1
|
Marranci A, Maresca L, Lodovichi S, Luserna di Rorà AG, Stecca B, Poliseno L. PARP1 in melanoma: Mechanistic insights and implications for basic and clinical research. Cancer Lett 2025; 617:217599. [PMID: 40024566 DOI: 10.1016/j.canlet.2025.217599] [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: 11/25/2024] [Revised: 02/04/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Targeted therapies and immunotherapies have revolutionized the treatment of metastatic melanoma and have set a successful example for the treatment of other cancers. A similar breakthrough was achieved with the advent of PARP inhibitors (PARPi) in breast and ovarian cancer. Recent evidence highlights the critical role of PARP1 in melanoma initiation and progression. High PARP1 expression correlates with aggressive melanoma characteristics and poor patient outcomes. Preclinical and clinical data suggest that PARPi, alone or in combination, can effectively reduce melanoma cell viability and inhibit tumor growth. However, integrating PARPi with current treatment approaches and identifying patients who could benefit the most from such combinations remain underexplored areas of investigation. This review highlights the need for further basic and clinical research on PARP1 in melanoma, to better understand its role and to tackle major challenges in the field, such as resistance to targeted therapies and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Andrea Marranci
- Oncohematology Unit, Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, 56017, Pisa, Italy. http://www.fpscience.it/
| | - Luisa Maresca
- Tumor Cell Biology Unit, Core Research Laboratory (CRL), Institute for Cancer Research and Prevention (ISPRO), 50139, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, 50139, Florence, Italy
| | - Samuele Lodovichi
- Department of Biosciences, University of Milan, 20133, Milan, Italy; Institute of Clinical Physiology, National Research Council (CNR-IFC), 56124, Pisa, Italy
| | | | - Barbara Stecca
- Tumor Cell Biology Unit, Core Research Laboratory (CRL), Institute for Cancer Research and Prevention (ISPRO), 50139, Florence, Italy
| | - Laura Poliseno
- Institute of Clinical Physiology, National Research Council (CNR-IFC), 56124, Pisa, Italy; Oncogenomics Unit, Core Research Laboratory, Institute for Cancer Research and Prevention (ISPRO), 56124, Pisa, Italy.
| |
Collapse
|
2
|
Qureshi Z, Fatima E, Safi A, Khanzada M, Altaf F. Talazoparib for the Treatment of Metastatic Castration-resistant Prostate Cancer: A Narrative Review. Am J Clin Oncol 2025; 48:206-214. [PMID: 39761644 DOI: 10.1097/coc.0000000000001159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Breast and prostate cancer are among the most commonly diagnosed cancers worldwide. Recent advances in tumor sequencing and gene studies have led to a paradigm shift from treatment centered on the type of tumor to therapy more focused on specific immune phenotype markers and molecular alterations. In this review, we discuss the utility and function of talazoparib concerning prostate cancer treatment and summarize recent and planned clinical trials on talazoparib. We searched medical databases for articles relating to the use of talazoparib in prostate cancer from inception. Poly ADP ribose polymerase (PARP) is a family of 17 necessary DNA repair enzymes responsible for base excision repair, single-strand break repair, and double-strand break repair. PARP inhibitors are a class of oral targeted therapies that compete for the NAD + binding site on PARP molecules. Talazoparib, a potent PARP inhibitor, has emerged as a significant therapeutic option in the treatment of metastatic castration-resistant prostate cancer (mCRPC), particularly for patients with specific genetic alterations. Its role as a PARP inhibitor makes it a targeted therapy, focusing on cancer cells with DNA repair deficiencies. Talazoparib's role as a biomarker-directed therapy in advanced prostate cancer has been increasingly recognized. The TALAPRO-1 demonstrated durable antitumor activity in mCRPC patients. TALAPRO-2 is a notable clinical trial, specifically examining the effectiveness of Talazoparib when used in combination therapies. Current investigations demonstrate a significant improvement in survival outcomes for the patients of mCRPC, making Talazoparib a promising intervention.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences
| | - Adnan Safi
- Department of Medicine, Lahore General Hospital
| | - Mikail Khanzada
- Department of Internal Medicine, Lahore Medical and Dental College, Lahore, Pakistan
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY
| |
Collapse
|
3
|
Sharma N, Bhati A, Aggarwal S, Shah K, Dewangan HK. PARP Pioneers: Using BRCA1/2 Mutation-targeted Inhibition to Revolutionize Breast Cancer Treatment. Curr Pharm Des 2025; 31:663-673. [PMID: 39421986 DOI: 10.2174/0113816128322894241004051814] [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: 05/06/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
Breast cancer stands on the second position in the world in being common and women happen to have it with high rate of about five-folds around the world. The causes of occurrence can matter with different humans be it external factors or the internal genetic ones. Breast cancer is primarily driven by mutations in the BRCA1 and BRCA2 susceptibility genes. These BC susceptibility genes encode proteins critical for DNA homologous recombination repair (HRR). Poly (ADP ribose) polymerases (PARP) are the essential enzymes involved in the repairing of the damaged DNA. So the inhibition of these inhibitors can be considered as the promising strategy for targeting cancers with defective damage in the deoxyribonucleic acid. Olaparib and talazoparib are PARP inhibitors (PARPi) are being employed for the monotherapies in case of the deleterious germline HER2-negative and BRCA-mutated breast cancer. The potency of PARP for trapping on DNA and causes cytotoxicity may have difference in the safety and efficacy with the PARPi. The PARPi have been found its place in the all different types of breast cancers and have shown potential benefits. The purpose of this review is to provide an update on the oral poly (ADP-ribose) polymerase (PARP) inhibitors for the improvement in the treatment and management of breast cancer.
Collapse
Affiliation(s)
- Navneet Sharma
- University Institute of Pharma Sciences (UIPS), Chandigarh University, NH-05, Chandigarh Ludhiana Highway, Mohali, Punjab, Pin: 160101, India
| | - Akash Bhati
- University Institute of Pharma Sciences (UIPS), Chandigarh University, NH-05, Chandigarh Ludhiana Highway, Mohali, Punjab, Pin: 160101, India
| | - Shagun Aggarwal
- University Institute of Pharma Sciences (UIPS), Chandigarh University, NH-05, Chandigarh Ludhiana Highway, Mohali, Punjab, Pin: 160101, India
| | - Kamal Shah
- Institute of Pharmaceutical Research (IPR), GLA University, NH-2, Delhi Mathura Road, PO-Chaumuhan, Mathura, Uttar Pradesh, India
| | - Hitesh Kumar Dewangan
- University Institute of Pharma Sciences (UIPS), Chandigarh University, NH-05, Chandigarh Ludhiana Highway, Mohali, Punjab, Pin: 160101, India
| |
Collapse
|
4
|
Giunta EF, Roviello G, Conteduca V, Verzoni E, Procopio G, De Giorgi U. Pharmacological treatment landscape of non-metastatic hormone-sensitive prostate cancer: A narrative review on behalf of the meet-URO Group. Crit Rev Oncol Hematol 2024; 204:104534. [PMID: 39447667 DOI: 10.1016/j.critrevonc.2024.104534] [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: 02/27/2024] [Revised: 09/20/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024] Open
Abstract
The definition of "non-metastatic hormone-sensitive prostate cancer" (nmHSPC) can be applied to patients with prostate cancer (PC) who are androgen-deprivation therapy-naïve and without evidence of metastatic disease. This definition includes heterogeneous situations; however, PC patients at high risk of metastatic spread - and who have not started a hormonal treatment - constitute a unique category with unmet clinical needs. This narrative review critically discusses the advances that characterize the rapidly evolving diagnostic and therapeutic scenario in the nmHSPC setting. We found that nmHSPC represents a grey zone in the context of PC. New clinical trials are trying to redefine the therapeutic algorithm of these patients, but escalating treatment seems not to be the right choice for the overall population. Biomarkers able to stratify patients - including molecular ones - are urgently needed, and biomarker-based clinical trials could clarify their prognostic and predictive role in the nmHSPC scenario.
Collapse
Affiliation(s)
- Emilio Francesco Giunta
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, Italy
| | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze, Italy
| | - Vincenza Conteduca
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Elena Verzoni
- SSD Genitourinary Medical Oncology and Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppe Procopio
- SSD Genitourinary Medical Oncology and Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, Italy.
| |
Collapse
|
5
|
Szczotka J, Szpila G, Hejduk M, Mucha E, Rudel J, Kępiński M, Kaletka J, Ryszawy J, Zapala P, Tsuboi I, Matsukawa A, Miszczyk M, Fazekas T, Zattoni F, Bryniarski P, Rajwa P. Role of PARP inhibitors in prostate cancer. Cent European J Urol 2024; 77:424-435. [PMID: 40115493 PMCID: PMC11921958 DOI: 10.5173/ceju.2024.72.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/26/2024] [Indexed: 03/23/2025] Open
Abstract
Introduction Olaparib, rucaparib, niraparib, and talazoparib are poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) targeted at recombination. To gain a comprehensive understanding of the mechanism of action of PARPi, scientists conducted research involving numerous studies that provided evidence regarding their efficacy and safety. Material and methods A literature review was performed using the PubMed® and Google Scholar databases. Articles were reviewed and categorized based on the most crucial and current information regarding the pharmacological properties and use of PARPi in treating metastatic castration-resistant prostate cancer (mCRPC), while also indicating the future therapeutic direction toward which these pharmaceuticals are progressing. Data were extracted, analyzed and summarized. Results PARP inhibitors like olaparib, rucaparib, niraparib, and talazoparib show promise in mCRPC, particularly for patients with specific genetic mutations (BRCA1/2, ATM). While they extend PFS and sometimes OS, side effects - especially anemia - are prevalent and impact treatment continuation. Conclusions Despite PARPi already being recognized as the standard treatment for mCRPC, further research is crucial to optimize their efficacy and safety, particularly in the context of combination therapies and use in the early stages of the disease.
Collapse
Affiliation(s)
- Julia Szczotka
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Gabriela Szpila
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Michał Hejduk
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Ewa Mucha
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Jolanta Rudel
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Michał Kępiński
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Julia Kaletka
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Jakub Ryszawy
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Piotr Zapala
- Clinic of General, Oncological, and Functional Urology, Medical University of Warsaw, Poland
| | - Ichiro Tsuboi
- Department of Urology, Medical University of Vienna, Austria
- Department of Urology, Shimane University Faculty of Medicine, Izumo, Shimane, Japan
| | - Akihiro Matsukawa
- Department of Urology, Medical University of Vienna, Austria
- Department of Urology, Jikei University School of Medicine, Tokyo, Japan
| | - Marcin Miszczyk
- Department of Urology, Medical University of Vienna, Austria
- Collegium Medicum, Faculty of Medicine, WSB University, Dabrowa Gornicza, Poland
| | - Tamas Fazekas
- Department of Urology, Medical University of Vienna, Austria
- Department of Urology, Semmelweis University, Budapest, Hungary
| | - Fabio Zattoni
- Department of Surgery, Oncology and Gastroenterology, Urologic Unit, University of Padua, Italy
- Department of Medicine - DIMED, University of Padua, Italy
| | - Piotr Bryniarski
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Paweł Rajwa
- Department of Urology, Medical University of Silesia, Zabrze, Poland
- Department of Urology, Medical University of Vienna, Austria
| |
Collapse
|
6
|
Thapa B, De Sarkar N, Giri S, Sharma K, Kim M, Kilari D. Integrating PARP Inhibitors in mCRPC Therapy: Current Strategies and Emerging Trends. Cancer Manag Res 2024; 16:1267-1283. [PMID: 39308935 PMCID: PMC11416116 DOI: 10.2147/cmar.s411023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 08/16/2024] [Indexed: 09/25/2024] Open
Abstract
Metastatic castrate-resistant prostate cancer (mCRPC) is associated with poor prognosis. DNA damage response (DDR) genes are commonly altered in mCRPC rendering them as promising therapeutic targets. Poly (ADP ribose) polymerase inhibitors (PARPi) demonstrated antitumor activity in mCRPC patients with DDR gene mutations through synthetic lethality. Multiple clinical trials with PARPi monotherapy exhibited encouraging clinical outcomes in selected patients with mCRPC. More recently, three Phase III randomized clinical trials (RCTs) combining PARPi with androgen receptor signaling inhibitors (ARSIs) demonstrated improved antitumor activity compared to ARSI monotherapy in mCRPC patients as the first-line therapy. Clinical benefit was more pronounced in patients harboring DDR alterations, specifically BRCA1/2. Interestingly, antitumor activity was also observed irrespective of DDR gene mutations, highlighting BRCAness phenotype with androgen receptor blockade resulting in synergistic activity between ARSIs and PARPi. In this review, we discuss the clinical efficacy and safety data of the combination of PARPi plus ARSI in all Phase 3 randomized controlled trials (RCTs), emphasizing strategies for patient selection and highlighting emerging trends based on clinical trial data.
Collapse
Affiliation(s)
- Bicky Thapa
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Navonil De Sarkar
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
- Data Science Institute, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Subhajit Giri
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Komal Sharma
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
- Data Science Institute, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mingee Kim
- School of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Deepak Kilari
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
7
|
Bilen MA, Khilfeh I, Rossi C, Muser E, Morrison L, Hilts A, Diaz L, Lefebvre P, Pilon D, George DJ. Homologous Recombination Repair Testing Patterns and Outcomes in mCRPC by Alteration Status and Race. CLINICOECONOMICS AND OUTCOMES RESEARCH 2024; 16:657-674. [PMID: 39257456 PMCID: PMC11385689 DOI: 10.2147/ceor.s468680] [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: 03/16/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
Background Alterations in DNA damage repair genes in advanced prostate cancer (PC) may impact responses to therapy and clinical outcomes. This study described homologous recombination repair (HRR) testing patterns and clinical outcomes among patients with metastatic castration-resistant prostate cancer (mCRPC) by HRR alteration status and race in the United States (US). Methods Clinical data in the nationwide (US-based) Flatiron Health-Foundation Medicine, Inc. (FMI) Metastatic PC Clinico-Genomic Database were evaluated (01/01/2011-12/31/2022). Patients initiating first-line (1L) mCRPC therapy on or after mCRPC diagnosis were included. Testing patterns, time-to-next treatment, overall survival (OS), and time-to-prostate specific antigen response were described. Results Of the 1367 patients with mCRPC and at least one HRR panel test prior to or on the date of 1L mCRPC therapy initiation, 332 (24.3%) were HRR positive (White patients: n = 219 [66.0%]; Black patients: n = 37 [11.1%]) and 1035 (75.7%) were HRR negative (White patients: n = 702 [67.8%]; Black patients: n = 84 [8.1%]). The mean time between first positive test and 1L mCRPC therapy initiation date was 588 days (White patients: 589 days; Black patients: 639 days). Among HRR positive relative to negative patients, trends for faster progression (respective 12-month rate overall: 71.1% and 63.7%; White patients: 72.5% and 64.0%; Black patients: 65.4% and 56.4%), shorter OS (respective 24-month rate overall: 46.8% and 51.9%; White patients: 48.6% and 46.2%; Black patients: 52.8% and 54.1%), and decreased treatment response (respective 12-month rate overall: 24.3% and 37.9%; White patients: 24.5% and 35.2%; Black patients: 17.0% and 43.9%) were observed. Conclusion Patients with mCRPC positive for HRR alterations tended to exhibit poorer treatment responses and clinical outcomes than those with a negative status. These findings highlight the importance of timely genetic testing in mCRPC, particularly among Black patients, and the need for improved 1L targeted therapies to address the unmet need in HRR positive mCRPC.
Collapse
Affiliation(s)
- Mehmet Asim Bilen
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | - Erik Muser
- Janssen Scientific Affairs, LLC, Horsham, PA, USA
| | | | | | | | | | | | - Daniel J George
- Department of Medicine, Duke University Cancer Center, Durham, NC, USA
| |
Collapse
|
8
|
Saleem MA, Mustafa MS. Promoter Hypermethylation of the BRCA1 Gene as a Novel Biomarker for Prostate Cancer. Cureus 2024; 16:e66467. [PMID: 39246954 PMCID: PMC11380563 DOI: 10.7759/cureus.66467] [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] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Prostate cancer (PCa) is recognized as one of the most common malignancies that greatly affects the male population globally. Breast cancer gene 1 (BRCA1) is an important tumor suppressor gene that plays a central role in the maintenance of genomic integrity by promoting the repair of double-strand breaks of DNA. Here, we present a pilot study to examine the promoter methylation and gene expression of the BRCA1 gene in patients with PCa in Erbil governorate, Iraq. The collection of samples took place in Erbil City, Iraq, specifically at Rizgary Hospital, PAR Hospital, and Al-Mufti's private laboratory. A total of 40 tissue samples were collected from age-matched individuals, comprising 30 pathologically confirmed PCa cases and 10 normal prostatic tissue taken from individuals who, during diagnosis, were found to be negative for PCa. Data on demographic and clinical information, such as pathological stage, age, and prostate-specific antigen (PSA) level, were gathered from the medical records. The impact of the promoter methylation was forecasted using the DNA bisulfite conversion technique and methyl-specific PCR (MSP) with specific primers for the BRCA1 promoter region. The assessment of BRCA1 expression was conducted using quantitative real-time PCR (qPCR). Among the 30 patients examined, 76.6% (23 cases) were found to have BRCA1 promoter methylation, and none of the normal tissues appeared to have DNA methylation. BRCA1 promoter methylation was positively associated with the advanced stage of disease (p=0.01) and Gleason score (p=0.007). The analysis revealed a significant downregulation of the BRCA1 gene expression in methylated tumor samples as compared to non-methylated tumors and normal tissues, suggesting the role of epigenetic silencing. To the best of our knowledge, this is the first study investigating methylation status and level of BRCA1 mRNA transcripts among PCa patients in Iraq. Our findings suggest that promoter hypermethylation of the BRCA1 gene could serve as a viable biomarker for PCa, marking a significant discovery.
Collapse
Affiliation(s)
- Mohammed A Saleem
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, IRQ
| | - Mustafa S Mustafa
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, IRQ
| |
Collapse
|
9
|
Garofoli M, Maiorano BA, Bruno G, Giordano G, Di Tullio P, Maselli FM, Landriscina M, Conteduca V. Androgen receptor, PARP signaling, and tumor microenvironment: the 'perfect triad' in prostate cancer? Ther Adv Med Oncol 2024; 16:17588359241258443. [PMID: 38887656 PMCID: PMC11181896 DOI: 10.1177/17588359241258443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
Aberrations in the homologous recombination repair (HRR) pathway in prostate cancer (PCa) provide a unique opportunity to develop therapeutic strategies that take advantage of the reduced tumor ability to repair DNA damage. Poly-ADP-ribose polymerase (PARP) inhibitors (PARPi) have been shown to prolong the survival of PCa patients with HRR defects, particularly in those with Breast Cancer type 1 susceptibility protein/Breast Cancer type 2 susceptibility protein alterations. To expand the benefit of PARPi to patients without detectable HRR alterations, multiple preclinical and clinical studies are addressing potential synergies between PARPi and androgen receptor signaling inhibitors, and these strategies are also being evaluated in combination with other drugs such as immune checkpoint inhibitors. However, the effectiveness of these combining therapies could be hindered by multiple mechanisms of resistance, including also the role played by the immunosuppressive tumor microenvironment. In this review, we summarize the use of PARPi in PCa and the potential synergies with different molecular pathways. However, numerous unanswered questions remain, including the identification of the patient population that could benefit most from PARPi, determining whether to use PARPi as monotherapy or in combination, and finding the optimal timing of PARPi, expanding the use of genomic tests, and optimizing combination therapies.
Collapse
Affiliation(s)
- Marianna Garofoli
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | | | - Giuseppina Bruno
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Guido Giordano
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Piergiorgio Di Tullio
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Felicia Maria Maselli
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Matteo Landriscina
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Viale Pinto, 1, Foggia 71122, Italy
| | - Vincenza Conteduca
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Viale Pinto, 1, Foggia 71122, Italy
| |
Collapse
|
10
|
Kwon WA. PARP Inhibitors in the Treatment of Prostate Cancer: From Scientific Rationale to Clinical Development. World J Mens Health 2024; 42:290-303. [PMID: 37853532 PMCID: PMC10949026 DOI: 10.5534/wjmh.230177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 07/19/2023] [Indexed: 10/20/2023] Open
Abstract
Prostate cancer (PC) treatment has reached a milestone with the introduction of poly(ADP-ribose) polymerase (PARP) inhibitors. PARP inhibitors (PARPi) induce breaks in single-stranded and/or double-stranded DNA, resulting in synthetic lethality in cancer cells lacking functional homologous recombination genes. Around 20% to 25% of patients with metastatic castration-resistant prostate cancer harbor mutations in DNA damage repair genes, either somatic or germline. The success of PARPi in these patients has prompted studies exploring its potential in tumors classified as "BRCAness," which refers to tumors without germline BRCA1 or BRCA2 mutations. Additionally, there is a proposed connection between androgen receptor signaling and synthetic lethality of PARPi. The inclusion of genetic mutation tests in the treatment algorithm for PC is a significant step towards precision and personalized medicine, marking a first in the field. The objectives of this review encompass understanding the mechanism of action of PARPi in both monotherapy and combination therapy, exploring patient selection criteria, discussing pivotal studies that led to its approval, and offering future prospects. However, numerous unanswered questions remain, including the identification of the patient population that could benefit most from PARPi, determining whether to use PARPi as monotherapy or in combination, and finding the optimal timing of PARPi administration in advanced or localized disease. To address these questions, several ongoing clinical trials are being conducted.
Collapse
Affiliation(s)
- Whi-An Kwon
- Department of Urology, Myongji Hospital, Hanyang University College of Medicine, Goyang, Korea.
| |
Collapse
|
11
|
Zaman N, Kushwah AS, Badriprasad A, Chakraborty G. Unravelling the molecular basis of PARP inhibitor resistance in prostate cancer with homologous recombination repair deficiency. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 389:257-301. [PMID: 39396849 PMCID: PMC11855062 DOI: 10.1016/bs.ircmb.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Prostate cancer is a disease with heterogeneous characteristics, making its treatability and curability dependent on the cancer's stage. While prostate cancer is often indolent, some cases can be aggressive and evolve into metastatic castration-resistant prostate cancer (mCRPC), which is lethal. A significant subset of individuals with mCRPC exhibit germline and somatic variants in components of the DNA damage repair (DDR) pathway. Recently, PARP inhibitors (PARPi) have shown promise in treating mCRPC patients who carry deleterious alterations in BRCA2 and 13 other DDR genes that are important for the homologous recombination repair (HRR) pathway. These inhibitors function by trapping PARP, resulting in impaired PARP activity and increased DNA damage, ultimately leading to cell death through synthetic lethality. However, the response to these inhibitors only lasts for 3-4 months, after which the cancer becomes PARPi resistant. Cancer cells can develop resistance to PARPi through numerous mechanisms, such as secondary reversion mutations in DNA repair pathway genes, heightened drug efflux, loss of PARP expression, HRR reactivation, replication fork stability, and upregulation of Wnt/Catenin and ABCB1 pathways. Overcoming PARPi resistance is a critical and complex process, and there are two possible ways to sensitize the resistance. The first approach is to potentiate the PARPi agents through chemo/radiotherapy and combination therapy, while the second approach entails targeting different signaling pathways. This review article highlights the latest evidence on the resistance mechanism of PARPi in lethal prostate cancer and discusses additional therapeutic opportunities available for prostate cancer patients with DDR gene alterations who do not respond to PARPi.
Collapse
Affiliation(s)
- Nabila Zaman
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Atar Singh Kushwah
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Anagha Badriprasad
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Goutam Chakraborty
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
12
|
Corpetti M, Müller C, Beltran H, de Bono J, Theurillat JP. Prostate-Specific Membrane Antigen-Targeted Therapies for Prostate Cancer: Towards Improving Therapeutic Outcomes. Eur Urol 2024; 85:193-204. [PMID: 38104015 DOI: 10.1016/j.eururo.2023.11.018] [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: 08/02/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023]
Abstract
CONTEXT Prostate-specific membrane antigen (PSMA) is a transmembrane glycoprotein overexpressed in most prostate cancers and exploited as a target for PSMA-targeted therapies. Different approaches to target PSMA-expressing cancer cells have been developed, showing promising results in clinical trials. OBJECTIVE To discuss the regulation of PSMA expression and the main PSMA-targeted therapeutic concepts illustrating their clinical development and rationalizing combination approaches with examples. EVIDENCE ACQUISITION We performed a detailed literature search using PubMed and reviewed the American Society of Clinical Oncology and European Society of Medical Oncology annual meeting abstracts up to September 2023. EVIDENCE SYNTHESIS We present an overarching description of the different strategies to target PSMA. The outcomes of PSMA-targeted therapies strongly rely on surface-bound PSMA expression. However, PSMA heterogeneity at different levels (interpatient and inter/intratumoral) limits the efficacy of PSMA-targeted therapies. We highlight the molecular mechanisms governing PSMA regulation, the understanding of which is crucial to designing therapeutic strategies aimed at upregulating PSMA expression. Thus far, homeobox B13 (HOXB13) and androgen receptor (AR) have emerged as critical transcription factors positively and negatively regulating PSMA expression, respectively. Furthermore, epigenetic regulation of PSMA has been also reported recently. In addition, many established therapeutic approaches harbor the potential to upregulate PSMA levels as well as potentiate DNA damage mediated by current radioligands. CONCLUSIONS PSMA-targeted therapies are rapidly advancing, but their efficacy is strongly limited by the heterogeneous expression of the target. A thorough comprehension of how PSMA is regulated will help improve the outcomes through increasing PSMA expression and will provide the basis for synergistic combination therapies. PATIENT SUMMARY Prostate-specific membrane antigen (PSMA) is overexpressed in most prostate cancers. PSMA-targeted therapies have shown promising results, but the heterogeneous expression of PSMA limits their efficacy. We propose to better elucidate the regulation of PSMA expression to increase the levels of the target and improve the therapeutic outcomes.
Collapse
Affiliation(s)
- Matteo Corpetti
- Institute of Oncology Research, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Cristina Müller
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland; Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Johann de Bono
- The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK
| | - Jean-Philippe Theurillat
- Institute of Oncology Research, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland.
| |
Collapse
|
13
|
Warli SM, Velaro AJ, Firsty NN, Tala ZZ. Addition of Olaparib to the New Hormonal Agent Regimen for Metastatic Castration-Resistant Prostate Cancer: A Systematic Review and Meta-Analysis. World J Oncol 2023; 14:518-528. [PMID: 38022404 PMCID: PMC10681786 DOI: 10.14740/wjon1685] [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: 08/01/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background The emergence of olaparib, a poly (adenosine diphosphate (ADP)-ribose) polymerase (PARP) inhibitor to treat metastatic castration-resistant prostate cancer (mCRPC), created a measurable clinical question on whether the agent positively influences the treatment outcomes and acceptable safety factors. The objective was to elaborate on the efficacy and safety of olaparib-added regimens in treating mCRPC patients as compared to the established guideline. Methods The literature search was performed on several scientific databases, e.g., PubMed, Cochrane, and ScienceDirect, by applying the Boolean Term method. Statistical and risk of bias (RoB) analyses were calculated through RevMan 5.4.1. to investigate our outcomes, i.e., progression-free survival (PFS) and overall survival (OS) with the reported adverse effects (AEs). These outcomes were presented in hazard ratio (HR) and risk ratio (RR). Results Three trials consisting of 1,325 individuals with comparable baseline characteristics were investigated. The meta-analysis showed that introducing olaparib into the regimens significantly improved the PFS (HR 0.59 (0.48 - 0.73); P < 0.05), which disclosed even better outcomes among mutated homologous recombinant repair (HRR) and ataxia-telangiectasia mutated (ATM) gene (HR 0.43 (0.30 - 0.62); P < 0.05) in 95% confidence interval (CI). Furthermore, similar outcomes were observed in OS analysis (HR 0.81 (0.67 - 0.99); P < 0.05), despite olaparib group disclosed higher AEs rate with insignificant difference in mortality rate. Conclusion The efficacy and safety of olaparib-added regimens in mCRPC patients need to be explored more extensively in trials because they are beneficial, particularly among HRR-mutated individuals.
Collapse
Affiliation(s)
- Syah Mirsya Warli
- Department of Urology, Universitas Sumatera Utara Hospital, Universitas Sumatera Utara, Medan, Indonesia
- Division of Urology, Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara-Haji Adam Malik General Hospital, Medan, Indonesia
| | - Adrian Joshua Velaro
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Naufal Nandita Firsty
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | | |
Collapse
|
14
|
Lourenço T, Vale N. Entecavir: A Review and Considerations for Its Application in Oncology. Pharmaceuticals (Basel) 2023; 16:1603. [PMID: 38004468 PMCID: PMC10675314 DOI: 10.3390/ph16111603] [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: 10/17/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Entecavir (ETV) is a drug used as a first-line treatment for chronic hepatitis B (CHB) virus infection because it is a guanosine nucleoside analogue with activity against the hepatitis B virus polymerase. The ETV dosage can range from 0.5 mg to 1 mg once a day and the most common side effects include headache, insomnia, fatigue, dizziness, somnolence, vomiting, diarrhea, nausea, dyspepsia, and increased liver enzyme levels. In addition to its conventional use, ETV acts as an inhibitor of lysine-specific demethylase 5B (KDM5B), an enzyme that is overexpressed in breast, lung, skin, liver, and prostate tumors and is involved in the hormonal response, stem cell regeneration, genomic stability, cell proliferation, and differentiation. The KDM5B enzyme acts as a transcriptional repressor in tumor suppressor genes, silencing them, and its overexpression leads to drug resistance in certain tumor types. Furthermore, the literature suggests that KDM5B activates the PI3K/AKT signaling pathway, while reducing KDM5B expression decreases AKT signaling, resulting in decreased tumor cell proliferation. In silico studies have demonstrated that ETV can inhibit tumor cell proliferation and induce apoptosis by reducing KDM5B expression. ETV also appears to inhibit PARP-1, has a high genetic barrier, reducing the chance of resistance development, and can also prevent the reactivation of the hepatitis B virus in cancer patients, which have proven to be significant advantages regarding its use as a repurposed drug in oncology. Therefore, ETV holds promise beyond its original therapeutic indication.
Collapse
Affiliation(s)
- Tânia Lourenço
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
15
|
Cancel-Tassin G, Koutros S. Use of genomic markers to improve epidemiologic and clinical research in urology. Curr Opin Urol 2023; 33:414-420. [PMID: 37642472 PMCID: PMC11382258 DOI: 10.1097/mou.0000000000001126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
PURPOSE OF REVIEW Urologic cancers result from the appearance of genomic alterations in the target organ due to the combination of genetic and environmental factors. Knowledge of the genomic markers involved in their etiology and mechanisms for their development continue to progress. This reviewed provides an update on recent genomic studies that have informed epidemiologic and clinical research in urology. RECENT FINDINGS Inherited variations are an established risk factor for urologic cancers with significant estimates of heritability for prostate, kidney, and bladder cancer. The roles of both rare germline variants, identified from family-based studies, and common variants, identified from genome-wide association studies, have provided important information about the genetic architecture for urologic cancers. Large-scale analyses of tumors have generated genomic, epigenomic, transcriptomic, and proteomic data that have also provided novel insights into etiology and mechanisms. These tumors characteristics, along with the associated tumor microenvironment, have attempted to provide more accurate risk stratification, prognosis of disease and therapeutic management. SUMMARY Genomic studies of inherited and acquired variation are changing the landscape of our understanding of the causes of urologic cancers and providing important translational insights for their management. Their use in epidemiologic and clinical studies is thus essential.
Collapse
Affiliation(s)
- Géraldine Cancel-Tassin
- Centre for Research on Prostatic Diseases (CeRePP), Paris, France
- GRC 5 Predictive Onco-Urology, Sorbonne University, Paris, France
| | - Stella Koutros
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| |
Collapse
|
16
|
Iannantuono GM, Chandran E, Floudas CS, Choo-Wosoba H, Butera G, Roselli M, Gulley JL, Karzai F. Efficacy and safety of PARP inhibitors in metastatic castration-resistant prostate cancer: A systematic review and meta-analysis of clinical trials. Cancer Treat Rev 2023; 120:102623. [PMID: 37716332 PMCID: PMC10591840 DOI: 10.1016/j.ctrv.2023.102623] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/18/2023]
Abstract
INTRODUCTION PARP inhibitors (PARPi) are a standard-of-care (SoC) treatment option for patients with metastatic castration-resistant prostate cancer (mCRPC). Several clinical trials have shown the potential of combining PARPi with other anticancer agents. Therefore, we conducted a systematic review and meta-analysis to comprehensively evaluate the efficacy and safety of PARPi in patients with metastatic prostate cancer. METHODS MEDLINE, Cochrane CENTRAL, EMBASE, CINAHL, and Web of Science were searched on March 22nd, 2023, for phase 2 or 3 clinical trials. Efficacy (progression-free survival [PFS], overall survival [OS], PSA decline >50% [PSA50], and objective response rate [ORR]) and safety outcomes were assessed in the included studies. RESULTS Seventeen clinical trials (PARPi monotherapy [n = 7], PARPi + androgen-receptor signaling inhibitors [ARSI] [n = 6], and PARPi + immune checkpoint inhibitors [ICI] [n = 4]) were included in the quantitative analyses. PARPi monotherapy improved radiographic PFS and OS over SoC in mCRPC patients with alterations in BRCA1 or BRCA2 genes but not in those with alterations in the ATM gene. Higher rates of PSA50 and ORR were reported in participants treated with PARPi + ARSI than in single-agent PARPi or PARPi + ICI. Although the rate of high-grade adverse events was similar across all groups, treatment discontinuation was higher in patients treated with PARPi-based combinations than PARPi monotherapy. CONCLUSION The efficacy of PARPi is not uniform across mCRPC patients with alterations in DNA damage repair genes, and optimal patient selection remains a clinical challenge. No unexpected safety signals for this class of agents emerged from this analysis.
Collapse
Affiliation(s)
- Giovanni Maria Iannantuono
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States; Medical Oncology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Elias Chandran
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Charalampos S Floudas
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hyoyoung Choo-Wosoba
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Gisela Butera
- Division of Library Services, Office of Research Services, National Institutes of Health, Bethesda, MD, United States
| | - Mario Roselli
- Medical Oncology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - James L Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Fatima Karzai
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
17
|
Alameddine Z, Niazi MRK, Rajavel A, Behgal J, Keesari PR, Araji G, Mustafa A, Wei C, Jahangir A, Terjanian TO. A Meta-Analysis of Randomized Clinical Trials Assessing the Efficacy of PARP Inhibitors in Metastatic Castration-Resistant Prostate Cancer. Curr Oncol 2023; 30:9262-9275. [PMID: 37887569 PMCID: PMC10605202 DOI: 10.3390/curroncol30100669] [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: 09/20/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023] Open
Abstract
Prostate cancer ranks as the second most common malignancy in males. Prostate cancer progressing on androgen deprivation therapy (ADT) is castration-resistant prostate cancer (CRPC). Poly-ADP ribose polymerase (PARP) inhibitors (PARPis) have been at the forefront of the treatment of CRPC. We aim to better characterize the progression-free survival (PFS) and overall survival (OS) in metastatic CRPC patients treated with PARPis. A systemic review search was conducted using National Clinical Trial (NCT), PubMed, Embase, Scopus, and Central Cochrane Registry. The improvement in overall survival was statistically significant, favoring PARPis (hazard ratio (HR) 0.855; 95% confidence interval (CI) 0.752-0.974; p = 0.018). The improvement in progression-free survival was also statistically significant, with results favoring PARPis (HR 0.626; 95%CI 0.566-0.692; p = 0.000). In a subgroup analysis, similar results were observed where the efficacy of PARPis was evaluated in a subgroup of patients without homologous recombination repair (HRR) gene mutation, which showed improvement in PFS favoring PARPis (HR 0.747; 95%CI 0.0.637-0.877; p = 0.000). Our meta-analysis of seven RCTs showed that PARPis significantly increased PFS and OS when used with or without antihormonal agents like abiraterone or enzalutamide.
Collapse
Affiliation(s)
- Zakaria Alameddine
- Staten Island University Hospital, Staten Island, NY 10305, USA; (Z.A.); (M.R.K.N.); (A.R.); (J.B.); (P.R.K.); (G.A.); (A.M.); (C.W.)
| | - Muhammad Rafay Khan Niazi
- Staten Island University Hospital, Staten Island, NY 10305, USA; (Z.A.); (M.R.K.N.); (A.R.); (J.B.); (P.R.K.); (G.A.); (A.M.); (C.W.)
| | - Anisha Rajavel
- Staten Island University Hospital, Staten Island, NY 10305, USA; (Z.A.); (M.R.K.N.); (A.R.); (J.B.); (P.R.K.); (G.A.); (A.M.); (C.W.)
| | - Jai Behgal
- Staten Island University Hospital, Staten Island, NY 10305, USA; (Z.A.); (M.R.K.N.); (A.R.); (J.B.); (P.R.K.); (G.A.); (A.M.); (C.W.)
| | - Praneeth Reddy Keesari
- Staten Island University Hospital, Staten Island, NY 10305, USA; (Z.A.); (M.R.K.N.); (A.R.); (J.B.); (P.R.K.); (G.A.); (A.M.); (C.W.)
| | - Ghada Araji
- Staten Island University Hospital, Staten Island, NY 10305, USA; (Z.A.); (M.R.K.N.); (A.R.); (J.B.); (P.R.K.); (G.A.); (A.M.); (C.W.)
| | - Ahmad Mustafa
- Staten Island University Hospital, Staten Island, NY 10305, USA; (Z.A.); (M.R.K.N.); (A.R.); (J.B.); (P.R.K.); (G.A.); (A.M.); (C.W.)
| | - Chapman Wei
- Staten Island University Hospital, Staten Island, NY 10305, USA; (Z.A.); (M.R.K.N.); (A.R.); (J.B.); (P.R.K.); (G.A.); (A.M.); (C.W.)
| | - Abdullah Jahangir
- University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA;
| | - Terenig O Terjanian
- Staten Island University Hospital, Staten Island, NY 10305, USA; (Z.A.); (M.R.K.N.); (A.R.); (J.B.); (P.R.K.); (G.A.); (A.M.); (C.W.)
| |
Collapse
|
18
|
Le TK, Duong QH, Baylot V, Fargette C, Baboudjian M, Colleaux L, Taïeb D, Rocchi P. Castration-Resistant Prostate Cancer: From Uncovered Resistance Mechanisms to Current Treatments. Cancers (Basel) 2023; 15:5047. [PMID: 37894414 PMCID: PMC10605314 DOI: 10.3390/cancers15205047] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/26/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Prostate cancer (PC) is the second most common cancer in men worldwide. Despite recent advances in diagnosis and treatment, castration-resistant prostate cancer (CRPC) remains a significant medical challenge. Prostate cancer cells can develop mechanisms to resist androgen deprivation therapy, such as AR overexpression, AR mutations, alterations in AR coregulators, increased steroidogenic signaling pathways, outlaw pathways, and bypass pathways. Various treatment options for CRPC exist, including androgen deprivation therapy, chemotherapy, immunotherapy, localized or systemic therapeutic radiation, and PARP inhibitors. However, more research is needed to combat CRPC effectively. Further investigation into the underlying mechanisms of the disease and the development of new therapeutic strategies will be crucial in improving patient outcomes. The present work summarizes the current knowledge regarding the underlying mechanisms that promote CRPC, including both AR-dependent and independent pathways. Additionally, we provide an overview of the currently approved therapeutic options for CRPC, with special emphasis on chemotherapy, radiation therapy, immunotherapy, PARP inhibitors, and potential combination strategies.
Collapse
Affiliation(s)
- Thi Khanh Le
- Centre de Recherche en Cancérologie de Marseille—CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, 13009 Marseille, France; (T.K.L.); (Q.H.D.); (V.B.); (M.B.); (D.T.)
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
| | - Quang Hieu Duong
- Centre de Recherche en Cancérologie de Marseille—CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, 13009 Marseille, France; (T.K.L.); (Q.H.D.); (V.B.); (M.B.); (D.T.)
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
- Vietnam Academy of Science and Technology (VAST), University of Science and Technology of Hanoi (USTH), Hanoi 10000, Vietnam
| | - Virginie Baylot
- Centre de Recherche en Cancérologie de Marseille—CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, 13009 Marseille, France; (T.K.L.); (Q.H.D.); (V.B.); (M.B.); (D.T.)
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
| | - Christelle Fargette
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
- Department of Nuclear Medicine, La Timone University Hospital, Aix-Marseille University, 13005 Marseille, France
| | - Michael Baboudjian
- Centre de Recherche en Cancérologie de Marseille—CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, 13009 Marseille, France; (T.K.L.); (Q.H.D.); (V.B.); (M.B.); (D.T.)
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
- Department of Urology AP-HM, Aix-Marseille University, 13005 Marseille, France
| | - Laurence Colleaux
- Faculté de Médecine Timone, INSERM, MMG, U1251, Aix-Marseille University, 13385 Marseille, France;
| | - David Taïeb
- Centre de Recherche en Cancérologie de Marseille—CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, 13009 Marseille, France; (T.K.L.); (Q.H.D.); (V.B.); (M.B.); (D.T.)
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
- Department of Nuclear Medicine, La Timone University Hospital, Aix-Marseille University, 13005 Marseille, France
| | - Palma Rocchi
- Centre de Recherche en Cancérologie de Marseille—CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, 13009 Marseille, France; (T.K.L.); (Q.H.D.); (V.B.); (M.B.); (D.T.)
- European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, 13005 Marseille, France;
| |
Collapse
|
19
|
Yip SM, Morash C, Kolinsky MP, Kapoor A, Ong M, Selvarajah S, Nuk J, Compton K, Pouliot F, Lavallée LT, Khalaf DJ, Hamilton RJ, Gotto GT, Rendon RA, Antebi E, Hotte SJ, Malone S, Chi KN, Drachenberg DE, Saad F, Chan J, Ferrario C, Ko J, Shayegan B, Parimi S, So AI, Feifer A, Jansz K, Finch D, Chin JL, Osborne B, Ho KF, Galamo CD, Zardan A, Niazi T. Genetic testing practices among specialist physicians who treat prostate cancer A Canadian, cross-sectional survey. Can Urol Assoc J 2023; 17:326-336. [PMID: 37494316 PMCID: PMC10581730 DOI: 10.5489/cuaj.8403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
INTRODUCTION In patients with prostate cancer (PCa), the identification of an alteration in genes associated with homologous recombination repair (HRR) has implications for prognostication, optimization of therapy, and familial risk mitigation. The aim of this study was to assess the genomic testing landscape of PCa in Canada and to recommend an approach to offering germline and tumor testing for HRR-associated genes. METHODS The Canadian Genitourinary Research Consortium (GURC) administered a cross-sectional survey to a largely academic, multidisciplinary group of investigators across 22 GURC sites between January and June 2022. RESULTS Thirty-eight investigators from all 22 sites responded to the survey. Germline genetic testing was initiated by 34%, while 45% required a referral to a genetic specialist. Most investigators (82%) reported that both germline and tumor testing were needed, with 92% currently offering germline and 72% offering tissue testing to patients with advanced PCa. The most cited reasons for not offering testing were an access gap (50%), uncertainties around who to test and which genes to test, (33%) and interpreting results (17%). A majority reported that patients with advanced PCa (74-80%) should be tested, with few investigators testing patients with localized disease except when there is a family history of PCa (45-55%). CONCLUSIONS Canadian physicians with academic subspecialist backgrounds in genitourinary malignancies recognize the benefits of both germline and somatic testing in PCa; however, there are challenges in accessing testing across practices and specialties. An algorithm to reduce uncertainty for providers when ordering genetic testing for patients with PCa is proposed.
Collapse
Affiliation(s)
- Steven M Yip
- Tom Baker Cancer Centre, University of Calgary, Calgary, AB, Canada
| | | | | | - Anil Kapoor
- St Joseph's Healthcare, McMaster University, Hamilton, ON, Canada
| | - Michael Ong
- Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Shamini Selvarajah
- Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network (UHN), Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jennifer Nuk
- Hereditary Cancer Program, BC Cancer, Vancouver, BC, Canada
| | - Katie Compton
- Hereditary Cancer Program, BC Cancer, Vancouver, BC, Canada
| | - Frederic Pouliot
- Centre hospitalier universitaire de Québec, Université Laval, Quebec City, QC, Canada
| | - Luke T Lavallée
- Division of Urology, Department of Surgery, University of Ottawa and Ottawa Hospital Research Institute Epidemiology Program, Ottawa, ON, Canada
| | - Daniel J Khalaf
- Department of Medicine, Medical Oncology Division, BC Cancer, Vancouver Centre, University of British Columbia, Vancouver, BC, Canada
| | - Robert J Hamilton
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Geoffrey T Gotto
- Southern Alberta Institute of Urology, University of Calgary, Calgary, AB, Canada
| | - Ricardo A Rendon
- Queen Elizabeth II Health Sciences Centre, Dalhousie University, Halifax, NS, Canada
| | - Elie Antebi
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Shawn Malone
- The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Kim N Chi
- BC Cancer Agency, University of British Columbia, Vancouver, BC, Canada
| | - Darrel E Drachenberg
- Section of Urology, Department of Surgery, University of Manitoba, Winnipeg, MB, Canada
| | - Fred Saad
- Centre Hospitalier de l'Université de Montréal, University of Montreal, Montreal, QC, Canada
| | - Jonathan Chan
- Scarborough Health Network, University of Toronto, ON, Canada
| | | | - Jenny Ko
- Department of Medical Oncology, BC Cancer - Abbotsford, Abbotsford, BC, Canada
| | - Bobby Shayegan
- St Joseph's Healthcare, McMaster University, Hamilton, ON, Canada
| | - Sunil Parimi
- Department of Medical Oncology, BC Cancer-Victoria, BC, Canada
| | - Alan I So
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Andrew Feifer
- Division of Urology, Department of Surgery, University of Toronto, Toronto, ON Canada
- Institute for Better Health, Trillium Health Partners, Mississauga, ON Canada
| | - Kenneth Jansz
- Joseph Brant Hospital, McMaster University, Burlington, ON, Canada
| | - Daygen Finch
- Department of Medical Oncology, BC Cancer Agency - Centre for the Southern Interior, Kelowna, BC, Canada
| | - Joseph L Chin
- Department of Surgery (Urology), University of Western Ontario, London Health Sciences Center, London, ON, Canada
| | | | | | | | | | - Tamim Niazi
- Jewish General Hospital, McGill University, Montreal, QC, Canada
| |
Collapse
|
20
|
Gulliver C, Huss S, Semjonow A, Baillie GS, Hoffmann R. Loss of PDE4D7 expression promotes androgen independence, neuroendocrine differentiation and alterations in DNA repair: implications for therapeutic strategies. Br J Cancer 2023; 129:1462-1476. [PMID: 37740039 PMCID: PMC10628190 DOI: 10.1038/s41416-023-02417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Androgen signalling remains the seminal therapeutic approach for the management of advanced prostate cancer. However, most tumours eventually shift towards an aggressive phenotype, characterised by androgen independence and treatment resistance. The cyclic adenosine monophosphate (cAMP) pathway plays a crucial role in regulating various cellular processes, with the phosphodiesterase PDE4D7 being a vital modulator of cAMP signalling in prostate cancer cells. METHODS Using shRNA-mediated PDE4D7 knockdown in LNCaP cells and downstream analysis via RNA sequencing and phenotypic assays, we replicate clinical observations that diminished PDE4D7 expression promotes an aggressive prostate cancer phenotype. RESULTS Our study provides evidence that loss of PDE4D7 expression represents a pivotal switch driving the transition from an androgen-sensitive state to hormone unresponsiveness and neuroendocrine differentiation. In addition, we demonstrate that PDE4D7 loss affects DNA repair pathways, conferring resistance to poly ADP ribose polymerase (PARP) inhibitors. CONCLUSION Reinstating PDE4D7 expression sensitises prostate cancer cells to anti-androgens, DNA damage response inhibitors, and cytotoxic therapies. These findings provide significant insight into the regulatory role of PDE4D7 in the development of lethal prostate cancer and the potential of its modulation as a novel therapeutic strategy.
Collapse
Affiliation(s)
- Chloe Gulliver
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, G12 8TA, Scotland, UK.
| | - Sebastian Huss
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, 48149, Münster, Germany
| | - Axel Semjonow
- Prostate Center, University Hospital Münster, 48149, Münster, Germany
| | - George S Baillie
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, G12 8TA, Scotland, UK
| | - Ralf Hoffmann
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, G12 8TA, Scotland, UK.
- Oncology Solutions, Philips Research Europe, High Tech Campus 34, 5656AE, Eindhoven, The Netherlands.
| |
Collapse
|
21
|
Giesen A, Baekelandt L, Devlies W, Devos G, Dumez H, Everaerts W, Claessens F, Joniau S. Double trouble for prostate cancer: synergistic action of AR blockade and PARPi in non-HRR mutated patients. Front Oncol 2023; 13:1265812. [PMID: 37810962 PMCID: PMC10551452 DOI: 10.3389/fonc.2023.1265812] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Prostate cancer (PCa) is the most common cancer in men worldwide. Despite better and more intensive treatment options in earlier disease stages, a large subset of patients still progress to metastatic castration-resistant PCa (mCRPC). Recently, poly-(ADP-ribose)-polymerase (PARP)-inhibitors have been introduced in this setting. The TALAPRO-2 and PROpel trials both showed a marked benefit of PARPi in combination with an androgen receptor signaling inhibitor (ARSI), compared with an ARSI alone in both the homologous recombination repair (HRR)-mutated, as well as in the HRR-non-mutated subgroup. In this review, we present a comprehensive overview of how maximal AR-blockade via an ARSI in combination with a PARPi has a synergistic effect at the molecular level, leading to synthetic lethality in both HRR-mutated and HRR-non-mutated PCa patients. PARP2 is known to be a cofactor of the AR complex, needed for decompacting the chromatin and start of transcription of AR target genes (including HRR genes). The inhibition of PARP thus reinforces the effect of an ARSI. The deep androgen deprivation caused by combining androgen deprivation therapy (ADT) with an ARSI, induces an HRR-like deficient state, often referred to as "BRCA-ness". Further, PARPi will prevent the repair of single-strand DNA breaks, leading to the accumulation of DNA double-strand breaks (DSBs). Due to the induced HRR-deficient state, DSBs cannot be repaired, leading to apoptosis.
Collapse
Affiliation(s)
- Alexander Giesen
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Loïc Baekelandt
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Wout Devlies
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, Catholic University Leuven (KU Leuven), Leuven, Belgium
| | - Gaëtan Devos
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Herlinde Dumez
- Department of Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Wouter Everaerts
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Frank Claessens
- Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, Catholic University Leuven (KU Leuven), Leuven, Belgium
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, Catholic University Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
22
|
Chi KN, Sandhu S, Smith MR, Attard G, Saad M, Olmos D, Castro E, Roubaud G, Pereira de Santana Gomes AJ, Small EJ, Rathkopf DE, Gurney H, Jung W, Mason GE, Dibaj S, Wu D, Diorio B, Urtishak K, Del Corral A, Francis P, Kim W, Efstathiou E. Niraparib plus abiraterone acetate with prednisone in patients with metastatic castration-resistant prostate cancer and homologous recombination repair gene alterations: second interim analysis of the randomized phase III MAGNITUDE trial. Ann Oncol 2023; 34:772-782. [PMID: 37399894 PMCID: PMC10849465 DOI: 10.1016/j.annonc.2023.06.009] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Patients with metastatic castration-resistant prostate cancer (mCRPC) and BRCA alterations have poor outcomes. MAGNITUDE found patients with homologous recombination repair gene alterations (HRR+), particularly BRCA1/2, benefit from first-line therapy with niraparib plus abiraterone acetate and prednisone (AAP). Here we report longer follow-up from the second prespecified interim analysis (IA2). PATIENTS AND METHODS Patients with mCRPC were prospectively identified as HRR+ with/without BRCA1/2 alterations and randomized 1 : 1 to niraparib (200 mg orally) plus AAP (1000 mg/10 mg orally) or placebo plus AAP. At IA2, secondary endpoints [time to symptomatic progression, time to initiation of cytotoxic chemotherapy, overall survival (OS)] were assessed. RESULTS Overall, 212 HRR+ patients received niraparib plus AAP (BRCA1/2 subgroup, n = 113). At IA2 with 24.8 months of median follow-up in the BRCA1/2 subgroup, niraparib plus AAP significantly prolonged radiographic progression-free survival {rPFS; blinded independent central review; median rPFS 19.5 versus 10.9 months; hazard ratio (HR) = 0.55 [95% confidence interval (CI) 0.39-0.78]; nominal P = 0.0007} consistent with the first prespecified interim analysis. rPFS was also prolonged in the total HRR+ population [HR = 0.76 (95% CI 0.60-0.97); nominal P = 0.0280; median follow-up 26.8 months]. Improvements in time to symptomatic progression and time to initiation of cytotoxic chemotherapy were observed with niraparib plus AAP. In the BRCA1/2 subgroup, the analysis of OS with niraparib plus AAP demonstrated an HR of 0.88 (95% CI 0.58-1.34; nominal P = 0.5505); the prespecified inverse probability censoring weighting analysis of OS, accounting for imbalances in subsequent use of poly adenosine diphosphate-ribose polymerase inhibitors and other life-prolonging therapies, demonstrated an HR of 0.54 (95% CI 0.33-0.90; nominal P = 0.0181). No new safety signals were observed. CONCLUSIONS MAGNITUDE, enrolling the largest BRCA1/2 cohort in first-line mCRPC to date, demonstrated improved rPFS and other clinically relevant outcomes with niraparib plus AAP in patients with BRCA1/2-altered mCRPC, emphasizing the importance of identifying this molecular subset of patients.
Collapse
Affiliation(s)
- K N Chi
- University of British Columbia, BC Cancer-Vancouver Center, Vancouver, Canada.
| | - S Sandhu
- Peter MacCallum Cancer Center, Melbourne, Australia; University of Melbourne, Melbourne, Australia
| | - M R Smith
- Massachusetts General Hospital Cancer Center, Boston, USA; Harvard Medical School, Boston, USA
| | - G Attard
- University College London Cancer Institute, London, UK; University College London Hospitals, London, UK
| | - M Saad
- Department of Clinical Oncology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - D Olmos
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid
| | - E Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - G Roubaud
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | | | - E J Small
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco
| | - D E Rathkopf
- Memorial Sloan Kettering Cancer Center, New York, USA; Weill Cornell Medicine, New York, USA
| | - H Gurney
- Macquarie University, Macquarie Park, Australia
| | - W Jung
- Keimyung University Dongsan Hospital, Daegu, South Korea
| | - G E Mason
- Janssen Research & Development, LLC, Spring House
| | - S Dibaj
- Janssen Research & Development, LLC, San Diego
| | - D Wu
- Janssen Research & Development, LLC, Los Angeles
| | - B Diorio
- Janssen Research & Development, LLC, Titusville
| | - K Urtishak
- Janssen Research & Development, LLC, Spring House
| | | | - P Francis
- Janssen Research & Development, LLC, Bridgewater
| | - W Kim
- Janssen Research & Development, LLC, Los Angeles
| | | |
Collapse
|
23
|
Rivero Belenchón I, Congregado Ruiz CB, Saez C, Osman García I, Medina López RA. Parp Inhibitors and Radiotherapy: A New Combination for Prostate Cancer (Systematic Review). Int J Mol Sci 2023; 24:12978. [PMID: 37629155 PMCID: PMC10455664 DOI: 10.3390/ijms241612978] [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: 07/20/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
PARPi, in combination with ionizing radiation, has demonstrated the ability to enhance cellular radiosensitivity in different tumors. The rationale is that the exposure to radiation leads to both physical and biochemical damage to DNA, prompting cells to initiate three primary mechanisms for DNA repair. Two double-stranded DNA breaks (DSB) repair pathways: (1) non-homologous end-joining (NHEJ) and (2) homologous recombination (HR); and (3) a single-stranded DNA break (SSB) repair pathway (base excision repair, BER). In this scenario, PARPi can serve as radiosensitizers by leveraging the BER pathway. This mechanism heightens the likelihood of replication forks collapsing, consequently leading to the formation of persistent DSBs. Together, the combination of PARPi and radiotherapy is a potent oncological strategy. This combination has proven its efficacy in different tumors. However, in prostate cancer, there are only preclinical studies to support it and, recently, an ongoing clinical trial. The objective of this paper is to perform a review of the current evidence regarding the use of PARPi and radiotherapy (RT) in PCa and to give future insight on this topic.
Collapse
Affiliation(s)
- Inés Rivero Belenchón
- Urology and Nephrology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain; (I.O.G.); (R.A.M.L.)
- Biomedical Institute of Seville (IBIS), 41013 Seville, Spain;
| | - Carmen Belen Congregado Ruiz
- Urology and Nephrology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain; (I.O.G.); (R.A.M.L.)
- Biomedical Institute of Seville (IBIS), 41013 Seville, Spain;
| | - Carmen Saez
- Biomedical Institute of Seville (IBIS), 41013 Seville, Spain;
| | - Ignacio Osman García
- Urology and Nephrology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain; (I.O.G.); (R.A.M.L.)
- Biomedical Institute of Seville (IBIS), 41013 Seville, Spain;
| | - Rafael Antonio Medina López
- Urology and Nephrology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain; (I.O.G.); (R.A.M.L.)
- Biomedical Institute of Seville (IBIS), 41013 Seville, Spain;
| |
Collapse
|
24
|
Ferretti S, Mercinelli C, Marandino L, Litterio G, Marchioni M, Schips L. Metastatic Castration-Resistant Prostate Cancer: Insights on Current Therapy and Promising Experimental Drugs. Res Rep Urol 2023; 15:243-259. [PMID: 37396015 PMCID: PMC10312338 DOI: 10.2147/rru.s385257] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 06/15/2023] [Indexed: 07/04/2023] Open
Abstract
The therapeutic landscape of metastatic hormone sensitive and metastatic castration-resistant prostate cancer (mCRPC) is rapidly changing. We reviewed the current treatment options for mCRPC, with insights on new available therapeutic strategies. Chemotherapy with docetaxel or cabazitaxel (for patients progressing on docetaxel), as well as treatment with androgen receptor axis targeted therapies, and Radium-223 are well-established treatment options for patients with mCRPC. The advent of theragnostic in prostate cancer established Lutetium-177 (177Lu)-PSMA-617 as a new standard of care for PSMA-positive mCRPC previously treated with ARAT and taxane-based chemotherapy. Olaparib, a poly-ADP-ribose polymerase (PARP) inhibitor, is approved for selected patients with mCRPC progressed on ARATs and in combination with abiraterone acetate as first-line treatment for mCRPC. Immunotherapy showed limited efficacy in unselected patients with mCRPC and novel immunotherapy strategies need to be explored. The search for biomarkers is a growing field of interest in mCRPC, and predictive biomarkers are needed to support the choice of treatment and the development of tailored strategies.
Collapse
Affiliation(s)
- Simone Ferretti
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio University of Chieti, Urology Unit, Chieti, Italy
| | - Chiara Mercinelli
- Division of Experimental Oncology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Marandino
- Division of Experimental Oncology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Litterio
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio University of Chieti, Urology Unit, Chieti, Italy
| | - Michele Marchioni
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio University of Chieti, Urology Unit, Chieti, Italy
| | - Luigi Schips
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio University of Chieti, Urology Unit, Chieti, Italy
| |
Collapse
|
25
|
Groen L, Kloots I, Englert D, Seto K, Estafanos L, Smith P, Verhaegh GW, Mehra N, Schalken JA. Transcriptome Profiling of Circulating Tumor Cells to Predict Clinical Outcomes in Metastatic Castration-Resistant Prostate Cancer. Int J Mol Sci 2023; 24:ijms24109002. [PMID: 37240349 DOI: 10.3390/ijms24109002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/04/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The clinical utility of circulating tumor cells (CTC) as a non-invasive multipurpose biomarker is broadly recognized. The earliest methods for enriching CTCs from whole blood rely on antibody-based positive selection. The prognostic utility of CTC enumeration using positive selection with the FDA-approved CellSearchTM system has been demonstrated in numerous studies. The capture of cells with specific protein phenotypes does not fully represent cancer heterogeneity and therefore does not realize the prognostic potential of CTC liquid biopsies. To avoid this selection bias, CTC enrichment based on size and deformability may provide better fidelity, i.e., facilitate the characterization of CTCs with any phenotype. In this study, the recently FDA-approved Parsortix® technology was used to enrich CTCs from prostate cancer (PCa) patients for transcriptome analysis using HyCEADTM technology. A tailored PCa gene panel allowed us to stratify metastatic castration-resistant prostate cancer (mCRPC) patients with clinical outcomes. In addition, our findings suggest that targeted CTC transcriptome profiling may be predictive of therapy response.
Collapse
Affiliation(s)
- Levi Groen
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Iris Kloots
- Department of Medical Oncology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | | | - Kelly Seto
- ANGLE Biosciences Inc., Toronto, ON M9W 1B3, Canada
| | | | - Paul Smith
- ANGLE Biosciences Inc., Toronto, ON M9W 1B3, Canada
| | - Gerald W Verhaegh
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jack A Schalken
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
26
|
Somanath PR, Chernoff J, Cummings BS, Prasad SM, Homan HD. Targeting P21-Activated Kinase-1 for Metastatic Prostate Cancer. Cancers (Basel) 2023; 15:2236. [PMID: 37190165 PMCID: PMC10137274 DOI: 10.3390/cancers15082236] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 05/17/2023] Open
Abstract
Metastatic prostate cancer (mPCa) has limited therapeutic options and a high mortality rate. The p21-activated kinase (PAK) family of proteins is important in cell survival, proliferation, and motility in physiology, and pathologies such as infectious, inflammatory, vascular, and neurological diseases as well as cancers. Group-I PAKs (PAK1, PAK2, and PAK3) are involved in the regulation of actin dynamics and thus are integral for cell morphology, adhesion to the extracellular matrix, and cell motility. They also play prominent roles in cell survival and proliferation. These properties make group-I PAKs a potentially important target for cancer therapy. In contrast to normal prostate and prostatic epithelial cells, group-I PAKs are highly expressed in mPCA and PCa tissue. Importantly, the expression of group-I PAKs is proportional to the Gleason score of the patients. While several compounds have been identified that target group-I PAKs and these are active in cells and mice, and while some inhibitors have entered human trials, as of yet, none have been FDA-approved. Probable reasons for this lack of translation include issues related to selectivity, specificity, stability, and efficacy resulting in side effects and/or lack of efficacy. In the current review, we describe the pathophysiology and current treatment guidelines of PCa, present group-I PAKs as a potential druggable target to treat mPCa patients, and discuss the various ATP-competitive and allosteric inhibitors of PAKs. We also discuss the development and testing of a nanotechnology-based therapeutic formulation of group-I PAK inhibitors and its significant potential advantages as a novel, selective, stable, and efficacious mPCa therapeutic over other PCa therapeutics in the pipeline.
Collapse
Affiliation(s)
- Payaningal R. Somanath
- Department of Clinical & Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
- MetasTx LLC, Basking Ridge, NJ 07920, USA
| | - Jonathan Chernoff
- MetasTx LLC, Basking Ridge, NJ 07920, USA
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Brian S. Cummings
- MetasTx LLC, Basking Ridge, NJ 07920, USA
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Sandip M. Prasad
- Morristown Medical Center, Atlantic Health System, Morristown, NJ 07960, USA
| | | |
Collapse
|
27
|
Congregado Ruiz B, Rivero Belenchón I, Lendínez Cano G, Medina López RA. Strategies to Re-Sensitize Castration-Resistant Prostate Cancer to Antiandrogen Therapy. Biomedicines 2023; 11:biomedicines11041105. [PMID: 37189723 DOI: 10.3390/biomedicines11041105] [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: 02/17/2023] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023] Open
Abstract
Since prostate cancer (PCa) was described as androgen-dependent, the androgen receptor (AR) has become the mainstay of its systemic treatment: androgen deprivation therapy (ADT). Although, through recent years, more potent drugs have been incorporated, this chronic AR signaling inhibition inevitably led the tumor to an incurable phase of castration resistance. However, in the castration-resistant status, PCa cells remain highly dependent on the AR signaling axis, and proof of it is that many men with castration-resistant prostate cancer (CRPC) still respond to newer-generation AR signaling inhibitors (ARSis). Nevertheless, this response is limited in time, and soon, the tumor develops adaptive mechanisms that make it again nonresponsive to these treatments. For this reason, researchers are focused on searching for new alternatives to control these nonresponsive tumors, such as: (1) drugs with a different mechanism of action, (2) combination therapies to boost synergies, and (3) agents or strategies to resensitize tumors to previously addressed targets. Taking advantage of the wide variety of mechanisms that promote persistent or reactivated AR signaling in CRPC, many drugs explore this last interesting behavior. In this article, we will review those strategies and drugs that are able to resensitize cancer cells to previously used treatments through the use of "hinge" treatments with the objective of obtaining an oncological benefit. Some examples are: bipolar androgen therapy (BAT) and drugs such as indomethacin, niclosamide, lapatinib, panobinostat, clomipramine, metformin, and antisense oligonucleotides. All of them have shown, in addition to an inhibitory effect on PCa, the rewarding ability to overcome acquired resistance to antiandrogenic agents in CRPC, resensitizing the tumor cells to previously used ARSis.
Collapse
Affiliation(s)
- Belén Congregado Ruiz
- Urology and Nephrology Department, Biomedical Institute of Seville (IBIS), University Hospital Virgen del Rocío, 41013 Seville, Spain
| | - Inés Rivero Belenchón
- Urology and Nephrology Department, Biomedical Institute of Seville (IBIS), University Hospital Virgen del Rocío, 41013 Seville, Spain
| | - Guillermo Lendínez Cano
- Urology and Nephrology Department, Biomedical Institute of Seville (IBIS), University Hospital Virgen del Rocío, 41013 Seville, Spain
| | - Rafael Antonio Medina López
- Urology and Nephrology Department, Biomedical Institute of Seville (IBIS), University Hospital Virgen del Rocío, 41013 Seville, Spain
| |
Collapse
|
28
|
Tisseverasinghe S, Bahoric B, Anidjar M, Probst S, Niazi T. Advances in PARP Inhibitors for Prostate Cancer. Cancers (Basel) 2023; 15:1849. [PMID: 36980735 PMCID: PMC10046616 DOI: 10.3390/cancers15061849] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Poly-adenosine diphosphate-ribose polymerase plays an essential role in cell function by regulating apoptosis, genomic stability and DNA repair. PARPi is a promising drug class that has gained significant traction in the last decade with good outcomes in different cancers. Several trials have sought to test its effectiveness in metastatic castration resistant prostate cancer (mCRPC). We conducted a comprehensive literature review to evaluate the current role of PARPi in this setting. To this effect, we conducted queries in the PubMed, Embase and Cochrane databases. We reviewed and compared all major contemporary publications on the topic. In particular, recent phase II and III studies have also demonstrated the benefits of olaparib, rucaparib, niraparib, talazoparib in CRPC. Drug effectiveness has been assessed through radiological progression or overall response. Given the notion of synthetic lethality and potential synergy with other oncological therapies, several trials are looking to integrate PARPi in combined therapies. There remains ongoing controversy on the need for genetic screening prior to treatment initiation as well as the optimal patient population, which would benefit most from PARPi. PARPi is an important asset in the oncological arsenal for mCRPC. New combinations with PARPi may improve outcomes in earlier phases of prostate cancer.
Collapse
Affiliation(s)
| | - Boris Bahoric
- Department of Radiation Oncology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Maurice Anidjar
- Department of Urology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Stephan Probst
- Department of Nuclear Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Tamim Niazi
- Department of Radiation Oncology, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
29
|
Zhang Z, Diao L, Zhang C, Wang F, Guan X, Yao X. Use of PARP inhibitors in prostate cancer: from specific to broader application. Front Endocrinol (Lausanne) 2023; 14:1164067. [PMID: 37152924 PMCID: PMC10162014 DOI: 10.3389/fendo.2023.1164067] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Prostate cancer (PC) is one of the major health issues of elderly men in the word. It is showed that there were approximately 1.414 million patients with PC in 2020 worldwide, with a high mortality rate in metastatic cases. In the present choices of treatment in PC, androgen deprivation therapy has long been as a backbone of them. But the clinical outcomes of patients with metastatic castration-resistant prostate cancer (mCRPC) were not ideal because of their poor prognosis, more effective therapeutic approaches are still necessary to further improve this problem. Poly (ADP-ribose) polymerase (PARP) inhibitors lead to the single-strand DNA breaks and/or double-strand DNA breaks, and result in synthetic lethality in cancer cells with impaired homologous recombination genes. It is estimated that approximately 20~25% of patients with mCRPC have a somatic or germinal DNA damage repair gene mutation. Furthermore, in "BRCAness" cases, which has been used to describe as tumors that have not arisen from a germline BRCA1 or BRCA2 mutation, there were also a number of studies sought to extend these promising results of PARP inhibitors. It is worth noting that an interaction between androgen receptor signaling and synthetic lethality with PARP inhibitors has been proposed. In this review, we discussed the mechanism of action and clinical research of PARP inhibitors, which may benefit population from "specific" to the "all-comer" in patients with PC when combined with novel hormonal therapies.
Collapse
Affiliation(s)
- Zhenting Zhang
- Department of Genitourinary Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lei Diao
- Department of Genitourinary Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chao Zhang
- Department of Genitourinary Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Feifei Wang
- PBG China Medical, Pfizer Inc, Shanghai, China
| | - Xin Guan
- PBG China Medical, Pfizer Inc, Shanghai, China
| | - Xin Yao
- Department of Genitourinary Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- *Correspondence: Xin Yao,
| |
Collapse
|