1
|
Palazzo L, Pieri V, Berzero G, Filippi M. CAR-T Cells for the Treatment of Central Nervous System Tumours: Known and Emerging Neurotoxicities. Brain Sci 2024; 14:1220. [PMID: 39766419 PMCID: PMC11727498 DOI: 10.3390/brainsci14121220] [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: 11/01/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 01/15/2025] Open
Abstract
The advent of chimeric antigen receptor (CAR)-T cells has recently changed the prognosis of relapsing/refractory diffuse large B-cell lymphomas, showing response rates as high as 60 to 80%. Common toxicities reported in the pivotal clinical trials include the cytokine release syndrome (CRS) and the Immune effector Cell-Associated Neurotoxicity Syndrome (ICANS), a stereotyped encephalopathy related to myeloid cell activation and blood-brain barrier dysfunction, presenting with a distinctive cascade of dysgraphia, aphasia, disorientation, attention deficits, vigilance impairment, motor symptoms, seizures, and diffuse brain oedema. The tremendous oncological efficacy of CAR-T cells observed in systemic B-cell malignancies is leading to their growing use in patients with primary or secondary central nervous system (CNS) lymphomas and in patients with solid tumours, including several CNS cancers. Early studies conducted in adult and paediatric patients with solid CNS tumours reported a distinct profile of neurotoxicity referred to as Tumour inflammation-associated neurotoxicity (TIAN), corresponding to local inflammation at the tumour site manifesting with focal neurological deficits or mechanical complications (e.g., obstructive hydrocephalus). The present review summarises available data on the efficacy and safety of CAR-T cells for solid and haematological CNS malignancies, emphasising known and emerging phenotypes, ongoing challenges, and future perspectives.
Collapse
Affiliation(s)
- Leonardo Palazzo
- Neurology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (L.P.); (V.P.); (M.F.)
- Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Valentina Pieri
- Neurology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (L.P.); (V.P.); (M.F.)
- Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Giulia Berzero
- Neurology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (L.P.); (V.P.); (M.F.)
- Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Massimo Filippi
- Neurology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (L.P.); (V.P.); (M.F.)
- Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Neurorehabilitation Unit, Neurophysiology Unit, Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| |
Collapse
|
2
|
Liu D, Wang L, Guo Y. Advances in and prospects of immunotherapy for prostate cancer. Cancer Lett 2024; 601:217155. [PMID: 39127338 DOI: 10.1016/j.canlet.2024.217155] [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: 05/30/2024] [Revised: 07/07/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Immunotherapy has shown promising therapeutic effects in hematological malignancies and certain solid tumors and has emerged as a critical and highly potential treatment modality for cancer. However, prostate cancer falls under the category of immune-resistant cold tumors, for which immunotherapy exhibits limited efficacy in patients with solid tumors. Thus, it is important to gain a deeper understanding of the tumor microenvironment in prostate cancer to facilitate immune system activation and overcome immune suppression to advance immunotherapy for prostate cancer. In this review, we discuss the immunosuppressive microenvironment of prostate cancer, which is characterized by the presence of few tumor-infiltrating lymphocytes, abundant immunosuppressive cells, low immunogenicity, and a noninflammatory phenotype, which significantly influences the efficacy of immunotherapy for prostate cancer. Immunotherapy is mainly achieved by activating the host immune system and overcoming immunosuppression. In this regard, we summarize the therapeutic advances in immune checkpoint blockade, immunogenic cell death, reversal of the immunosuppressive tumor microenvironment, tumor vaccines, immune adjuvants, chimeric antigen receptor T-cell therapy, and overcoming penetration barriers in prostate cancer, with the aim of providing novel research insights and approaches to enhance the effectiveness of immunotherapy for prostate cancer.
Collapse
Affiliation(s)
- Deng Liu
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China; Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Luofu Wang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
3
|
Zhou R, Xu CL. Application effect of case management nursing based on patient safety in patients with prostate cancer. World J Clin Cases 2024; 12:6070-6076. [PMID: 39328862 PMCID: PMC11326094 DOI: 10.12998/wjcc.v12.i27.6070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/10/2024] [Accepted: 07/02/2024] [Indexed: 07/29/2024] Open
Abstract
BACKGROUND Globally, prostate cancer has become a major threat to men's health, with an increasing incidence and causes serious effects on the quality and length of life of patients. Despite the rapid development of medical technology, which provides treatments, including surgery, radiotherapy, and endocrine therapy, the treatment of patients with prostate cancer, especially with endocrine therapy, has become a major challenge in clinical treatment owing to the lengthy course of treatment, side effects of drugs, and impact of the disease on the psychological and physiological functioning of the patient, producing poor treatment adherence and a decline in quality of life. AIM To explore effects of nursing intervention prioritizing patient safety and case management in patients with prostate cancer undergoing endocrine therapy. METHODS Eighty patients with prostate cancer who received endocrine therapy at our hospital between January 2022 and January 2024 were divided into observation and control groups with 40 cases per group. The control group was treated using a routine nursing workflow while the observation group received case management nursing guidance prioritizing patient safety. Scores for anxiety and depression, prostate cancer symptoms, and quality of life and patient compliance and satisfaction were compared between the groups after three months of intervention. RESULTS After the nursing intervention, the anxiety and depression scores of the observation group were significantly lower than those of the control group (P < 0.05). The quality of life score, sexual function, and hormone function were significantly higher than those in the control group (P < 0.05). CONCLUSION Case management guidance based on patient safety effectively reduced anxiety and depression in patients undergoing endocrine therapy for prostate cancer and improved their quality of life, treatment compliance, and satisfaction.
Collapse
Affiliation(s)
- Ru Zhou
- Department of Comprehensive Surgery II, Jiangnan University Affiliated Hospital, Wuxi 214101, Jiangsu Province, China
| | - Chan-Ling Xu
- Department of Comprehensive Surgery II, Jiangnan University Affiliated Hospital, Wuxi 214101, Jiangsu Province, China
| |
Collapse
|
4
|
Zhang J, Chadha JS. Developmental Therapeutics in Metastatic Prostate Cancer: New Targets and New Strategies. Cancers (Basel) 2024; 16:3098. [PMID: 39272956 PMCID: PMC11393880 DOI: 10.3390/cancers16173098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
There is an unmet need to develop new treatments for metastatic prostate cancer. With the development of targeted radioligand therapies, bispecific T cell engagers, antibody-drug conjugates and chimeric antigen receptor T cell (CAR T) therapies, tumor-associated cell surface antigens have emerged as new therapeutic targets in metastatic prostate cancer. Ongoing and completed clinical trials targeting prostate-specific membrane antigen (PSMA), six transmembrane epithelial antigens of the prostate 1 (STEAP1), kallikrein-related peptidase 2 (KLK2), prostate stem cell antigen (PSCA), and delta-like protein 3 (DLL3) in metastatic prostate cancer were reviewed. Strategies for sequential or combinational therapy were discussed.
Collapse
Affiliation(s)
- Jingsong Zhang
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Juskaran S Chadha
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| |
Collapse
|
5
|
Sun H, Li X, Liu Q, Sheng H, Zhu L. pH-responsive self-assembled nanoparticles for tumor-targeted drug delivery. J Drug Target 2024; 32:672-706. [PMID: 38682299 DOI: 10.1080/1061186x.2024.2349124] [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: 10/07/2023] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Recent advances in the field of drug delivery have opened new avenues for the development of novel nanodrug delivery systems (NDDS) in cancer therapy. Self-assembled nanoparticles (SANPs) based on tumour microenvironment have great advantages in improving antitumor effect, and pH-responsive SANPs prepared by the combination of pH-responsive nanomaterials and self-assembly technology can effectively improve the efficacy and reduce the systemic toxicity of antitumor drugs. In this review, we describe the characteristics of self-assembly and its driving force, the mechanism of pH-responsive NDDS, and the nanomaterials for pH-responsive SANPs type. A series of pH-responsive SANPs for tumour-targeted drug delivery are discussed, with an emphasis on the relation between structural features and theranostic performance.
Collapse
Affiliation(s)
- Henglai Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinyu Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qian Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
6
|
Li X, Zhao L, Li W, Gao P, Zhang N. HER2-targeting CAR-T cells show highly efficient anti-tumor activity against glioblastoma both in vitro and in vivo. Genes Immun 2024; 25:201-208. [PMID: 38702509 PMCID: PMC11178492 DOI: 10.1038/s41435-024-00275-6] [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: 02/04/2024] [Revised: 04/16/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Glioblastoma (GBM) is the most common and aggressive malignant primary brain tumor in adults. Current treatment options for GBM include surgical resection, radiation, and chemotherapy, which predominantly slow cancer growth and reduce symptoms, resulting in a 5-year survival rate of no more than 10%. Chimeric antigen receptor (CAR) T-cell therapy is a new class of cellular immunotherapy that has made great progress in treating malignant tumors. Human epidermal growth factor receptor 2 (HER2) is overexpressed in GBM and may provide a potential therapeutic target for GBM treatment. In this study, we constructed third-generation CAR-T cells targeting the HER2 antigen in GBM. HER2-CAR-T cells showed effective anti-tumor activity both in vitro and in vivo. Furthermore, HER2-specific CAR-T cells exhibited strong cytotoxicity and cytokine-secreting abilities against GBM cells in vitro. Anti-HER2 CAR-T cells also exhibited increased cytotoxicity with increasing effector-to-target ratios. Anti-HER2 CAR-T cells delivered via peritumoral injection successfully stunted tumor progression in vivo. Moreover, peritumoral intravenous administration of anti-HER2 CAR-T cells resulted in therapeutic improvement against GBM cells compared with intravenous administration. In conclusion, our study shows that HER2 CAR-T cells represent an emerging immunotherapy for treating GBM.
Collapse
Affiliation(s)
- Xueying Li
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, China
- Research Institute for Microbial Diseases and World Premier International Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Lifen Zhao
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, China
| | - Wenzhe Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong Province, 515041, China
| | - Peng Gao
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, China.
| | - Nianzhu Zhang
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, 116023, China.
| |
Collapse
|
7
|
Chao Z, Wang Z, Li L, Jiang Y, Tang Y, Wang Y, Hao X, Zhang C, Guo X, Yu W, Cheng F, Wang Z. Poly (ADP-ribose) Polymerase Inhibitors in Patients with Metastatic Castration-Resistant Prostate Cancer: A Meta-Analysis of Randomized Controlled Trials. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:2198. [PMID: 38138301 PMCID: PMC10744677 DOI: 10.3390/medicina59122198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
Context: Several recent randomized controlled trials (RCTs) have reported on the survival benefits of poly (ADP-ribose) polymerase inhibitors (PARPi) compared to standard-of-care (SOC) treatment (enzalutamide, abiraterone, or docetaxel) in patients with metastatic castration-resistant prostate cancer (mCRPC). However, there is a limited integrated analysis of high-quality evidence comparing the efficacy and safety of PARPi and SOC treatments in this context. Objective: This study aims to comprehensively analyze the survival benefits and adverse events associated with PARPi and SOC treatments through a head-to-head meta-analysis in mCRPC. Evidence acquisition: A systematic review search was conducted in PubMed, Embase, Clinical trials, and the Central Cochrane Registry in July 2023. RCTs were assessed following the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. The systematic review was prospectively registered on PROSPERO (CRD42023441034). Evidence synthesis: A total of 8 studies, encompassing 2341 cases in the PARPi treatment arm and 1810 cases in the controlled arm, were included in the qualitative synthesis. The hazard ratio (HR) for radiographic progression-free survival (rPFS) and overall survival (OS) were 0.74 (95% CI, 0.61-0.90) and 0.89 (95% CI, 0.80-0.99), respectively, in the intention-to-treatment patients. For subgroup analysis, HRs for rPFS and OS in the BRCA-mutated subgroup were 0.39 (95% CI, 0.28-0.55) and 0.62 (95% CI, 0.38-0.99), while in the HRR-mutated subgroup, HR for rPFS was 0.57 (95% CI, 0.48-0.69) and for OS was 0.77 (95% CI, 0.64-0.93). The odds ratio (OR) for all grades of adverse events (AEs) and AEs with severity of at least grade 3 were 3.86 (95% CI, 2.53-5.90) and 2.30 (95% CI, 1.63-3.26), respectively. Conclusions: PARP inhibitors demonstrate greater effectiveness than SOC treatments in HRR/BRCA-positive patients with mCRPC. Further research is required to explore ways to reduce adverse event rates and investigate the efficacy of HRR/BRCA-negative patients.
Collapse
Affiliation(s)
- Zheng Chao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.C.)
| | - Zefeng Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430030, China
| | - Le Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.C.)
| | - Yi Jiang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yunxing Tang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing 100034, China
| | - Yanan Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.C.)
| | - Xiaodong Hao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.C.)
| | - Chunyu Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.C.)
| | - Xiangdong Guo
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.C.)
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430030, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430030, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.C.)
| |
Collapse
|
8
|
Ou L, Su C, Liang L, Duan Q, Li Y, Zang H, He Y, Zeng R, Li Y, Zhou H, Xiao L. Current status and future prospects of chimeric antigen receptor-T cell therapy in lymphoma research: A bibliometric analysis. Hum Vaccin Immunother 2023; 19:2267865. [PMID: 37846106 PMCID: PMC10583622 DOI: 10.1080/21645515.2023.2267865] [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: 05/31/2023] [Accepted: 10/04/2023] [Indexed: 10/18/2023] Open
Abstract
CAR-T cell therapy, a novel therapeutic approach that has attracted much attention in the field of cancer treatment at present, has become the subject of many studies and has shown great potential in the treatment of hematological malignancies, such as leukemia and lymphoma. This study aims to analyze the characteristics of articles published on CAR-T cell therapy in the lymphoma field and explore the existing hotspots and frontiers. The relevant articles published from 2013 to 2022 were retrieved from the Web of Science Core Collection. CiteSpace, VOSviewer, Bibliometric online analysis platform, Microsoft Excel, and R software were used for bibliometric analysis and visualization. The number of publications related to the research has been increasing year by year, including 1023 articles and 760 reviews from 62 countries and regions, 2092 institutions, 1040 journals, and 8727 authors. The United States, China, and Germany are the main publishing countries in this research field. The top 10 institutions are all from the United States, the journal with the highest impact factor is BLOOD, the author with the most publications is Frederick L Locke, and the most influential author is Carl H June. The top three keywords are "Lymphoma," "Immunotherapy," and "Therapy." "Maude (2014)" is the most cited and strongest burstiness reference over the past decade. This study provides a comprehensive bibliometric analysis of CAR-T cell therapy in lymphoma, which can help researchers understand the current research hotspots in this field, explore potential research directions, and identify future development trends.
Collapse
Affiliation(s)
- Lijia Ou
- Department of Lymphoma & Hematology, The Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Chang Su
- Department of Lymphoma & Hematology, The Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Liang Liang
- Department of Lymphoma & Hematology, The Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qintong Duan
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yufeng Li
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hui Zang
- Department of Human Anatomy and Histoembryology of School of Basic Medical Sciences, Yiyang Medical College, Yiyang, Hunan, China
| | - Yizi He
- Department of Lymphoma & Hematology, The Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Ruolan Zeng
- Department of Lymphoma & Hematology, The Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Yajun Li
- Department of Lymphoma & Hematology, The Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Hui Zhou
- Department of Lymphoma & Hematology, The Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Ling Xiao
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
9
|
Pu Z, Wang TB, Mou L. Revolutionizing cancer immunotherapy in solid tumor: CAR engineering and single-cell sequencing insights. Front Immunol 2023; 14:1310285. [PMID: 38090577 PMCID: PMC10712310 DOI: 10.3389/fimmu.2023.1310285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
The global increase in cancer incidence presents significant economic and societal challenges. While chimeric antigen receptor-modified T cell (CAR-T) therapy has demonstrated remarkable success in hematologic malignancies and has earned FDA approval, its translation to solid tumors encounters faces significant obstacles, primarily centered around identifying reliable tumor-associated antigens and navigating the complexities of the tumor microenvironment. Recent developments in single-cell RNA sequencing (scRNA-seq) have greatly enhanced our understanding of tumors by offering high-resolution, unbiased analysis of cellular heterogeneity and molecular patterns. These technologies have revolutionized our comprehension of tumor immunology and have led to notable progress in cancer immunotherapy. This mini-review explores the progress of chimeric antigen receptor (CAR) cell therapy in solid tumor treatment and the application of scRNA-seq at various stages following the administration of CAR cell products into the body. The advantages of scRNA-seq are poised to further advance the investigation of the biological characteristics of CAR cells in vivo, tumor immune evasion, the impact of different cellular components on clinical efficacy, the development of clinically relevant biomarkers, and the creation of new targeted drugs and combination therapy approaches. The integration of scRNA-seq with CAR therapy represents a promising avenue for future innovations in cancer immunotherapy. This synergy holds the potential to enhance the precision and efficacy of CAR cell therapies while expanding their applications to a broader range of malignancies.
Collapse
Affiliation(s)
- Zuhui Pu
- Imaging Department, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
- MetaLife Lab, Shenzhen Institute of Translational Medicine, Shenzhen, Guangdong, China
| | - Tony Bowei Wang
- Biology Department, Skidmore College, Saratoga Springs, NY, United States
| | - Lisha Mou
- Imaging Department, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
- MetaLife Lab, Shenzhen Institute of Translational Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
10
|
Ma HY, Das J, Prendergast C, De Jong D, Braumuller B, Paily J, Huang S, Liou C, Giarratana A, Hosseini M, Yeh R, Capaccione KM. Advances in CAR T Cell Therapy for Non-Small Cell Lung Cancer. Curr Issues Mol Biol 2023; 45:9019-9038. [PMID: 37998743 PMCID: PMC10670348 DOI: 10.3390/cimb45110566] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
Since its first approval by the FDA in 2017, tremendous progress has been made in chimeric antigen receptor (CAR) T cell therapy, the adoptive transfer of engineered, CAR-expressing T lymphocyte. CAR T cells are all composed of three main elements: an extracellular antigen-binding domain, an intracellular signaling domain responsible for T cell activation, and a hinge that joins these two domains. Continuous improvement has been made in CARs, now in their fifth generation, particularly in the intracellular signaling domain responsible for T cell activation. CAR T cell therapy has revolutionized the treatment of hematologic malignancies. Nonetheless, the use of CAR T cell therapy for solid tumors has not attained comparable levels of success. Here we review the challenges in achieving effective CAR T cell therapy in solid tumors, and emerging CAR T cells that have shown great promise for non-small cell lung cancer (NSCLC). A growing number of clinical trials have been conducted to study the effect of CAR T cell therapy on NSCLC, targeting different types of surface antigens. They include epidermal growth factor receptor (EGFR), mesothelin (MSLN), prostate stem cell antigen (PSCA), and mucin 1 (MUC1). Potential new targets such as erythropoietin-producing hepatocellular carcinoma A2 (EphA2), tissue factor (TF), and protein tyrosine kinase 7 (PTK7) are currently under investigation in clinical trials. The challenges in developing CAR T for NSCLC therapy and other approaches for enhancing CAR T efficacy are discussed. Finally, we provide our perspective on imaging CAR T cell action by reviewing the two main radionuclide-based CAR T cell imaging techniques, the direct labeling of CAR T cells or indirect labeling via a reporter gene.
Collapse
Affiliation(s)
- Hong Yun Ma
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Jeeban Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Conor Prendergast
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | | | - Brian Braumuller
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Jacienta Paily
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Sophia Huang
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Connie Liou
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Anna Giarratana
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Mahdie Hosseini
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Randy Yeh
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Kathleen M. Capaccione
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| |
Collapse
|
11
|
Meng L, Yang Y, Mortazavi A, Zhang J. Emerging Immunotherapy Approaches for Treating Prostate Cancer. Int J Mol Sci 2023; 24:14347. [PMID: 37762648 PMCID: PMC10531627 DOI: 10.3390/ijms241814347] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Immunotherapy has emerged as an important approach for cancer treatment, but its clinical efficacy has been limited in prostate cancer compared to other malignancies. This review summarizes key immunotherapy strategies under evaluation for prostate cancer, including immune checkpoint inhibitors, bispecific T cell-engaging antibodies, chimeric antigen receptor (CAR) T cells, therapeutic vaccines, and cytokines. For each modality, the rationale stemming from preclinical studies is discussed along with outcomes from completed clinical trials and strategies to improve clinical efficacy that are being tested in ongoing clinical trials. Imperative endeavors include biomarker discovery for patient selection, deciphering resistance mechanisms, refining cellular therapies such as CAR T cells, and early-stage intervention were reviewed. These ongoing efforts instill optimism that immunotherapy may eventually deliver significant clinical benefits and expand treatment options for patients with advanced prostate cancer.
Collapse
Affiliation(s)
- Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (L.M.); (Y.Y.); (A.M.)
| | - Yuanquan Yang
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (L.M.); (Y.Y.); (A.M.)
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (L.M.); (Y.Y.); (A.M.)
| | - Jingsong Zhang
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center & Research Institute, University of South Florida, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
12
|
Porter LH, Zhu JJ, Lister NL, Harrison SG, Keerthikumar S, Goode DL, Urban RQ, Byrne DJ, Azad A, Vela I, Hofman MS, Neeson PJ, Darcy PK, Trapani JA, Taylor RA, Risbridger GP. Low-dose carboplatin modifies the tumor microenvironment to augment CAR T cell efficacy in human prostate cancer models. Nat Commun 2023; 14:5346. [PMID: 37660083 PMCID: PMC10475084 DOI: 10.1038/s41467-023-40852-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 08/11/2023] [Indexed: 09/04/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells have transformed the treatment landscape for hematological malignancies. However, CAR T cells are less efficient against solid tumors, largely due to poor infiltration resulting from the immunosuppressive nature of the tumor microenvironment (TME). Here, we assessed the efficacy of Lewis Y antigen (LeY)-specific CAR T cells in patient-derived xenograft (PDX) models of prostate cancer. In vitro, LeY CAR T cells directly killed organoids derived from androgen receptor (AR)-positive or AR-null PDXs. In vivo, although LeY CAR T cells alone did not reduce tumor growth, a single prior dose of carboplatin reduced tumor burden. Carboplatin had a pro-inflammatory effect on the TME that facilitated early and durable CAR T cell infiltration, including an altered cancer-associated fibroblast phenotype, enhanced extracellular matrix degradation and re-oriented M1 macrophage differentiation. In a PDX less sensitive to carboplatin, CAR T cell infiltration was dampened; however, a reduction in tumor burden was still observed with increased T cell activation. These findings indicate that carboplatin improves the efficacy of CAR T cell treatment, with the extent of the response dependent on changes induced within the TME.
Collapse
Affiliation(s)
- L H Porter
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
| | - J J Zhu
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - N L Lister
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
| | - S G Harrison
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Physiology, Monash University, Clayton, VIC, 3800, Australia
| | - S Keerthikumar
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - D L Goode
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - R Quezada Urban
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - D J Byrne
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - A Azad
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - I Vela
- Queensland Bladder Cancer Initiative, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, QLD, 4102, Australia
- Australian Prostate Cancer Research Center, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, QLD, 4102, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD, 4102, Australia
| | - M S Hofman
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - P J Neeson
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - P K Darcy
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - J A Trapani
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - R A Taylor
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Physiology, Monash University, Clayton, VIC, 3800, Australia.
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
| | - G P Risbridger
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia.
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
| |
Collapse
|
13
|
Bellone M, Mondino A. Leveraging the Tumor Immune Microenvironment To Overcome Genitourinary Cancers. Eur Urol Oncol 2023; 6:273-274. [PMID: 37147215 DOI: 10.1016/j.euo.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 05/07/2023]
Affiliation(s)
- Matteo Bellone
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Anna Mondino
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
14
|
Chen C, Liu X, Chang CY, Wang HY, Wang RF. The Interplay between T Cells and Cancer: The Basis of Immunotherapy. Genes (Basel) 2023; 14:genes14051008. [PMID: 37239368 DOI: 10.3390/genes14051008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Over the past decade, immunotherapy has emerged as one of the most promising approaches to cancer treatment. The use of immune checkpoint inhibitors has resulted in impressive and durable clinical responses in the treatment of various cancers. Additionally, immunotherapy utilizing chimeric antigen receptor (CAR)-engineered T cells has produced robust responses in blood cancers, and T cell receptor (TCR)-engineered T cells are showing promising results in the treatment of solid cancers. Despite these noteworthy advancements in cancer immunotherapy, numerous challenges remain. Some patient populations are unresponsive to immune checkpoint inhibitor therapy, and CAR T cell therapy has yet to show efficacy against solid cancers. In this review, we first discuss the significant role that T cells play in the body's defense against cancer. We then delve into the mechanisms behind the current challenges facing immunotherapy, starting with T cell exhaustion due to immune checkpoint upregulation and changes in the transcriptional and epigenetic landscapes of dysfunctional T cells. We then discuss cancer-cell-intrinsic characteristics, including molecular alterations in cancer cells and the immunosuppressive nature of the tumor microenvironment (TME), which collectively facilitate tumor cell proliferation, survival, metastasis, and immune evasion. Finally, we examine recent advancements in cancer immunotherapy, with a specific emphasis on T-cell-based treatments.
Collapse
Affiliation(s)
- Christina Chen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Xin Liu
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Che-Yu Chang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Helen Y Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Rong-Fu Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
15
|
Chimeric antigen receptor T cells therapy in solid tumors. Clin Transl Oncol 2023:10.1007/s12094-023-03122-8. [PMID: 36853399 DOI: 10.1007/s12094-023-03122-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/10/2023] [Indexed: 03/01/2023]
Abstract
Chimeric antigen receptor T cells therapy (CAR-T therapy) is a class of ACT therapy. Chimeric antigen receptor (CAR) is an engineered synthetic receptor of CAR-T, which give T cells the ability to recognize tumor antigens in a human leukocyte antigen-independent (HLA-independent) manner and enables them to recognize more extensive target antigens than natural T cell surface receptor (TCR), resulting in tumor destruction. CAR-T is composed of an extracellular single-chain variable fragment (scFv) of antibody, which serves as the targeting moiety, hinge region, transmembrane spacer, and intracellular signaling domain(s). CAR-T has been developing in many generations, which differ according to costimulatory domains. CAR-T therapy has several limitations that reduce its wide availability in immunotherapy which we can summarize in antigen escape that shows either partial or complete loss of target antigen expression, so multiplexing CAR-T cells are promoted to enhance targeting of tumor profiles. In addition, the large diversity in the tumor microenvironment also plays a major role in limiting this kind of treatment. Therefore, engineered CAR-T cells can evoke immunostimulatory signals that rebalance the tumor microenvironment. Using CAR-T therapy in treating the solid tumor is mainly restricted by the difficulty of CAR-T cells infiltrating the tumor site, so local administration was developed to improve the quality of treatment. The most severe toxicity after CAR-T therapy is on-target/on-tumor toxicity, such as cytokine release syndrome (CRS). Another type of toxicity is on-target/off-tumor toxicity which originates from the binding of CAR-T cells to target antigen that has shared expression on normal cells leading to damage in healthy cells and organs. Toxicity management should become a focus of implementation to permit management beyond specialized centers.
Collapse
|
16
|
Maalej KM, Merhi M, Inchakalody VP, Mestiri S, Alam M, Maccalli C, Cherif H, Uddin S, Steinhoff M, Marincola FM, Dermime S. CAR-cell therapy in the era of solid tumor treatment: current challenges and emerging therapeutic advances. Mol Cancer 2023; 22:20. [PMID: 36717905 PMCID: PMC9885707 DOI: 10.1186/s12943-023-01723-z] [Citation(s) in RCA: 198] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
In the last decade, Chimeric Antigen Receptor (CAR)-T cell therapy has emerged as a promising immunotherapeutic approach to fight cancers. This approach consists of genetically engineered immune cells expressing a surface receptor, called CAR, that specifically targets antigens expressed on the surface of tumor cells. In hematological malignancies like leukemias, myeloma, and non-Hodgkin B-cell lymphomas, adoptive CAR-T cell therapy has shown efficacy in treating chemotherapy refractory patients. However, the value of this therapy remains inconclusive in the context of solid tumors and is restrained by several obstacles including limited tumor trafficking and infiltration, the presence of an immunosuppressive tumor microenvironment, as well as adverse events associated with such therapy. Recently, CAR-Natural Killer (CAR-NK) and CAR-macrophages (CAR-M) were introduced as a complement/alternative to CAR-T cell therapy for solid tumors. CAR-NK cells could be a favorable substitute for CAR-T cells since they do not require HLA compatibility and have limited toxicity. Additionally, CAR-NK cells might be generated in large scale from several sources which would suggest them as promising off-the-shelf product. CAR-M immunotherapy with its capabilities of phagocytosis, tumor-antigen presentation, and broad tumor infiltration, is currently being investigated. Here, we discuss the emerging role of CAR-T, CAR-NK, and CAR-M cells in solid tumors. We also highlight the advantages and drawbacks of CAR-NK and CAR-M cells compared to CAR-T cells. Finally, we suggest prospective solutions such as potential combination therapies to enhance the efficacy of CAR-cells immunotherapy.
Collapse
Affiliation(s)
- Karama Makni Maalej
- grid.413548.f0000 0004 0571 546XTranslational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, P.O. Box: 3050, Doha, Qatar
| | - Maysaloun Merhi
- grid.413548.f0000 0004 0571 546XTranslational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, P.O. Box: 3050, Doha, Qatar
| | - Varghese P. Inchakalody
- grid.413548.f0000 0004 0571 546XTranslational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, P.O. Box: 3050, Doha, Qatar
| | - Sarra Mestiri
- grid.413548.f0000 0004 0571 546XTranslational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, P.O. Box: 3050, Doha, Qatar
| | - Majid Alam
- grid.413548.f0000 0004 0571 546XTranslational Research Institute, Academic Health System, Dermatology Institute, Hamad Medical Corporation, Doha, Qatar ,grid.413548.f0000 0004 0571 546XDepartment of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Cristina Maccalli
- grid.467063.00000 0004 0397 4222Laboratory of Immune and Biological Therapy, Research Department, Sidra Medicine, Doha, Qatar
| | - Honar Cherif
- grid.413548.f0000 0004 0571 546XDepartment of Hematology, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- grid.413548.f0000 0004 0571 546XTranslational Research Institute, Academic Health System, Dermatology Institute, Hamad Medical Corporation, Doha, Qatar
| | - Martin Steinhoff
- grid.413548.f0000 0004 0571 546XTranslational Research Institute, Academic Health System, Dermatology Institute, Hamad Medical Corporation, Doha, Qatar ,grid.413548.f0000 0004 0571 546XDepartment of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar ,grid.416973.e0000 0004 0582 4340Department of Dermatology, Weill Cornell Medicine-Qatar, Doha, Qatar ,grid.412603.20000 0004 0634 1084College of Medicine, Qatar University, Doha, Qatar ,grid.5386.8000000041936877XDepartment of Dermatology, Weill Cornell Medicine, New York, USA
| | - Francesco M. Marincola
- grid.418227.a0000 0004 0402 1634Global Head of Research, Kite Pharma, Santa Monica, California USA
| | - Said Dermime
- grid.413548.f0000 0004 0571 546XTranslational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, P.O. Box: 3050, Doha, Qatar ,grid.452146.00000 0004 1789 3191College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
17
|
Zarrabi KK, Narayan V, Mille PJ, Zibelman MR, Miron B, Bashir B, Kelly WK. Bispecific PSMA antibodies and CAR-T in metastatic castration-resistant prostate cancer. Ther Adv Urol 2023; 15:17562872231182219. [PMID: 37359737 PMCID: PMC10285603 DOI: 10.1177/17562872231182219] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Prostate cancer is the most common cancer among men and the second leading cause of cancer-related deaths in men in the United States. The treatment paradigm for prostate cancer has evolved with the emergence of a variety of novel therapies which have improved survival; however, treatment-related toxicities are abundant and durable responses remain rare. Immune checkpoint inhibitors have shown modest activity in a small subset of patients with prostate cancer and have not had an impact on most men with advanced disease. The discovery of prostate-specific membrane antigen (PSMA) and the understanding of its specificity to prostate cancer has identified it as an ideal tumor-associated antigen and has revived the enthusiasm for immunotherapeutics in prostate cancer. T-cell immunotherapy in the form of bispecific T-cell engagers (BiTEs) and chimeric antigen receptor (CAR) T-cell therapy have shown exceptional success in treating various hematologic malignancies, and are now being tested in patients with prostate cancer with drug design centered on various target ligands including not just PSMA, but others as well including six-transmembrane epithelial antigen of the prostate 1 (STEAP1) and prostate stem cell antigen (PSCA). This summative review will focus on the data surrounding PSMA-targeting T-cell therapies. Early clinical studies with both classes of T-cell redirecting therapies have demonstrated antitumor activity; however, there are multiple challenges with this class of agents, including dose-limiting toxicity, 'on-target, off-tumor' immune-related toxicity, and difficulty in maintaining sustained immune responses within a complex and overtly immunosuppressive tumor microenvironment. Reflecting on experiences from recent trials has been key toward understanding mechanisms of immune escape and limitations in developing these drugs in prostate cancer. Newer generation BiTE and CAR T-cell constructs, either alone or as part of combination therapy, are currently under investigation with modifications in drug design to overcome these barriers. Ongoing innovation in drug development will likely foster successful implementation of T-cell immunotherapy bringing transformational change to the treatment of prostate cancer.
Collapse
Affiliation(s)
| | - Vivek Narayan
- Department of Medical Oncology, Abramson Cancer Center and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick J. Mille
- Department of Medical Oncology and Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Matthew R. Zibelman
- Department of Medical Oncology, Fox Chase Cancer Center, Temple University, Philadelphia, PA, USA
| | - Benjamin Miron
- Department of Medical Oncology, Fox Chase Cancer Center, Temple University, Philadelphia, PA, USA
| | - Babar Bashir
- Department of Medical Oncology and Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, USA
- Department of Pharmacology, Physiology, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - William Kevin Kelly
- Department of Medical Oncology and Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| |
Collapse
|
18
|
Steinbach C, Merchant A, Zaharie AT, Horak P, Marhold M, Krainer M. Current Developments in Cellular Therapy for Castration Resistant Prostate Cancer: A Systematic Review of Clinical Studies. Cancers (Basel) 2022; 14:5719. [PMID: 36428811 PMCID: PMC9688882 DOI: 10.3390/cancers14225719] [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: 09/14/2022] [Accepted: 10/21/2022] [Indexed: 11/23/2022] Open
Abstract
Recently, the development of immunotherapies such as cellular therapy, monoclonal antibodies, vaccines and immunomodulators has revolutionized the treatment of various cancer entities. In order to close the existing gaps in knowledge about cellular immunotherapy, specifically focusing on the chimeric antigen receptors (CAR) T-cells, their benefits and application in clinical settings, we conducted a comprehensive systematic review. Two co-authors independently searched the literature and characterized the results. Out of 183 records, 26 were considered eligible. This review provides an overview of the cellular immunotherapy landscape in treating prostate cancer, honing in on the challenges of employing CAR T-cell therapy. CAR T-cell therapy is a promising avenue for research due to the presence of an array of different tumor specific antigens. In prostate cancer, the complex microenvironment of the tumor vastly contributes to the success or failure of immunotherapies.
Collapse
Affiliation(s)
- Christina Steinbach
- Internal Medicine I, Department of Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Almas Merchant
- Internal Medicine I, Department of Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Peter Horak
- Nationales Centrum für Tumorerkrankungen (NCT), 69120 Heidelberg, Germany
| | - Maximilian Marhold
- Internal Medicine I, Department of Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Krainer
- Internal Medicine I, Department of Oncology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
19
|
Zhang Q, Zhang Z, Liu G, Li D, Gu Z, Zhang L, Pan Y, Cui X, Wang L, Liu G, Tian X, Zhang Z. B7-H3 targeted CAR-T cells show highly efficient anti-tumor function against osteosarcoma both in vitro and in vivo. BMC Cancer 2022; 22:1124. [PMID: 36320072 PMCID: PMC9628043 DOI: 10.1186/s12885-022-10229-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/25/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Osteosarcoma (OS) mainly happens in children and youths. Surgery, radiotherapy and chemotherapy are the common therapies for osteosarcoma treatment but all their anti-tumor effects are limited. In recent years, a new cellular therapy, CAR-T, a cellular immunotherapy with genetically engineered T cells bearing chimeric antigen receptor targeting specific tumor-associated antigen, has been proved to be an effective therapy against acute lymphoblastic leukemia. Thus, CAR-T is a potentially effective therapy for osteosarcoma treatment. METHODS A CAR gene targeting B7-H3 antigen was constructed into lentiviral vector through molecular biology techniques. Then, the CAR gene was transferred to T cells through lentiviral delivery system, and the CAR-T cells were largely expanded using in vitro culture technology. The in vitro anti-tumor effect of CAR-T cells was evaluated through Real Time Cell Analysis system (RTCA) and ELISA assay. The in vivo anti-tumor capabilities of CAR-T cells were evaluated using the patient-derived xenografts (PDX) model of osteosarcoma. RESULTS The third-generation CAR-T cells we constructed could target the B7-H3 antigen, and the phenotype of CAR-T cells was consistent with normal T cells; The CAR-T cells showed superior antitumor effects both in vitro and in vivo. CONCLUSION Our study showed that B7-H3 targeted CAR-T cells had high anti-tumor efficacy against osteosarcoma both in vitro and in vivo, which proved that B7-H3 targeted CAR-T therapy is potentially effective for osteosarcoma treatment.
Collapse
Affiliation(s)
- Qian Zhang
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231 China
| | - Zhiqiang Zhang
- grid.411333.70000 0004 0407 2968Department of Pediatric Orthopedics, National Children’s Medical Center & Children’s Hospital of Fudan University, Shanghai, 201102 China
| | - Guodi Liu
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231 China ,grid.28056.390000 0001 2163 4895State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237 China
| | - Dehua Li
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231 China
| | - Zhangjie Gu
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231 China
| | - Linsong Zhang
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231 China
| | - Yingjiao Pan
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231 China
| | - Xingbing Cui
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231 China
| | - Lu Wang
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231 China
| | - Guoping Liu
- grid.411525.60000 0004 0369 1599Department of General Surgery, Changhai Hospital, Shanghai, 200433 China
| | - Xiaoli Tian
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231 China ,Shanghai Beautiful Life Medical Technology Co., Ltd., Shanghai, 200231 China
| | - Ziming Zhang
- grid.412987.10000 0004 0630 1330Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092 China ,grid.415625.10000 0004 0467 3069Department of Orthopaedics, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China
| |
Collapse
|
20
|
Liu C, Wang M, Zhang H, Li C, Zhang T, Liu H, Zhu S, Chen J. Tumor microenvironment and immunotherapy of oral cancer. Eur J Med Res 2022; 27:198. [PMID: 36209263 PMCID: PMC9547678 DOI: 10.1186/s40001-022-00835-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022] Open
Abstract
Oral cancer is one of the most common malignant tumors of the head and neck, not only affects the appearance, but also affects eating and even endangers life. The clinical treatments of oral cancer mainly include surgery, radiotherapy, and chemotherapy. However, unsatisfactory therapeutic effect and toxic side effects are still the main problems in clinical treatment. Tumor microenvironment (TME) is not only closely related to the occurrence, growth, and metastasis of tumor but also works in the diagnosis, prevention, and treatment of tumor and prognosis. Future studies should continue to investigate the relationship of TME and oral cancer therapy. This purpose of this review was to analyze the characteristics of oral cancer microenvironment, summarize the traditional oral cancer therapy and immunotherapy strategies, and finally prospect the development prospects of oral cancer immunotherapy. Immunotherapy targeting tumor microenvironment is expected to provide a new strategy for clinical treatment of oral cancer.
Collapse
Affiliation(s)
- Chang Liu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Min Wang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Haiyang Zhang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Chunyan Li
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Tianshou Zhang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Hong Liu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Song Zhu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China.
| | - Jie Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People's Republic of China.
| |
Collapse
|
21
|
Zhang X, Sun S, Miao Y, Yuan Y, Zhao W, Li H, Wei X, Huang C, Hu X, Wang B, Xu H, Zhang W, Gao X, Song J, Zheng J, Zhang Q. Docetaxel enhances the therapeutic efficacy of PSMA-specific CAR-T cells against prostate cancer models by suppressing MDSCs. J Cancer Res Clin Oncol 2022; 148:3511-3520. [PMID: 35962287 DOI: 10.1007/s00432-022-04248-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 08/01/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE Prostate cancer can undergo curative effects by radical prostatectomy or radical radiotherapy. However, the best treatment for more aggressive high-risk prostate cancer remains controversial. Insufficient infiltration capacity and dysfunction are commonly occurrences in engineered T lymphocytes expressing chimeric antigen receptor (CAR-T), characterizing cancer immunotherapy failure. We conducted this study to investigate whether the combinative application of docetaxel and PSMA-CAR-T cells could be a more effective treatment to prostate cancer. METHODS Expressions of prostate specific membrane antigen (PSMA) on prostate cancer cells were examined by Flow cytometry. The efficaciousness of PSMA-CAR-T was evaluated in vitro using ELISA and RTCA. The effect of intermixed therapy was assessed in vivo utilizing a human prostate cancer liver metastasis mouse model and a human prostate cancer cell xenograft mouse model. RESULTS The outcome of cytokine discharge and cell killing assays demonstrated that PSMA-CAR-T cells have characteristic effector capacity against PSMA+ prostate cancer cells in vitro. Additionally, collaborative treatment of PSMA-CAR-T cells and docetaxel have cooperative efficacy in a mouse model of human prostate cancer. The merged strategy could be seen as an undeveloped avenue to augmenting adoptive CAR-T cell immunotherapy and mitigating the adverse side effects of chemotherapy. CONCLUSIONS Cooperation of PSMA-specific CAR-T cells and the chemotherapy drug docetaxel can impressively ameliorate antitumor effectiveness against an installed metastatic human prostate cancer model in NPG mice.
Collapse
Affiliation(s)
- Xiaokang Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Shishuo Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Yangna Miao
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Yifan Yuan
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Wanxin Zhao
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Hailong Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Xiaohuan Wei
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Chao Huang
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Xiaolei Hu
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Bixi Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Heng Xu
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Wei Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Xiaoge Gao
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Jingyuan Song
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.,School of Nursing, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.
| | - Qing Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China. .,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
22
|
Di Sarno V, Giovannelli P, Medina-Peris A, Ciaglia T, Di Donato M, Musella S, Lauro G, Vestuto V, Smaldone G, Di Matteo F, Bifulco G, Castoria G, Migliaccio A, Fernandez-Carvajal A, Campiglia P, Gomez-Monterrey I, Ostacolo C, Bertamino A. New TRPM8 blockers exert anticancer activity over castration-resistant prostate cancer models. Eur J Med Chem 2022; 238:114435. [DOI: 10.1016/j.ejmech.2022.114435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 11/04/2022]
|
23
|
Perera MP, Thomas PB, Risbridger GP, Taylor R, Azad A, Hofman MS, Williams ED, Vela I. Chimeric Antigen Receptor T-Cell Therapy in Metastatic Castrate-Resistant Prostate Cancer. Cancers (Basel) 2022; 14:cancers14030503. [PMID: 35158771 PMCID: PMC8833489 DOI: 10.3390/cancers14030503] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Prostate cancer is one of the most frequently diagnosed cancers amongst men worldwide. Treatment for metastatic disease is often in the form of androgen deprivation therapy. However, over the course of treatment affected men may become castrate-resistant. Options for men with metastatic castrate-resistant cancer are limited. This review focuses on the role of chimeric antigen receptor T-cell therapy (CAR-T) in men with metastatic castrate-resistant prostate cancer. This review is a contemporary appraisal of preclinical and clinical studies conducted in this emerging form of immunotherapy. A thorough evaluation of the role of CAR-T therapy in prostate cancer is provided, as well as the obstacles we must overcome to clinically translate this therapy for men affected with this rapidly fatal disease. Abstract Prostate cancer is the most commonly diagnosed solid-organ cancer amongst males worldwide. Metastatic castrate-resistant prostate cancer (mCRPC) is a rapidly fatal end-sequelae of prostate cancer. Therapeutic options for men with mCRPC are limited and are not curative in nature. The recent development of chimeric antigen receptor T-cell (CAR-T) therapy has revolutionised the treatment of treatment-resistant haematological malignancies, and several studies are underway investigating the utility of this technology in the treatment of solid tumours. In this review, we evaluate the current treatment options for men with mCRPC as well as the current landscape of preclinical and clinical trials of CAR-T cell therapy against prostate cancer. We also appraise the various prostate cancer-specific tumour-associated antigens that may be targeted by CAR-T cell technology. Finally, we examine the potential translational barriers of CAR-T cell therapy in solid tumours. Despite preclinical success, preliminary clinical trials in men with prostate cancer have had limited efficacy. Therefore, further clinically translatable preclinical models are required to enhance the understanding of the role of this investigational therapeutic in men with mCRPC. In the era of precision medicine, tailored immunotherapy administered to men in a tumour-agnostic approach provides hope to a group of men who otherwise have few treatment options available.
Collapse
Affiliation(s)
- Mahasha P.J. Perera
- School of Biomedical Sciences at Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.); (E.D.W.)
- Queensland Bladder Cancer Initiative (QBCI), Woolloongabba, QLD 4102, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD 4102, Australia
- Correspondence: (M.P.P.); (I.V.)
| | - Patrick B. Thomas
- School of Biomedical Sciences at Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.); (E.D.W.)
- Queensland Bladder Cancer Initiative (QBCI), Woolloongabba, QLD 4102, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD 4102, Australia
| | - Gail P. Risbridger
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3168, Australia; (G.P.R.); (R.T.)
| | - Renea Taylor
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3168, Australia; (G.P.R.); (R.T.)
| | - Arun Azad
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (A.A.); (M.S.H.)
| | - Michael S. Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (A.A.); (M.S.H.)
| | - Elizabeth D. Williams
- School of Biomedical Sciences at Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.); (E.D.W.)
- Queensland Bladder Cancer Initiative (QBCI), Woolloongabba, QLD 4102, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD 4102, Australia
| | - Ian Vela
- School of Biomedical Sciences at Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD 4102, Australia; (P.B.T.); (E.D.W.)
- Queensland Bladder Cancer Initiative (QBCI), Woolloongabba, QLD 4102, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD 4102, Australia
- Correspondence: (M.P.P.); (I.V.)
| |
Collapse
|
24
|
Network models of prostate cancer immune microenvironments identify ROMO1 as heterogeneity and prognostic marker. Sci Rep 2022; 12:192. [PMID: 34996995 PMCID: PMC8741951 DOI: 10.1038/s41598-021-03946-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/07/2021] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) is the fifth leading cause of death from cancer in men worldwide. Its treatment remains challenging due to the heterogeneity of the tumor, mainly because of the lack of effective and targeted prognostic markers at the system biology level. First, the data were retrieved from TCGA dataset, and valid samples were obtained by consistent clustering and principal component analysis; next, key genes were analyzed for prognosis of PCa using WGCNA, MEGENA, and LASSO Cox regression model analysis, while key genes were screened based on disease-free survival significance. Finally, TIMER data were selected to explore the relationship between genes and tumor immune infiltration, and GSCAlite was used to explore the small-molecule targeted drugs that act with them. Here, we used tumor subtype analysis and an energetic co-expression network algorithm of WGCNA and MEGENA to identify a signal dominated by the ROMO1 to predict PCa prognosis. Cox regression analysis of ROMO1 was an independent influence, and the prognostic value of this biomarker was validated in the training set, the validated data itself, and external data, respectively. This biomarker correlates with tumor immune infiltration and has a high degree of infiltration, poor prognosis, and strong correlation with CD8+T cells. Gene function annotation and other analyses also implied a potential molecular mechanism for ROMO1. In conclusion, we putative ROMO1 as a portal key prognostic gene for the diagnosis and prognosis of PCa, which provides new insights into the diagnosis and treatment of PCa.
Collapse
|
25
|
Tan H, Xie Y, Zhang X, Wu S, Zhao H, Wu J, Wang W, Lin C. Integrative Analysis of MALT1 as a Potential Therapeutic Target for Prostate Cancer and its Immunological Role in Pan-Cancer. Front Mol Biosci 2021; 8:714906. [PMID: 34926571 PMCID: PMC8674617 DOI: 10.3389/fmolb.2021.714906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Mucosa-associated lymphoma antigen 1 (MALT1) is an oncogene in subsets of diffuse large B cell lymphoma (DLBCL) and mucosa-associated lymphoid tissue type (MALT) lymphoma. However, the role of MALT1 across cancers, especially in prostate cancer is still poorly understood. Methods: Here, we used several public datasets to evaluate MALT1 expression. Then, PCa cell lines and nude mice were used to investigate the cellular functions in vitro and in vivo. Microarray data were downloaded from The Cancer Genome Atlas and MALT1 was subjected to gene set enrichment analysis (GSEA) and Gene Ontology (GO) analysis to identify the biological functions and relevant pathways. Additionally, the correlations between MALT1 expression and mismatch repair (MMR) gene mutation, immune checkpoint gene expression, tumor mutational burden (TMB), and microsatellite instability (MSI) were investigated by Pearson correlation analysis. Moreover, the correlation between MALT1 expression and tumor immune infiltration was analyzed by the Tumor Immune Evaluation Resource (TIMER) database. Results: MALT1 overexpression was significantly correlated with MMR gene mutation levels and crucially promoted proliferation and colony genesis while reducing PCa cell apoptosis levels in vivo and in vitro. MALT1 expression showed strong correlations with immune checkpoint genes, TMB, and MSI in most cancers. The GO analysis indicated that MALT1-coexpressed genes were involved in heterotypic cell-cell adhesion, actin filament-based movement regulation, and action potential regulation. GSEA revealed that MALT1 expression was associated with several signaling pathways, including the NF-κB signaling, Wnt/β-catenin and TGF-β signaling pathways, in PCa. Additionally, MALT1 expression was significantly correlated with the infiltration of immune cells, including B cells, CD8+ T cells, dendritic cells and macrophages, and negatively correlated with CD4+ cell infiltration in PCa. Conclusion: MALT1 expression is higher in pancancer samples than in normal tissues. MALT1 promoted proliferation and colony genesis while reducing PCa cell apoptosis levels, and MALT1 suppression could inhibit xenograft tumor establishment in nude mice. Furthermore, MALT1 expression is closely related to the occurrence and development of multiple tumors in multiple ways. Therefore, MALT1 may be an emerging therapeutic target for a variety of cancers especially PCa.
Collapse
Affiliation(s)
- Haotian Tan
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yaqi Xie
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Binzhou Medical University, Yantai, China
| | - Xuebao Zhang
- Department of Reproductive Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Shuang Wu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Hongwei Zhao
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jitao Wu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Wenting Wang
- Central Laboratory, Yantai Yuhuangding Hospital, Yantai, China
| | - Chunhua Lin
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| |
Collapse
|
26
|
Abstract
Chimeric antigen receptor (CAR) T cell immunotherapy involves the genetic modification of the patient's own T cells so that they specifically recognize and destroy tumour cells. Considerable clinical success has been achieved using this technique in patients with lymphoid malignancies, but clinical studies that investigated treating solid tumours using this emerging technology have been disappointing. A number of developments might be able to increase the efficacy of CAR T cell therapy for treatment of prostate cancer, including improved trafficking to the tumour, techniques to overcome the immunosuppressive tumour microenvironment, as well as methods to enhance CAR T cell persistence, specificity and safety. Furthermore, CAR T cell therapy has the potential to be combined with other treatment modalities, such as androgen deprivation therapy, radiotherapy or chemotherapy, and could be applied as focal CAR T cell therapy for prostate cancer.
Collapse
|
27
|
Cheng G, Dong H, Yang C, Liu Y, Wu Y, Zhu L, Tong X, Wang S. A review on the advances and challenges of immunotherapy for head and neck cancer. Cancer Cell Int 2021; 21:406. [PMID: 34332576 PMCID: PMC8325213 DOI: 10.1186/s12935-021-02024-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
Head and neck cancer (HNC), which includes lip and oral cavity, larynx, nasopharynx, oropharynx, and hypopharynx malignancies, is one of the most common cancers worldwide. Due to the interaction of tumor cells with immune cells in the tumor microenvironment, immunotherapy of HNCs, along with traditional treatments such as chemotherapy, radiotherapy, and surgery, has attracted much attention. Four main immunotherapy strategies in HNCs have been developed, including oncolytic viruses, monoclonal antibodies, chimeric antigen receptor T cells (CAR-T cells), and therapeutic vaccines. Oncorine (H101), an approved oncolytic adenovirus in China, is the pioneer of immunotherapy for the treatment of HNCs. Pembrolizumab and nivolumab are mAbs against PD-L1 that have been approved for recurrent and metastatic HNC patients. To date, several clinical trials using immunotherapy agents and their combination are under investigation. In this review, we summarize current the interaction of tumor cells with immune cells in the tumor microenvironment of HNCs, the main strategies that have been applied for immunotherapy of HNCs, obstacles that hinder the success of immunotherapies in patients with HNCs, as well as solutions for overcoming the challenges to enhance the response of HNCs to immunotherapies.
Collapse
Affiliation(s)
- Gang Cheng
- Department of Stomatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China.,Department of Stomatology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, 233030, China
| | - Hui Dong
- Department of Stomatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China.,Department of Stomatology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, 233030, China
| | - Chen Yang
- Department of Ultrasonography, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Yang Liu
- Department of Ultrasonography, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Yi Wu
- Phase I Clinical Research Center, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Lifen Zhu
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, 310014, Zhejiang, People's Republic of China.,Molecular Diagnosis Laboratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiangmin Tong
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, 310014, Zhejiang, People's Republic of China.,Molecular Diagnosis Laboratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Shibing Wang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, 310014, Zhejiang, People's Republic of China. .,Molecular Diagnosis Laboratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
28
|
Adenocarcinoma of the Prostate: Future Directions for Translational Science. Prostate Cancer 2021. [DOI: 10.36255/exonpublications.prostatecancer.translationalscience.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] Open
|
29
|
Venkatas J, Singh M. Nanomedicine-mediated optimization of immunotherapeutic approaches in cervical cancer. Nanomedicine (Lond) 2021; 16:1311-1328. [PMID: 34027672 DOI: 10.2217/nnm-2021-0044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cervical cancer shows immense complexity at the epigenetic, genetic and cellular levels, limiting conventional treatment. Immunotherapy has revolutionized nanomedicine and rejuvenated the field of tumor immunology. Although several immunotherapeutic approaches have shown favorable clinical responses, their efficacies vary, with subsets of patients benefitting. The success of cancer immunotherapy requires the enhancement of cytokines and antitumor effector cell production and activation. Recently, the feasibility of nanoparticle-based cytokine approaches in tumor immunotherapy has been highlighted. Immunotherapeutic nanoparticle-based platforms form a novel strategy enabling researchers to co-deliver immunomodulatory agents, target tumors, improve pharmacokinetics and minimize collateral toxicity to healthy cells. This review looks at the potential of immunotherapy and nanotechnologically enhanced immunotherapeutic approaches for cervical cancer.
Collapse
Affiliation(s)
- Jeaneen Venkatas
- Nano-Gene & Drug Delivery Group, Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, 4000, KwaZulu-Natal, South Africa
| | - Moganavelli Singh
- Nano-Gene & Drug Delivery Group, Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, 4000, KwaZulu-Natal, South Africa
| |
Collapse
|
30
|
Miao L, Zhang Z, Ren Z, Tang F, Li Y. Obstacles and Coping Strategies of CAR-T Cell Immunotherapy in Solid Tumors. Front Immunol 2021; 12:687822. [PMID: 34093592 PMCID: PMC8170155 DOI: 10.3389/fimmu.2021.687822] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/04/2021] [Indexed: 12/31/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell immunotherapy refers to an adoptive immunotherapy that has rapidly developed in recent years. It is a novel type of treatment that enables T cells to express specific CARs on their surface, then returns these T cells to tumor patients to kill the corresponding tumor cells. Significant strides in CAR-T cell immunotherapy against hematologic malignancies have elicited research interest among scholars in the treatment of solid tumors. Nonetheless, in contrast with the efficacy of CAR-T cell immunotherapy in the treatment of hematologic malignancies, its general efficacy against solid tumors is insignificant. This has been attributed to the complex biological characteristics of solid tumors. CAR-T cells play a better role in solid tumors, for instance by addressing obstacles including the lack of specific targets, inhibition of tumor microenvironment (TME), homing barriers of CAR-T cells, differentiation and depletion of CAR-T cells, inhibition of immune checkpoints, trogocytosis of CAR-T cells, tumor antigen heterogeneity, etc. This paper reviews the obstacles influencing the efficacy of CAR-T cell immunotherapy in solid tumors, their mechanism, and coping strategies, as well as economic restriction of CAR-T cell immunotherapy and its solutions. It aims to provide some references for researchers to better overcome the obstacles that affect the efficacy of CAR-T cells in solid tumors.
Collapse
Affiliation(s)
- Lele Miao
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhengchao Zhang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhijian Ren
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Futian Tang
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
31
|
Lei X, Ou Z, Yang Z, Zhong J, Zhu Y, Tian J, Wu J, Deng H, Lin X, Peng Y, Li B, He L, Tu Z, Chen W, Li Q, Liu N, Zhang H, Wang Z, Fang Z, Yamada T, Lv X, Tian T, Pan G, Wu F, Xiao L, Zhang L, Cai T, Wang X, Tannous BA, Li J, Kontos F, Ferrone S, Fan S. A Pan-Histone Deacetylase Inhibitor Enhances the Antitumor Activity of B7-H3-Specific CAR T Cells in Solid Tumors. Clin Cancer Res 2021; 27:3757-3771. [PMID: 33811153 DOI: 10.1158/1078-0432.ccr-20-2487] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/03/2020] [Accepted: 03/29/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE The limited efficacy of chimeric antigen receptor (CAR) T-cell therapies with solid malignancies prompted us to test whether epigenetic therapy could enhance the antitumor activity of B7-H3.CAR T cells with several solid cancer types. EXPERIMENTAL DESIGN We evaluated B7-H3 expression in many human solid cancer and normal tissue samples. The efficacy of the combinatorial therapy with B7-H3.CAR T cells and the deacetylase inhibitor SAHA with several solid cancer types and the potential underlying mechanisms were characterized with in vitro and ex vivo experiments. RESULTS B7-H3 is expressed in most of the human solid tumor samples tested, but exhibits a restricted expression in normal tissues. B7-H3.CAR T cells selectively killed B7-H3 expressing human cancer cell lines in vitro. A low dose of SAHA upregulated B7-H3 expression in several types of solid cancer cells at the transcriptional level and B7-H3.CAR expression on human transgenic T-cell membrane. In contrast, the expression of immunosuppressive molecules, such as CTLA-4 and TET2, by T cells was downregulated upon SAHA treatment. A low dose of SAHA significantly enhanced the antitumor activity of B7-H3.CAR T cells with solid cancers in vitro and ex vivo, including orthotopic patient-derived xenograft and metastatic models treated with autologous CAR T-cell infusions. CONCLUSIONS Our results show that our novel strategy which combines SAHA and B7-H3.CAR T cells enhances their therapeutic efficacy with solid cancers and justify its translation to a clinical setting.
Collapse
Affiliation(s)
- Xinyuan Lei
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China.,State University of New York at Stony Brook, Stony Brook, New York
| | - Zhanpeng Ou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Zhaohui Yang
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianglong Zhong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Yanliang Zhu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Jing Tian
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiannan Wu
- Department of Breast Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Heran Deng
- Department of Breast Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinyu Lin
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Peng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Bowen Li
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lile He
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Zhiming Tu
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Weixiong Chen
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qunxing Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Niu Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Hanqing Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Zhangsong Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Zezhen Fang
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Teppei Yamada
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Xiaobin Lv
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, Center Laboratory, the Third Affiliated Hospital, Nanchang University, Nanchang, China
| | - Tian Tian
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guokai Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Fan Wu
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liping Xiao
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lizao Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Tingting Cai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Xinhui Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Lab, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jinsong Li
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Filippos Kontos
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Song Fan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China. .,Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
32
|
Zhang Y, He L, Sadagopan A, Ma T, Dotti G, Wang Y, Zheng H, Gao X, Wang D, DeLeo AB, Fan S, Sun R, Yu L, Zhang L, Wang G, Ferrone S, Wang X. Targeting Radiation-Resistant Prostate Cancer Stem Cells by B7-H3 CAR T Cells. Mol Cancer Ther 2021; 20:577-588. [PMID: 33653946 PMCID: PMC7952034 DOI: 10.1158/1535-7163.mct-20-0446] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 11/13/2020] [Accepted: 01/04/2021] [Indexed: 01/16/2023]
Abstract
Radiotherapy (RT) is a key treatment for prostate cancer. However, RT resistance can contribute to treatment failure. Prostate cancer stem cells (PCSCs) are radioresistant. We recently found that fractionated irradiation (FIR) upregulates expression of the immune checkpoint B7-H3 (CD276) on PCSCs and bulk cells in each prostate cancer cell line tested. These findings prompted us to investigate whether B7-H3 targeting chimeric antigen receptor (CAR) T cells, which may abrogate function of an immune checkpoint and mediate lysis of targeted cells, can target RT-resistant PCSCs in vitro and in vivo. B7-H3 expression is naturally higher on PCSCs than bulk prostate cancer cells and cytotoxicity of B7-H3 CAR T cells to PCSCs is more potent than to bulk prostate cancer cells. Furthermore, FIR significantly upregulates B7-H3 expression on PCSCs and bulk prostate cancer cells. The duration of FIR or single-dose irradiation-induced further upregulation of B7-H3 on bulk prostate cancer cells and PCSCs lasts for up to 3 days. B7-H3 CAR T-cell cytotoxicity against FIR-resistant PCSCs at a low effector to target ratio of 1:1 was assessed by flow cytometry and sphere formation assays. Further upregulation of B7-H3 expression by FIR made PCSCs even more sensitive to B7-H3 CAR T-cell-mediated killing. Consequently, the FIR and B7-H3 CAR T-cell therapy combination is much more effective than FIR or CAR T cells alone in growth inhibition of hormone-insensitive prostate cancer xenografts in immunodeficient mice. Our work provides a sound basis for further development of this unique combinatorial model of RT and B7-H3 CAR T-cell therapy for prostate cancer. SIGNIFICANCE: We demonstrate that FIR significantly upregulates B7-H3 expression by RT-resistant PCSCs and bulk cells; cytotoxicity of B7-H3 CAR T cells to FIR-treated PCSCs is potent and results in significantly improved antitumor efficacy in mice.
Collapse
Affiliation(s)
- Yida Zhang
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Lile He
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ananthan Sadagopan
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tao Ma
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina
| | - Yufeng Wang
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hui Zheng
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xin Gao
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dian Wang
- Department of Radiation Oncology, Rush University Medical Center, Chicago, Illinois
| | - Albert B DeLeo
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Song Fan
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ruochuan Sun
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ling Yu
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Liyuan Zhang
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gongxian Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Soldano Ferrone
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xinhui Wang
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
33
|
Wong RL, Yu EY. Refining Immuno-Oncology Approaches in Metastatic Prostate Cancer: Transcending Current Limitations. Curr Treat Options Oncol 2021; 22:13. [PMID: 33433743 DOI: 10.1007/s11864-020-00808-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 02/07/2023]
Abstract
OPINION STATEMENT Due to its immunosuppressive tumor microenvironment, prostate cancer has historically been difficult to treat with immuno-oncology approaches. Other than pembrolizumab, which is now regulatory-approved for all microsatellite instability (MSI)-high and tumor mutational burden (TMB)-high advanced solid tumors, sipuleucel-T is the only immunotherapeutic agent approved by the US Food and Drug Administration (FDA) for prostate cancer. However, sipuleucel-T efficacy is optimal for select patients with indolent metastatic castration-resistant prostate cancer. Although manipulation of immune regulation by blocking immune checkpoints has led to substantial benefit in many cancers, experience with single-agent CTLA-4 and PD-1 or PD-L1 antibodies has shown limited effect for the majority of patients with prostate cancer, especially when administered as monotherapy. Combination therapies are now being attempted, in addition to enrichment strategies employing patient clinicopathologic and biologic characteristics that may heighten responses to immuno-oncology treatment, such as PD-L1 expression, TMB, MSI status, and alterations in CDK12. More work is needed to overcome the immune-exclusive barriers in prostate cancer, such as relatively low TMB, increased activity of myeloid-derived suppressor cells (MDSCs) and regulatory T cells, and defects in major histocompatibility complex (MHC) class I expression and interferon (IFN)-1 signaling. A promising approach and the likely next step in immuno-oncology for prostate cancer involves forced direction to markers expressed by prostate cancer tumor cells, such as prostate-specific membrane antigen (PSMA), that bypass the typical requirements for MHC class I interaction. The future will incorporate bispecific antibodies and chimeric antigen receptor (CAR)-T cells, potentially targeted towards phenotypic markers identified by next-generation PET imaging as part of the next wave of "precision medicine" in prostate cancer. Ultimately, we believe that the immune-exclusive prostate cancer tumor microenvironment can be overcome, and that patient outcomes can be enhanced through these more refined immuno-oncology approaches.
Collapse
Affiliation(s)
- Risa L Wong
- Division of Oncology, Department of Medicine, University of Washington, 825 Eastlake Ave E, G4830, Seattle, WA, 98109-1023, USA. .,Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D5-100, Seattle, WA, 98109-1024, USA.
| | - Evan Y Yu
- Division of Oncology, Department of Medicine, University of Washington, 825 Eastlake Ave E, G4830, Seattle, WA, 98109-1023, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D5-100, Seattle, WA, 98109-1024, USA
| |
Collapse
|
34
|
Bansal R, Reshef R. Revving the CAR - Combination strategies to enhance CAR T cell effectiveness. Blood Rev 2021; 45:100695. [PMID: 32402724 DOI: 10.1016/j.blre.2020.100695] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/22/2020] [Accepted: 04/11/2020] [Indexed: 12/14/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy is currently approved for treatment of refractory B-cell malignancies. Response rates in these diseases are impressive by historical standards, but most patients do not have a durable response and there remains room for improvement. To date, CAR T cell activity has been even more limited in solid malignancies. These limitations are thought to be due to several pathways of resistance to CAR T cells, including cell-intrinsic mechanisms and the immunosuppressive tumor microenvironment. In this review, we discuss current experimental strategies that combine small molecules and monoclonal antibodies with CAR T cells to overcome these resistance mechanisms. We describe the biological rationale, pre-clinical data and clinical trials in progress that test the efficacy and safety of these combinations.
Collapse
Affiliation(s)
- Rajat Bansal
- Division of Hematology/Oncology, Columbia University Irving Medical Center, 177 Ft. Washington Ave, Floor: 6GN-435, New York, NY 10032, USA.
| | - Ran Reshef
- Division of Hematology/Oncology, Columbia University Irving Medical Center, 630 W. 168(th) Street Mailbox 127, New York, NY 10032, USA.
| |
Collapse
|
35
|
Abstract
Cancer gene therapy emerged as a promising treatment modality 3 decades ago. However, the failure of the first gene therapy trials in cancer treatment has decreased its popularity. Likewise, immunotherapy has followed a similar course. While it was a popular and promising treatment with IL-2 and interferon and cancer vaccines in the 1980s, it later lost its popularity. Immunotherapy became one of the main options for cancer treatment with the successful use of immune checkpoint inhibitors in clinics approximately 10 years ago. The success of immunotherapy has increased even more with the introduction of cancer gene therapy methods in this area. With the identification of the oncolytic herpes simplex virus and Chimeric antigen receptor (CAR) T-cells, immune gene therapy has become an essential modality in cancer treatments such as surgery, radiotherapy, chemotherapy, and targeted therapies.
Collapse
Affiliation(s)
- Hakan Akbulut
- Department of Basic Oncology, Ankara University Cancer Research Institute, Ankara, Turkey,Department of Medical Oncology, Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
36
|
Shuvalova ML, Kopylov AT, Mazurov DV, Pichugin AV, Bovin NV, Filatov AV. CD44-Associated Tn Antigen as a New Biomarker of Tumor Cells with Aberrant Glycosylation. BIOCHEMISTRY (MOSCOW) 2020; 85:1064-1081. [DOI: 10.1134/s0006297920090060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
37
|
Shang X, Li J, Wang H, Li Z, Lin J, Chen D, Wang H. CMTM6 is positively correlated with PD-L1 expression and immune cells infiltration in lung squamous carcinoma. Int Immunopharmacol 2020; 88:106864. [PMID: 32866782 DOI: 10.1016/j.intimp.2020.106864] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/13/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The aim of this study was to clarify the association between CMTM6 and PD-L1 expression as well as microenvironment in lung squamous carcinoma (LUSC). MATERIAL AND METHODS Using Spearman's correlation and Tumor Immune Estimation Resource (TIMER), we analyzed the relationship between CMTM6 and PD-L1 mRNA in LUSC. Immunohistochemistry (IHC) assay was applied to validate the correlation between CMTM6 and PD-L1 protein level in 80 LUSC samples originated from Shandong Provincial Hospital. Then, using The Cancer Genome Atlas (TCGA) database and fisher test, we analyzed the differential mutation genes in high and low CMTM6 expression group. TISIDB was used to explore the distribution of CMTM6 across immune- and molecular-subtypes. TCGA database and Gene Set variation analysis (GSVA) were used to analyze the relationship between CMTM6 and immune genes, immune related pathways. RESULT Positive correlation between CMTM6 and PD-L1 in mRNA and protein level was found in LUSC patients. More gene mutations were found in CMTM6 high expression group compared with low expression group. Meanwhile, we also found the correlation between CMTM6 expression and molecular subtypes, immune genes, immune related pathways. Furthermore, our result revealed that B cells memory, T cells memory testing, T cells folicular helper, macrophages M0, macrophages M1 and neutrophils varied significantly between patients with CMTM6 high and low expression group. Finally, we found that CMTM6 expression was positively related to CD8 + T cell, macrophage, neutrophil and dendtritic cell (all, P < 0.05) and negatively related to CD4 + T cell (P = 0.018). CONCLUSION CMTM6 is positively associated with PD-L1 expression and correlates with infiltration of immune cells in microenvironment of lung squamous carcinoma.
Collapse
Affiliation(s)
- Xiaoling Shang
- Department of Clinical Laboratory, Shandong University, Jinan 250012, China; Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Jia Li
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai 200032, China
| | - Hui Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Zhenxiang Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Jiamao Lin
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Dawei Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China.
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China.
| |
Collapse
|
38
|
Light A, Ahmed A, Dasgupta P, Elhage O. The genetic landscapes of urological cancers and their clinical implications in the era of high-throughput genome analysis. BJU Int 2020; 126:26-54. [PMID: 32306543 DOI: 10.1111/bju.15084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE With the advent of high-throughput genome analysis, we are increasingly able to sequence and hence understand the pathogenic processes underlying individual cancers. Recently, consortiums such as The Cancer Genome Atlas (TCGA) have performed large-scale projects to this end, providing significant amounts of information regarding the genetic landscapes of several cancers. PATIENTS AND METHODS We performed a narrative review of studies from the TCGA and other major studies. We aimed to summarise data exploring the clinical implications of specific genetic alterations, both prognostically and therapeutically, in four major urological cancers. These were renal cell carcinoma, muscle-invasive bladder cancer/carcinoma, prostate cancer, and testicular germ cell tumours. RESULTS With these four urological cancers, great strides have been made in the molecular characterisation of tumours. In particular, recent studies have focussed on identifying molecular subtypes of tumours with characteristic genetic alterations and differing prognoses. Other prognostic alterations have also recently been identified, including those pertaining to epigenetics and microRNAs. In regard to treatment, numerous options are emerging for patients with these cancers such as including immune checkpoint inhibition, epigenetic-based treatments, and agents targeting MAPK, PI3K, and DNA repair pathways. There are a multitude of trials underway investigating the effects of these novel agents, the results of which are eagerly awaited. CONCLUSIONS As medicine chases the era of personalised care, it is becoming increasingly important to provide individualised prognoses for patients. Understanding how specific genetic alterations affects prognosis is key for this. It will also be crucial to provide highly targeted treatments against the specific genetics of a patient's tumour. With work performed by the TCGA and other large consortiums, these aims are gradually being achieved. Our review provides a succinct overview of this exciting field that may underpin personalised medicine in urological oncology.
Collapse
Affiliation(s)
- Alexander Light
- Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, University of Cambridge, Cambridge, UK.,Bedford Hospital NHS Trust, Bedford Hospital, Bedford, UK
| | - Aamir Ahmed
- Centre for Stem Cell and Regenerative Medicine, King's College London, London, UK
| | - Prokar Dasgupta
- Department of Urology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Oussama Elhage
- Department of Urology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
39
|
Huang Y, Zeng J, Liu T, Xu Q, Song X, Zeng J. DNAM1 and 2B4 Costimulatory Domains Enhance the Cytotoxicity of Anti-GPC3 Chimeric Antigen Receptor-Modified Natural Killer Cells Against Hepatocellular Cancer Cells in vitro. Cancer Manag Res 2020; 12:3247-3255. [PMID: 32440221 PMCID: PMC7217313 DOI: 10.2147/cmar.s253565] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/04/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose Hepatocellular cancer (HCC) is the sixth most prevalent cancer and the third leading cause of cancer-related death worldwide. Cellular immunotherapy against glypican 3 (GPC3) has recently been used in the treatment of HCC, following the success of chimeric antigen receptor (CAR)-T therapy in treatment of B cell malignancy. However, CAR-T cells are not “off-the-shelf” and always cause cytokine release syndrome, which can be eliminated by using natural killer (NK) cells as effector cells. Since a costimulatory signal is necessary for the activation, persistence, or cytotoxicity of CAR-T cells, we speculated that the costimulatory signal is also required for CAR-NK cells in HCC treatment. Methods Five anti-GPC3 CAR plasmids containing different costimulatory domains were constructed. They included Z (only the CD3ζ domain, no costimulatory domain), CD28.Z (T-cell costimulatory domain CD28), DNAM1/2B4.Z (NK-cell-associated costimulatory domain DNAM1 or 2B4), and DNAM1.2B4.Z (both NK-cell-associated costimulatory domains). Respective CAR-NK-92 cells were generated. The MTT viability assay was performed to evaluate the effect of the different costimulatory domains on CAR-NK-cell proliferation. The effect on persistence was analyzed using an apoptosis assay and flow cytometry. Special cytotoxicity against normal hepatocellular cells and GPC3+ malignant cells was investigated in vitro. The concentration of cytokines (TNF-α and IFN-γ) released by CAR-NK-92 cells was also measured by ELISA. Results NK-cell-associated costimulatory signal was necessary for CAR-NK-92 cells. CAR-NK-92 cells with DNAM1 and/or 2B4 expanded more quickly and persisted with a lower apoptotic ratio, compared to the presence of CD28 or no costimulatory signal. All CAR-NK-92 cells showed special cellular cytotoxicity in vitro. CAR-NK-92 cells with NK-cell-associated costimulatory domains exhibited higher cytotoxic ability compared with those without any costimulatory domain or with T-cell costimulatory domain. CAR-NK-92 cells with both DNAM1 and 2B4 displayed the highest cytotoxicity. The cytokine release assay results were consistent with those of the cytotoxicity assay. Conclusion We provided the first evidence supporting a strategy using DNAM1 and 2B4 costimulatory domains to generate anti-GPC3 CAR-NK-92 cells, which exhibits enhanced cytotoxicity against hepatocellular cancer cells in vitro.
Collapse
Affiliation(s)
- Yao Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China.,Department of Hepatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Jianxing Zeng
- Department of Hepatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Teng Liu
- Department of Hepatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Qingyi Xu
- Department of Hepatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Xianglin Song
- Department of Hepatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Jinhua Zeng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China.,Department of Hepatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| |
Collapse
|
40
|
Slovin SF. Chimeric Antigen Receptor T-cell Therapy in Prostate Cancer: Reality or Folly? Eur Urol 2019; 77:309-310. [PMID: 31864760 DOI: 10.1016/j.eururo.2019.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/04/2019] [Indexed: 01/21/2023]
Affiliation(s)
- Susan F Slovin
- Genitourinary Oncology Service, Sidney Kimmel Center for Prostate and Urologic Cancer, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|