1
|
Chen J, Ma N, Chen B, Huang Y, Li J, Li J, Chen Z, Wang P, Ran B, Yang J, Bai J, Ning S, Ai J, Wei Q, Liu L, Cao D. Synergistic effects of immunotherapy and adjunctive therapies in prostate cancer management. Crit Rev Oncol Hematol 2025; 207:104604. [PMID: 39732304 DOI: 10.1016/j.critrevonc.2024.104604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024] Open
Abstract
In recent years, cancer immunotherapy has received widespread attention due to significant tumor clearance in some malignancies. Various immunotherapy approaches, including vaccines, immune checkpoint inhibitors, oncolytic virotherapy, bispecific T cell engagers, and adoptive T cell transfer, have completed or are undergoing clinical trials for prostate cancer. Despite immune checkpoint blockade's extraordinary effectiveness in treating a variety of cancers, targeted prostate cancer treatment using the immune system is still in its infancy. Multiple factors including the heterogeneity of prostate cancer, the cold tumor microenvironment, and a low level of neoantigens, contribute to the poor immunotherapy response. Significant effort is being devoted to improving immune-based prostate cancer therapy. Recently, several key discoveries demonstrate that prostate cancer immunotherapy agents may be used to promise better prognosis for patients as part of combination strategies with other agents targeting tumor-associated immune mechanism of resistance. Here, this review comprehensively examines the recent advancements in immunotherapy for prostate cancer, exploring its potential synergistic effects when combined with other treatment modalities to enhance clinical efficacy.
Collapse
Affiliation(s)
- Jie Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Ma
- Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd section, South Renmin Road, Chengdu 610041, China
| | - Bo Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yin Huang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinze Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zeyu Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Puze Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Biao Ran
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiahao Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingxing Bai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shu Ning
- Department of Urologic Surgery, University of California Davis, Davis, CA, USA
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liangren Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Dehong Cao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Phung SK, Zorko NA, Soignier Y, Waller RL, Shackelford M, Walker JT, Nelson TD, Selleck C, Bendzick LE, Kotz LE, Kile QM, Bozicevich AJ, Miller SE, Khaw M, Shetty M, Hinderlie P, Ehrhardt M, Li Y, Luo X, Dehm SM, Antonarakis ES, Kennedy PR, Miller JS, Felices M. A PSMA-Targeted Tri-Specific Killer Engager Enhances NK Cell Cytotoxicity against Prostate Cancer. Cancer Immunol Res 2025; 13:258-272. [PMID: 39545924 PMCID: PMC11790377 DOI: 10.1158/2326-6066.cir-24-0273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/07/2024] [Accepted: 11/14/2024] [Indexed: 11/17/2024]
Abstract
NK cell tumor infiltration is associated with good prognosis in patients with metastatic castration-resistant prostate cancer (mCRPC). NK cells recognize and kill targets by a process called natural cytotoxicity. We hypothesized that promoting an antigen-specific synapse with coactivation may enhance NK cell function in mCRPC. We describe a tri-specific killer engager (TriKE) construct that engages with the activating receptor CD16 on NK cells and prostate-specific membrane antigen (PSMA) on mCRPC cells and has an IL15 moiety that is essential for NK cell survival, proliferation, and priming. We show that the PSMA TriKE specifically binds to PSMA-expressing cells and significantly enhances expansion, degranulation, and cytokine production of NK cells derived from healthy donors or patients with prostate cancer. Bystander killing of PSMA-negative tumor cells was also achieved with PSMA TriKE treatment when cocultured with PSMA-positive cells, suggesting potential PSMA TriKE benefit in controlling tumor antigen escape. When tested under physiologic conditions recapitulating the mCRPC tumor microenvironment, NK cells treated with PSMA TriKE and prolonged exposure to hypoxia or myeloid-derived suppressor cells maintained their potent function whereas IL15-treated NK cells showed greatly impaired cytotoxicity. Finally, in vivo testing of PSMA TriKE showed improved tumor control and survival of mice as compared with IL15-treated and untreated control groups. In conclusion, PSMA TriKE demonstrates potential as a new therapy for advanced prostate cancer by providing additional signals to NK cells to maximize their antitumor potential in prostate cancer, especially in the setting of a hostile tumor microenvironment.
Collapse
Affiliation(s)
| | - Nicholas A. Zorko
- Masonic Cancer Center, Minneapolis, MN, USA
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | | | | | | | | | - Carly Selleck
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN, USA
| | - Laura E. Bendzick
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN, USA
| | - Laura E. Kotz
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN, USA
| | | | | | | | | | - Mihir Shetty
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN, USA
| | | | - Michael Ehrhardt
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | | | - Xianghua Luo
- Masonic Cancer Center, Minneapolis, MN, USA
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Scott M. Dehm
- Masonic Cancer Center, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Department of Urology, University of Minnesota, Minneapolis, MN, USA
| | - Emmanuel S. Antonarakis
- Masonic Cancer Center, Minneapolis, MN, USA
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Philippa R. Kennedy
- Masonic Cancer Center, Minneapolis, MN, USA
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey S. Miller
- Masonic Cancer Center, Minneapolis, MN, USA
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Martin Felices
- Masonic Cancer Center, Minneapolis, MN, USA
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
3
|
San-Jose Manso L, Alfranca A, Moreno-Pérez I, Ruiz-Vico M, Velasco C, Toquero P, Pacheco M, Zapatero A, Aldave D, Celada G, Albers E, Fenor de la Maza MD, García J, Castro E, Olmos D, Colomer R, Romero-Laorden N. Immunome profiling in prostate cancer: a guide for clinicians. Front Immunol 2024; 15:1398109. [PMID: 39635522 PMCID: PMC11614818 DOI: 10.3389/fimmu.2024.1398109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024] Open
Abstract
Tumor immune microenvironment (TIME) plays a key role to understand how tumors respond to prostate cancer (PC) therapies and potential mechanisms of resistance. Previous research has suggested that specific genomic aberrations, such as microsatellite instability (MSI) or CDK12 bi-allelic loss can allow PC patients more likely to respond to immune checkpoint inhibitors (ICI) or other immune therapies. However, responses to these treatments remain highly variable even in selected patients. Thus, it is essential to obtain more information about tumor immune cells that infiltrate these tumors, and on their plasticity and interactions, in order to better understand the underlying biology to allow development of new therapeutic strategies. This review analyzes: 1) How interactions among immune cell populations and other cells infiltrating the tumor stroma can modulate the progression of PC, 2) How the standard therapies to treat PC (such as androgen deprivation therapy, new androgen-directed hormone therapy or chemotherapy) may influence the dynamic changes of the immunome and 3) What are the limitations in characterizing the immune landscape of the host´s response to tumors.
Collapse
Affiliation(s)
| | - Arantzazu Alfranca
- Immunology Department, Hospital Universitario La Princesa, Madrid, Spain
- Personalized Precision Medicine Chair, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ignacio Moreno-Pérez
- Medical Oncology Department, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - María Ruiz-Vico
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Clara Velasco
- Urology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - Patricia Toquero
- Personalized Precision Medicine Chair, Universidad Autónoma de Madrid, Madrid, Spain
- Medical Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - María Pacheco
- GU Translational Research Unit, Instituto de Investigación Sanitaria de la Princesa, Madrid, Spain
| | - Almudena Zapatero
- Radiation Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - Diego Aldave
- Radiation Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - Guillermo Celada
- Urology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - Eduardo Albers
- Urology Department, Hospital Universitario La Princesa, Madrid, Spain
| | | | - Jorge García
- Biocomputing Unit, Hospital Niño Jesús, Instituto de Investigación Sanitaria de la Princesa, Madrid, Spain
| | - Elena Castro
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - David Olmos
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Ramón Colomer
- Personalized Precision Medicine Chair, Universidad Autónoma de Madrid, Madrid, Spain
- Medical Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - Nuria Romero-Laorden
- Personalized Precision Medicine Chair, Universidad Autónoma de Madrid, Madrid, Spain
- Medical Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
| |
Collapse
|
4
|
Alshehri AHD. Bone-Targeting Radionuclides in the Treatment of Metastatic Castration-Resistant Prostate Cancer: A Review on Radium-223 Chloride (Alpharadin) in Combination with Other Therapies. Diagnostics (Basel) 2024; 14:2407. [PMID: 39518374 PMCID: PMC11544963 DOI: 10.3390/diagnostics14212407] [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: 09/05/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Recent advances have broadened the range of therapeutic options for mCRPC, with several new treatments, including novel hormonal therapies (enzalutamide, abiraterone), chemotherapeutic agents (docetaxel, cabazitaxel), immunotherapies (sipuleucel-T), and bone targeting radiopharmaceuticals (radium-223) showing improved clinical outcomes and receiving U.S. Food and Drug Administration approval. These new treatments provide new avenues for improving patient survival and quality of life. Radium-223, a targeted alpha-emitter, specifically targets bone metastases, offering palliative benefits and a potential increase in life expectancy. The integration of radium-223 with other treatments shows promise for managing mCRPC. However, the optimal sequencing and combination of radium-223 with other therapies are still being explored, with various clinical trials investigating new therapeutic approaches. The integration of these therapies, especially to provide more effective, personalized treatment strategies, requires further investigation. A thorough literature review was conducted on current treatments for mCRPC, including chemotherapeutic agents, oral hormonal therapies targeting the androgen receptor axis, immunotherapies, and radium-223. Ongoing clinical trials investigating radium-233 in the context of other therapies for the treatment of mCRPC patients were also reviewed. Further studies should focus on determining the optimal sequencing and dosing and identifying biomarkers that predict treatment response to enhance outcomes of mCRPC patients. This review underlines the rational strategies of combining radium-223 with other therapies, investigating their impact on bone in terms of delaying skeletal-related events, and managing bone disease progression in mCRPC patients.
Collapse
Affiliation(s)
- Ali H D Alshehri
- Department of Radiological Sciences, Faculty of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| |
Collapse
|
5
|
Pandit-Taskar N, O'Donoghue JA, Chetty D, Max S, Wanik D, Ilovich O, Russell M, Nyima T, Divgi CR, Yu M, Morris MJ. A Phase 0 Study to Assess the Biodistribution and Pharmacokinetics of a Radiolabeled Antibody Targeting Human Kallikrein 2 in Participants with Metastatic Castration-Resistant Prostate Cancer. J Nucl Med 2024; 65:1051-1056. [PMID: 38782459 PMCID: PMC11218722 DOI: 10.2967/jnumed.124.267416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Despite the inclusion of multiple agents within the prostate cancer treatment landscape, new treatment options are needed to address the unmet need for patients with metastatic castration-resistant prostate cancer (mCRPC). Although prostate-specific membrane antigen is the only cell-surface target to yield clinical benefit in men with advanced prostate cancer, additional targets may further advance targeted immune, cytotoxic, radiopharmaceutical, and other tumor-directed therapies for these patients. Human kallikrein 2 (hK2) is a novel prostate-specific target with little to no expression in nonprostate tissues. This first-in-human phase 0 trial uses an 111In-radiolabeled anti-hK2 monoclonal antibody, [111In]-DOTA-h11B6, to credential hK2 as a potential target for prostate cancer treatment. Methods: Participants with progressive mCRPC received a single infusion of 2 mg of [111In]-DOTA-h11B6 (185 MBq of 111In), with or without 8 mg of unlabeled h11B6 to assess antibody mass effects. Sequential imaging and serial blood samples were collected to determine [111In]-DOTA-h11B6 biodistribution, dosimetry, serum radioactivity, and pharmacokinetics. Safety was assessed within a 2-wk follow-up period from the time of [111In]-DOTA-h11B6 administration. Results: Twenty-two participants received [111In]-DOTA-h11B6 and are included in this analysis. Within 6-8 d of administration, [111In]-DOTA-h11B6 visibly accumulated in known mCRPC lesions, with limited uptake in other organs. Two treatment-emergent adverse events unrelated to treatment occurred, including tumor-related bleeding in 1 patient, which led to early study discontinuation. Serum clearance, biodistribution, and tumor targeting were independent of total antibody mass (2 or 10 mg). Conclusion: This first-in-human study demonstrates that tumor-associated hK2 can be identified and targeted using h11B6 as a platform as the h11B6 antibody selectively accumulated in mCRPC metastases with mass-independent clearance kinetics. These data support the feasibility of hK2 as a target for imaging and hK2-directed agents as potential therapies in patients with mCRPC.
Collapse
Affiliation(s)
- Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical Center, New York, New York
| | - Joseph A O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dushen Chetty
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Steven Max
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | | | | | - Michael Russell
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Tenzin Nyima
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Margaret Yu
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Michael J Morris
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York; and
- Department of Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
6
|
Zhang YW, Gvozdenovic A, Aceto N. A Molecular Voyage: Multiomics Insights into Circulating Tumor Cells. Cancer Discov 2024; 14:920-933. [PMID: 38581442 DOI: 10.1158/2159-8290.cd-24-0218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 04/08/2024]
Abstract
Circulating tumor cells (CTCs) play a pivotal role in metastasis, the leading cause of cancer-associated death. Recent improvements of CTC isolation tools, coupled with a steady development of multiomics technologies at single-cell resolution, have enabled an extensive exploration of CTC biology, unlocking insights into their molecular profiles. A detailed molecular portrait requires CTC interrogation across various levels encompassing genomic, epigenetic, transcriptomic, proteomic and metabolic features. Here, we review how state-of-the-art multiomics applied to CTCs are shedding light on how cancer spreads. Further, we highlight the potential implications of CTC profiling for clinical applications aimed at enhancing cancer diagnosis and treatment. SIGNIFICANCE Exploring the complexity of cancer progression through cutting-edge multiomics studies holds the promise of uncovering novel aspects of cancer biology and identifying therapeutic vulnerabilities to suppress metastasis.
Collapse
Affiliation(s)
- Yu Wei Zhang
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Ana Gvozdenovic
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| |
Collapse
|
7
|
Zhou S, Hou X, Li L, Guo L, Wang H, Mao L, Shi L, Yuan M. Discovery of dolutegravir-1,2,3-triazole derivatives against prostate cancer via inducing DNA damage. Bioorg Chem 2023; 141:106926. [PMID: 37871389 DOI: 10.1016/j.bioorg.2023.106926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Prostate cancer (PCa) is the second most frequently diagnosed cancer among men, causing a huge number of deaths each year. Traditional chemotherapy for PCa mostly focused on targeting androgen receptors. However, some of the patients would develop resistance to hormonal therapy. In these cases, it is suggested for these patients to administer treatments in combination with other chemotherapeutics. Current chemotherapeutics for metastatic castration-resistant PCa could hardly reach satisfying effects, therefore it is crucial to explore novel agents with low cytotoxicity. Herein, a common drug against the human immunodeficiency virus (HIV), the dolutegravir (DTG) was modified to become a series of dolutegravir-1,2,3-triazole derivatives. Among these compounds, the 4d and 4q derivatives were verified with high anti-tumor efficiency, suppressing the proliferation of the prostate cancer cells PC3 and DU145. These compounds function by binding to the poly (adenosine diphosphate-ribose) polymerase (PARP), inactivating the PARP and inducing DNA damage in cancer cells. It is noteworthy that the 4d and 4q derivatives showed almost no impact on normal cells and mice. Thereby, the results reveal that these dolutegravir-1,2,3-triazole compounds are potential chemotherapeutics for PCa treatment.
Collapse
Affiliation(s)
- Shuyi Zhou
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xixi Hou
- Key Laboratory of Green Chemical Media and Reactions (Ministry of Education), Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Ling Li
- Department of Pharmacology, the Eighth Affiliated Hospital, Sun Yat-sen University, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Shenzhen, China
| | - LiHao Guo
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Huili Wang
- University of North Carolina Hospitals, 101 Manning Dr, Chapel Hill, Orange County, NC27599, USA
| | - Longfei Mao
- Key Laboratory of Green Chemical Media and Reactions (Ministry of Education), Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China; College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, 263 Kaiyuan Road, Luoyang 471003, China.
| | - Leilei Shi
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Miaomiao Yuan
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
8
|
Pungsrinont T, Schneider MA, Baniahmad A. Androgen receptor agonist and antagonist reduce response of cytokine-induced killer cells on prostate cancer cells. J Cell Mol Med 2023; 27:2970-2982. [PMID: 37639523 PMCID: PMC10538273 DOI: 10.1111/jcmm.17923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/31/2023] [Accepted: 08/12/2023] [Indexed: 08/31/2023] Open
Abstract
Despite many advances, prostate cancer (PCa) is still the second most frequently diagnosed cancer and fifth leading cause of cancer death in men worldwide. So far, the promising field of onco-immunology has not yet provided a satisfactory treatment option for PCa. Here we show that the ex vivo expansion and activation of cytokine-induced killer (CIK) cells isolated from primary peripheral blood mononuclear cells induce immune-mediated apoptosis in both human PCa LNCaP and C4-2 cells. Interestingly, pretreating LNCaP and C4-2 cells with either androgen or the androgen receptor (AR) antagonist enzalutamide mediates resistance to this immunogenic attack. This is associated with a reduction of both total cell loss and apoptosis levels suggesting one possible mechanism blunting onco-immunological activity. The data also suggest that secreted factors from AR ligand-treated PCa cell suppress lymphocyte proliferation. Further, we analysed immune-mediated killing activity using conditioned media from LNCaP and C4-2 treated cells. The obtained data suggest that the conditioned media from PCa treated cells does not influence a measurable lymphocyte-mediated apoptosis. However, analysing clonal expansion of activated lymphocytes, the androgen-derived conditioned media suppresses lymphocyte proliferation/expansion suggesting inhibition of onco-immunological activity by pretreatment of PCa cells with AR ligands.
Collapse
Affiliation(s)
- Thanakorn Pungsrinont
- Institute of Human Genetics, Jena University HospitalFriedrich Schiller UniversityJenaGermany
| | - Margret Ann Schneider
- Institute of Human Genetics, Jena University HospitalFriedrich Schiller UniversityJenaGermany
| | - Aria Baniahmad
- Institute of Human Genetics, Jena University HospitalFriedrich Schiller UniversityJenaGermany
| |
Collapse
|
9
|
Sooi K, Walsh R, Kumarakulasinghe N, Wong A, Ngoi N. A review of strategies to overcome immune resistance in the treatment of advanced prostate cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:656-673. [PMID: 37842236 PMCID: PMC10571060 DOI: 10.20517/cdr.2023.48] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/06/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023]
Abstract
Immunotherapy has become integral in cancer therapeutics over the past two decades and is now part of standard-of-care treatment in multiple cancer types. While various biomarkers and pathway alterations such as dMMR, CDK12, and AR-V7 have been identified in advanced prostate cancer to predict immunotherapy responsiveness, the vast majority of prostate cancer remain intrinsically immune-resistant, as evidenced by low response rates to anti-PD(L)1 monotherapy. Since regulatory approval of the vaccine therapy sipuleucel-T in the biomarker-unselected population, there has not been much success with immunotherapy treatment in advanced prostate cancer. Researchers have looked at various strategies to overcome immune resistance, including the identification of more biomarkers and the combination of immunotherapy with existing effective prostate cancer treatments. On the horizon, novel drugs using bispecific T-cell engager (BiTE) and chimeric antigen receptors (CAR) technology are being explored and have shown promising early efficacy in this disease. Here we discuss the features of the tumour microenvironment that predispose to immune resistance and rational strategies to enhance antitumour responsiveness in advanced prostate cancer.
Collapse
Affiliation(s)
| | | | | | | | - Natalie Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore 119228, Singapore
| |
Collapse
|
10
|
Saleem S, Rashid AB, Shehzadi S, Mumtaz H, Saqib M, Bseiso A, Villasenor AV, Ahmed A, Sonia SN. Contemporaneous and upcoming trends in immunotherapy for prostate cancer: review. Ann Med Surg (Lond) 2023; 85:4005-4014. [PMID: 37554896 PMCID: PMC10406079 DOI: 10.1097/ms9.0000000000001070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/02/2023] [Indexed: 08/10/2023] Open
Abstract
Prostate cancer (PCa) is the most common cancer in men worldwide. It affects more than 1.4 million men worldwide and kills up to 37 5000 people. PCa is routinely managed with chemotherapy and androgen deprivation therapy, but the success rate of these treatments is unsatisfactory. Immunotherapy is a novel method of treating different types of cancers, and it utilizes the body's own immune system to fight cancer. Different types of cancer respond differently to immunotherapy, with some showing excellent responses, while others do not show very satisfactory responses. PCa is known to be an immunologically cold tumor, such that conventional immunotherapy does not work as effectively as it works in other cancers. In the past decade, multiple studies and trials have been conducted to test different types of therapies, ranging from immune checkpoint inhibitors to anticancer vaccines to anticancer cytokines. Even after many studies, there is still a drug to be discovered that can completely cure any stage of PCa. Recent immunotherapeutic drug trials have started using immunotherapy in conjunction with chemotherapy and radiotherapy and have shown promising results. In this paper, the authors present a comprehensive overview of the currently used immunotherapeutic drugs as well as emerging immunotherapies, including modalities of combination immunotherapy with radiotherapy and chemotherapy. This review can help readers gain the latest knowledge about emerging trends in the current immunotherapy landscape for the treatment of PCa, as well as a general overview of the already used immunotherapy drugs for PCa.
Collapse
Affiliation(s)
| | | | | | | | | | - Anan Bseiso
- Hebron University, Palestine, State of Hebron
| | | | | | | |
Collapse
|
11
|
Makkena VK, Jaramillo AP, Awosusi BL, Ayyub J, Dabhi KN, Gohil NV, Tanveer N, Hussein S, Pingili S, Khan S. Probing the Relationship Between the Human Gut Microbiome and Prospects of Prostate Cancer: A Systematic Review. Cureus 2023; 15:e43892. [PMID: 37746426 PMCID: PMC10511825 DOI: 10.7759/cureus.43892] [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: 07/24/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Prostate neoplasia is one of the most commonly occurring neoplasias in males and has a high mortality rate. Prostate cancer (PCA) risk factors include tall stature, male sex, known family history, obesity, high blood pressure, lack of fitness, higher levels of testosterone for a long time, increasing age, and ethnicity are well known. The association and role of the gut microbiota in different diseases in our body have been highlighted recently. Therefore, finding the influence of gut microbiota on the prostatic cells can be useful for preventing prostatic neoplasia and/or reducing its severity. We aimed to assess its impact on PCA risk. We thoroughly searched databases for the relevant literature for our systematic review. The final research papers analyzed how bacteria played a role in the risk of PCA, either through inflammation or the production of metabolites that increase/decrease the risk of PCA. Based on the studies reviewed, we found that some gut bacteria play a role in the formation of PCA. In contrast, some bacteria can help prevent PCA, but the metabolism of the dietary components is the major factor for PCA.
Collapse
Affiliation(s)
- Vijaya Krishna Makkena
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Department of Medicine, Osmania Medical College, Hyderabad, IND
| | - Arturo P Jaramillo
- Department of Internal Medicine, Universidad Estatal de Guayaquil, Machala, ECU
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Babatope L Awosusi
- Department of Pathology and Laboratory Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Javaria Ayyub
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Karan Nareshbha Dabhi
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Namra V Gohil
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Department of Internal Medicine, Medical College Baroda, Vadodara, IND
| | - Nida Tanveer
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Sally Hussein
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Shravya Pingili
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Department of Medicine, Kakatiya Medical College, Hyderabad, IND
| | - Safeera Khan
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
12
|
Lauriola A, Davalli P, Marverti G, Santi S, Caporali A, D'Arca D. Targeting the Interplay of Independent Cellular Pathways and Immunity: A Challenge in Cancer Immunotherapy. Cancers (Basel) 2023; 15:cancers15113009. [PMID: 37296972 DOI: 10.3390/cancers15113009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/19/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Immunotherapy is a cancer treatment that exploits the capacity of the body's immune system to prevent, control, and remove cancer. Immunotherapy has revolutionized cancer treatment and significantly improved patient outcomes for several tumor types. However, most patients have not benefited from such therapies yet. Within the field of cancer immunotherapy, an expansion of the combination strategy that targets independent cellular pathways that can work synergistically is predicted. Here, we review some consequences of tumor cell death and increased immune system engagement in the modulation of oxidative stress and ubiquitin ligase pathways. We also indicate combinations of cancer immunotherapies and immunomodulatory targets. Additionally, we discuss imaging techniques, which are crucial for monitoring tumor responses during treatment and the immunotherapy side effects. Finally, the major outstanding questions are also presented, and directions for future research are described.
Collapse
Affiliation(s)
- Angela Lauriola
- Department of Biotechnology, University of Verona, 37134 Verona, Italy
| | - Pierpaola Davalli
- Department of Biomedical, Metabolic and Neural Sciences, Via G. Campi 287, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Gaetano Marverti
- Department of Biomedical, Metabolic and Neural Sciences, Via G. Campi 287, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Spartaco Santi
- Consiglio Nazionale delle Ricerche (CNR) Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", 40136 Bologna, Italy
- IRCCS, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Andrea Caporali
- BHF Centre for Cardiovascular Science, University of Edinburgh, Scotland EH4 2XU, UK
| | - Domenico D'Arca
- Department of Biomedical, Metabolic and Neural Sciences, Via G. Campi 287, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
13
|
Roy P, Singh KP. Epigenetic mechanism of therapeutic resistance and potential of epigenetic therapeutics in chemorefractory prostate cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 380:173-210. [PMID: 37657858 DOI: 10.1016/bs.ircmb.2023.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/03/2023]
Abstract
Prostate cancer is the second leading cause of cancer death among men in the United States. Depending upon the histopathological subtypes of prostate cancers, various therapeutic options, such as androgen deprivation therapy (ADT), androgen receptor signaling inhibitors (ARSI), immunotherapy, and chemotherapy, are available to treat prostate cancer. While these therapeutics are effective in the initial stages during treatments, the tumors subsequently develop resistance to these therapies. Despite all the progress made so far, therapeutic resistance remains a major challenge in the treatment of prostate cancer. Although various mechanisms have been reported for the resistance development in prostate cancer, altered expression of genes either directly or indirectly involved in drug response pathways is a common event. In addition to the genetic basis of gene regulation such as mutations and gene amplifications, epigenetic alterations involved in the aberrant expression of genes have frequently been shown to be associated not only with cancer initiation and progression but also with therapeutic resistance development. There are several review articles compiling reports on genetic mechanisms involved in therapeutic resistance in prostate cancer. However, epigenetic mechanisms for the therapeutic resistance development in prostate cancer have not yet been summarized in a review article. Therefore, the objective of this article is to compile various reports and provide a comprehensive review of the epigenetic aberrations, and aberrant expression of genes by epigenetic mechanisms involved in CRPCs and therapeutic resistance development in prostate cancer. Additionally, the potential of epigenetic-based therapeutics in the treatment of chemorefractory prostate cancer as evidenced by clinical trials has also been discussed.
Collapse
Affiliation(s)
- Priti Roy
- Department of Environmental Toxicology, Texas Tech University, Lubbock, TX, United States
| | - Kamaleshwar P Singh
- Department of Environmental Toxicology, Texas Tech University, Lubbock, TX, United States.
| |
Collapse
|
14
|
He W, Xiao Y, Yan S, Zhu Y, Ren S. Cell-free DNA in the management of prostate cancer: Current status and future prospective. Asian J Urol 2022. [PMID: 37538150 PMCID: PMC10394290 DOI: 10.1016/j.ajur.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective With the escalating prevalence of prostate cancer (PCa) in China, there is an urgent demand for novel diagnostic and therapeutic approaches. Extensive investigations have been conducted on the clinical implementation of circulating free DNA (cfDNA) in PCa. This review aims to provide a comprehensive overview of the present state of cfDNA as a biomarker for PCa and to examine its merits and obstacles for future clinical utilization. Methods Relevant peer-reviewed manuscripts on cfDNA as a PCa marker were evaluated by PubMed search (2010-2022) to evaluate the roles of cfDNA in PCa diagnosis, prognosis, and prediction, respectively. Results cfDNA is primarily released from cells undergoing necrosis and apoptosis, allowing for non-invasive insight into the genomic, transcriptomic, and epigenomic alterations within various PCa disease states. Next-generation sequencing, among other detection methods, enables the assessment of cfDNA abundance, mutation status, fragment characteristics, and epigenetic modifications. Multidimensional analysis based on cfDNA can facilitate early detection of PCa, risk stratification, and treatment monitoring. However, standardization of cfDNA detection methods is still required to expedite its clinical application. Conclusion cfDNA provides a non-invasive, rapid, and repeatable means of acquiring multidimensional information from PCa patients, which can aid in guiding clinical decisions and enhancing patient management. Overcoming the application barriers of cfDNA necessitates increased data sharing and international collaboration.
Collapse
|