapse
Affiliation(s)
- Songlin Liu
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Haiyang Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
- Qingdao University of Science and Technology, Qingdao, 266042, People's Republic of China
| | - Xinzhe Shao
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Haonan Chen
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Shushu Chao
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Yanyan Zhang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Zhaoju Gao
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Qingqiang Yao
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| | - Pingping Zhang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China.
| |
Collapse
|
247
|
Yang Y, Li J, Jing C, Zhai Y, Bai Z, Yang Y, Deng W. Inhibition of neuroactive ligand-receptor interaction pathway can enhance immunotherapy response in colon cancer: an in silico study. Expert Rev Anticancer Ther 2023; 23:1205-1215. [PMID: 37555253 DOI: 10.1080/14737140.2023.2245567] [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: 02/08/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND The potential mechanism underlying the association between Homologous recombination deficiency (HRD) and immunotherapy in colon cancer has not been investigated. METHODS The exon sequencing data and transcriptome data of 456 colon adenocarcinoma (COAD) patients were obtained from the TCGA database. Pathway activity score was calculated by GSVA methods and engaged in further survival analysis. The prognostic value of the candidate pathways was validated in an external GEO cohort and an immunotherapy cohort. RESULTS Patients with high HRD were associated with poor prognosis, lower tumor mutation burden and microsatellite instability, higher fraction genome alteration, and less sensitivity to immunotherapy in COAD. And then, the neuroactive ligand-receptor interaction pathway was over-activated in high-HRD tumors and associated with immunosuppression in colon cancer with high HRD. Besides, the pathway was associated with prognosis and immunotherapy response in COAD. Moreover, genes in this pathway such as LTB4R2 can be used as a novel target for therapy development in colon cancer. CONCLUSION Our study not only revealed the potential mechanism of HRD and the function of the neuroactive ligand-receptor interaction pathway in colon cancer but also provided new clues for the improvement of immunotherapy response in colon cancer.
Collapse
Affiliation(s)
- Yun Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jun Li
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chao Jing
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yuhao Zhai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhigang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yingchi Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Wei Deng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
248
|
Pulanco MC, Madsen AT, Tanwar A, Corrigan DT, Zang X. Recent advancements in the B7/CD28 immune checkpoint families: new biology and clinical therapeutic strategies. Cell Mol Immunol 2023; 20:694-713. [PMID: 37069229 PMCID: PMC10310771 DOI: 10.1038/s41423-023-01019-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/25/2023] [Indexed: 04/19/2023] Open
Abstract
The B7/CD28 families of immune checkpoints play vital roles in negatively or positively regulating immune cells in homeostasis and various diseases. Recent basic and clinical studies have revealed novel biology of the B7/CD28 families and new therapeutics for cancer therapy. In this review, we discuss the newly discovered KIR3DL3/TMIGD2/HHLA2 pathways, PD-1/PD-L1 and B7-H3 as metabolic regulators, the glycobiology of PD-1/PD-L1, B7x (B7-H4) and B7-H3, and the recently characterized PD-L1/B7-1 cis-interaction. We also cover the tumor-intrinsic and -extrinsic resistance mechanisms to current anti-PD-1/PD-L1 and anti-CTLA-4 immunotherapies in clinical settings. Finally, we review new immunotherapies targeting B7-H3, B7x, PD-1/PD-L1, and CTLA-4 in current clinical trials.
Collapse
Affiliation(s)
- Marc C Pulanco
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, 10461, USA
| | - Anne T Madsen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, 10461, USA
- Department of Urology, Albert Einstein College of Medicine, New York, NY, 10461, USA
| | - Ankit Tanwar
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, 10461, USA
- Department of Oncology, Albert Einstein College of Medicine, New York, NY, 10461, USA
| | - Devin T Corrigan
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, 10461, USA
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, 10461, USA.
- Department of Urology, Albert Einstein College of Medicine, New York, NY, 10461, USA.
- Department of Oncology, Albert Einstein College of Medicine, New York, NY, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, 10461, USA.
| |
Collapse
|
249
|
Jiang Y, Yu J, Zhu T, Bu J, Hu Y, Liu Y, Zhu X, Gu X. Involvement of FAM83 Family Proteins in the Development of Solid Tumors: An Update Review. J Cancer 2023; 14:1888-1903. [PMID: 37476189 PMCID: PMC10355199 DOI: 10.7150/jca.83420] [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: 02/10/2023] [Accepted: 06/16/2023] [Indexed: 07/22/2023] Open
Abstract
FAM83 family members are a group of proteins that have been implicated in various solid tumors. In this updated review, we mainly focus on the cellular localization, molecular composition, and biological function of FAM83 family proteins in solid tumors. We discussed the factors that regulate abnormal protein expression and alterations in the functional activities of solid tumor cells (including non-coding microRNAs and protein modifiers) and potential mechanisms of tumorigenesis (including the MAPK, WNT, and TGF-β signaling pathways). Further, we highlighted the application of FAM83 family proteins in the diagnoses and treatment of different cancers, such as breast, lung, liver, and ovarian cancers from two aspects: molecular marker diagnosis and tumor drug resistance. We described the overexpression of FAM83 genes in various human malignant tumor cells and its relationship with tumor proliferation, migration, invasion, transformation, and drug resistance. Moreover, we explored the prospects and challenges of using tumor treatments based on the FAM83 proteins. Overall, we provide a theoretical basis for harnessing FAM83 family proteins as novel targets in cancer treatment. We believe that this review opens up open new directions for solid tumor treatment in clinical practice.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning province, P.R. China
| | - Jiahui Yu
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Tong Zhu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning province, P.R. China
| | - Jiawen Bu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning province, P.R. China
| | - Yueting Hu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning province, P.R. China
| | - Yang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning province, P.R. China
| | - Xudong Zhu
- Department of General Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Xi Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning province, P.R. China
| |
Collapse
|
250
|
Liu G, Zhang Z, Wu Y, Feng J, Lan Y, Dong D, Liu Y, Yuan H, Tai G, Li S, Ni W. Anti-PD-L1 antibody reverses the immune tolerance induced by multiple MUC1-MBP vaccine immunizations by increasing the CD80/PD-L1 ratio, resulting in DC maturation, and decreasing Treg activity in B16-MUC1 melanoma-bearing mice. Int Immunopharmacol 2023; 121:110487. [PMID: 37364328 DOI: 10.1016/j.intimp.2023.110487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/28/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023]
Abstract
In this study, we explored the possible mechanism of tumor tolerance induced by multiple repeated immunizations with a tumor vaccine (MUC1-MBP fusion protein plus CpG2006). We first analyzed the mechanism of tolerance by immunizing tumor-bearing mice 2, 5, or 8 times and found that compared with five immunizations with the M-M vaccine, eight immunizations increased tumor volume and weight and Treg levels, while the proportions of Th1 and Tc1 cells in the spleen and lymph nodes were decreased. In particular, the M-M vaccine induced PD-L1 expression in CD11c + DCs and decreased their CD80/PD-L1 ratio. Therefore, the mechanism of tolerance induction by multiple immunizations with the M-M vaccine was investigated by focusing on the CD80/PD-L1 ratio, and an anti-PD-L1 antibody (αPD-L1) and the M-M vaccine were used in combination to treat melanoma. The results showed that αPD-L1 increased the CD80/PD-L1 ratio and enhanced the maturation of cDC1s by blocking PD-L1 on DCs, which potentially increased the activity of Th1 and Tc1 cells. Furthermore, the combination of the M-M vaccine with αPD-L1 decreased the activity and proportion of Tregs, which reversed the immune tolerance induced by eight immunizations with the vaccine. This study reveals the mechanism of the combination of M-M and αPD-L1 and provides a new combination strategy for improving the therapeutic effect of the M-M vaccine, laying a theoretical basis for the clinical application of the vaccine.
Collapse
Affiliation(s)
- Guomu Liu
- Department of Dermatology and Venereology, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Zenan Zhang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yixuan Wu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Jingyue Feng
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yue Lan
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Dai Dong
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yu Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Hongyan Yuan
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Guixiang Tai
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Shanshan Li
- Department of Dermatology and Venereology, The First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Weihua Ni
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|