1
|
Segalés L, Bellmunt J, Perera-Bel J, Vargas-Parra G, Juanpere N, López D, Rodriguez-Vida A, Colomo L, Cecchini L, Lloreta-Trull J, Yélamos J, Fumadó L, Hernández-Llodrà S. Prognostic Value of PARP1 and PARP2 Copy Number Alterations in Prostate Cancer. J Transl Med 2025; 105:104171. [PMID: 40210166 DOI: 10.1016/j.labinv.2025.104171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/14/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
PARP1/2 have overlapping yet nonredundant biological functions in DNA repair and androgen receptor-transcriptional regulation. Studies on PARP alterations in human tumors have yielded conflicting results. In prostate cancer (PCa), PARP1/2 protein overexpression has been related to androgen deprivation therapy resistance, biochemical recurrence, and progression to metastases. PARP inhibitors have been approved for treating metastatic castration-resistant PCa with homologous recombination repair gene mutations. However, the significance of PARP1/2 genomic alterations is not fully studied. We aimed to analyze PARP1/2 alterations in PCa, assess their value as prognostic markers, and explore their relevance for potential therapeutic stratification. PARP1/2 copy number status was evaluated in 121 PCa primary tumors using real-time PCR. In 29 of them, a regional pelvic lymph node involvement was also analyzed. BRCA1/2 somatic mutations were analyzed in 24 PCa cases. Relationship with clinicopathological features, progression to metastases, and prostate-specific antigen recurrence was assessed. PARP1 loss and PARP2 gain were detected in 34.7% and 32.2% of primary tumors, respectively, with a high frequency of co-occurrence (P < .001). Both alterations were statistically associated with locally advanced disease at the time of diagnosis (P = .036; P = .006), metastatic dissemination (P = .014; P = .003), and other aggressive clinicopathological characteristics (such as the presence of Gleason pattern 5, high-grade, and high-stage). Cases with exclusive PARP2 gain had the shortest time to prostate-specific antigen recurrence, whereas double wt patients displayed the best outcome (P = .007). In 29 paired primary tumors and regional pelvic lymph node involvement, PARP1 loss showed strong concordance (P = .001), whereas PARP2 gain did not (P = .411). In conclusion, loss of PARP1 and gain of PARP2 show strong co-occurrence and are associated with clinicopathological characteristics of aggressiveness. PARP2 alterations appear to have a particularly significant impact on disease prognosis. Furthermore, these data suggest that the analysis of PARP1/2 copy number status could be useful in predicting PCa outcomes. Its role in therapy warrants further evaluation.
Collapse
Affiliation(s)
- Laura Segalés
- Departament of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain
| | - Joaquim Bellmunt
- Hospital del Mar Research Institute, Barcelona, Spain; Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | | | - Nuria Juanpere
- Hospital del Mar Research Institute, Barcelona, Spain; Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - David López
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Alejo Rodriguez-Vida
- Hospital del Mar Research Institute, Barcelona, Spain; Department of Medical Oncology, Hospital del Mar, Barcelona, Spain
| | - Lluís Colomo
- Hospital del Mar Research Institute, Barcelona, Spain; Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Lluís Cecchini
- Hospital del Mar Research Institute, Barcelona, Spain; Department of Urology, Hospital del Mar, Barcelona, Spain
| | - Josep Lloreta-Trull
- Departament of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - José Yélamos
- Hospital del Mar Research Institute, Barcelona, Spain; Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Lluís Fumadó
- Hospital del Mar Research Institute, Barcelona, Spain; Department of Urology, Hospital del Mar, Barcelona, Spain
| | - Silvia Hernández-Llodrà
- Departament of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
2
|
Shaik R, Mounika V, Begum S, Rajkumar A, Mallikarjun B, Sri Harshini V, Kolure R, Sreevani B, Thakur S. Monoclonal Antibodies in Clinical Trials for Breast Cancer Treatment. Monoclon Antib Immunodiagn Immunother 2025; 44:17-39. [PMID: 40171653 DOI: 10.1089/mab.2024.0018] [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] [Indexed: 04/04/2025] Open
Abstract
One of the most potent therapeutic and diagnostic agents in contemporary medicine is the monoclonal antibody (mAb). mAbs can perform a variety of tasks in breast cancer (BC), including identifying and delivering therapeutic medications to targets, preventing cell development, and suppressing immune system inhibitors including directly attacking cancer cells. mAbs are one of the most effective therapeutic options, particularly for HER2, but they have not been well studied for their use in treating other forms of BC, particularly triple negative breast tumors. Bispecific and trispecific mAbs have created new opportunities for more targeted specific efficacy, which has a positive impact on the viability of antigen specificity. They are more versatile and effective than other forms of treatment, emerging as most popular option for treating BC. However, mAbs have a limit in treatment due to certain adverse effects, including fever, shaking, exhaustion, headache, nausea, and vomiting, as well as rashes, bleeding, and difficulty breathing. To examine the current and prospective future capacities of mAbs with regard to the detection and treatment of BC, the present review highlights advantages and disadvantages of mAb approach.
Collapse
Affiliation(s)
- Rahaman Shaik
- School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Varikuppala Mounika
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Shireen Begum
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Agolapu Rajkumar
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Bathurasi Mallikarjun
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Vollala Sri Harshini
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | - Rajini Kolure
- Department of Pharmacology, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| | | | - Sneha Thakur
- Department of Pharmacognosy, St. Pauls College of Pharmacy, Turkayamjal, Hyderabad-501510, India
| |
Collapse
|
3
|
Lin L, Li Z, Chen K, Shao Y, Li X. Uncovering somatic genetic drivers in prostate cancer through comprehensive genome-wide analysis. GeroScience 2025:10.1007/s11357-025-01623-8. [PMID: 40156736 DOI: 10.1007/s11357-025-01623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/16/2025] [Indexed: 04/01/2025] Open
Abstract
Given that hereditary prostate cancer (PCa) accounts for only a small fraction of PCa phenotypes, there is still a substantial journey ahead in exploring the somatic genetic drivers contributing to sporadic PCa. The expression quantitative trait loci (eQTLs) data were sourced from the GTEx dataset for prostate-specific genes, and the summary statistic information was collected for 5854 genes. Genetic associations with PCa were extracted from three well-established consortiums: the UK Biobank (9131 cases and 173,493 controls), the PRACTICAL study (79,148 cases and 61,106 controls), and the FinnGen cohort (13,216 cases and 119,948 controls). To prioritize potential causal targets, additional analysis, including the protein-protein interaction (PPI), The Cancer Genome Atlas (TCGA) dataset, and the single-cell-type expression analysis, was performed. Generally, a total of 150 common significant genes with the same causal association with PCa were identified. Out of the 150 genes examined, 67.33% (101/150) were found to have protein-coding functions, while only 30.67% (46/150) of these genes had prior mentions in the scientific literature. Notably, the analysis of the TCGA dataset showed that only 44.67% (67/150) of the genes produced consistent results with the Mendelian randomization (MR) analysis. Furthermore, the evaluation of single-cell RNA-seq data and colocalization analysis singled out MSMB as a critical gene associated with the occurrence of PCa. We pinpointed a range of prostate-specific genes that display causal associations with the onset of PCa. Among these, the MSMB gene emerged as a pivotal factor linked to PCa, demonstrating robust consistency across all four assessments, including the MR, TCGA dataset, single-cell RNA-seq data, and colocalization analysis. These findings provided fresh perspectives on the pathogenesis of PCa and presented potential targets for drug development.
Collapse
Affiliation(s)
- Lede Lin
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhen Li
- Department of Urology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Kai Chen
- Department of Urology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Yanxiang Shao
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Xiang Li
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
4
|
Cruz Garcia Villa P, Izunza Laisequilla A, Puga Ortega E, Alaez Verson C. Prevalence of DNA-Repair Gene mutations in Mexican men with prostate cancer. Actas Urol Esp 2024; 48:588-595. [PMID: 38735436 DOI: 10.1016/j.acuroe.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/07/2024] [Indexed: 05/14/2024]
Abstract
INTRODUCTION AND OBJECTIVE Mexico reported 26,742 new cases of prostate cancer in 2020. Different risk factors have been identified in the pathogenesis of prostate cancer. Among them, genetic factors and alterations or mutations in specific genes have been described in different ethnic groups worldwide. The aim of our study is to report the prevalence of germline DNA-repair gene mutations in Mexican patients with prostate cancer. MATERIAL AND METHOD We performed germline genetic testing in 50 patients with localized prostate cancer and 50 patients with metastatic prostate cancer. Demographic, clinical, and histopathological data were collected. RESULTS Thirty-seven germline mutations were identified in 32 patients. The most commonly affected genes were ATM in 6%, followed by FANCA (5%), and ATR (4%). BRCA2 mutations were identified in 3%. The frequency of mutations was higher in the metastatic group. DISCUSSION AND CONCLUSION The results of our study show different mutations from those reported in different populations or regions. The use of PARP inhibitors is indicated in patients with germline mutations, specifically BRCA2, showing improvement in overall survival and progression free survival. To our knowledge, this is the first study reporting the prevalence of mutations in DNA-repair genes in Mexican patients with prostate cancer.
Collapse
Affiliation(s)
- P Cruz Garcia Villa
- Servicio de Urología, Hospital Regional Lic. Adolfo López Mateos, ISSSTE, Ciudad de México, Mexico.
| | - A Izunza Laisequilla
- Servicio de Urología, Hospital Regional Lic. Adolfo López Mateos, ISSSTE, Ciudad de México, Mexico
| | - E Puga Ortega
- Servicio de Urología, Hospital Regional Lic. Adolfo López Mateos, ISSSTE, Ciudad de México, Mexico
| | - C Alaez Verson
- Departamento de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica, Ciudad de México, Mexico
| |
Collapse
|
5
|
Foley GR, Marthick JR, Lucas SE, Raspin K, Banks A, Stanford JL, Ostrander EA, FitzGerald LM, Dickinson JL. Germline Sequencing of DNA Damage Repair Genes in Two Hereditary Prostate Cancer Cohorts Reveals New Disease Risk-Associated Gene Variants. Cancers (Basel) 2024; 16:2482. [PMID: 39001544 PMCID: PMC11240467 DOI: 10.3390/cancers16132482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Rare, inherited variants in DNA damage repair (DDR) genes have a recognised role in prostate cancer (PrCa) susceptibility. In addition, these genes are therapeutically targetable. While rare variants are informing clinical management in other common cancers, defining the rare disease-associated variants in PrCa has been challenging. Here, whole-genome and -exome sequencing data from two independent, high-risk Australian and North American familial PrCa datasets were interrogated for novel DDR risk variants. Rare DDR gene variants (predicted to be damaging and present in two or more family members) were identified and subsequently genotyped in 1963 individuals (700 familial and 459 sporadic PrCa cases, 482 unaffected relatives, and 322 screened controls), and association analyses accounting for relatedness (MQLS) undertaken. In the combined datasets, rare ERCC3 (rs145201970, p = 2.57 × 10-4) and BRIP1 (rs4988345, p = 0.025) variants were significantly associated with PrCa risk. A PARP2 (rs200603922, p = 0.028) variant in the Australian dataset and a MUTYH (rs36053993, p = 0.031) variant in the North American dataset were also associated with risk. Evaluation of clinicopathological characteristics provided no evidence for a younger age or higher-grade disease at diagnosis in variant carriers, which should be taken into consideration when determining genetic screening eligibility criteria for targeted, gene-based treatments in the future. This study adds valuable knowledge to our understanding of PrCa-associated DDR genes, which will underpin effective clinical screening and treatment strategies.
Collapse
Affiliation(s)
- Georgea R Foley
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - James R Marthick
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Sionne E Lucas
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Kelsie Raspin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Annette Banks
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Janet L Stanford
- Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., M4-B874, Seattle, WA 98109, USA
| | - Elaine A Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Liesel M FitzGerald
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Joanne L Dickinson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| |
Collapse
|
6
|
Sivaganesh V, Ta TM, Peethambaran B. Pentagalloyl Glucose (PGG) Exhibits Anti-Cancer Activity against Aggressive Prostate Cancer by Modulating the ROR1 Mediated AKT-GSK3β Pathway. Int J Mol Sci 2024; 25:7003. [PMID: 39000112 PMCID: PMC11241829 DOI: 10.3390/ijms25137003] [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: 05/09/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Androgen-receptor-negative, androgen-independent (ARneg-AI) prostate cancer aggressively proliferates and metastasizes, which makes treatment difficult. Hence, it is necessary to continue exploring cancer-associated markers, such as oncofetal Receptor Tyrosine Kinase like Orphan Receptor 1 (ROR1), which may serve as a form of targeted prostate cancer therapy. In this study, we identify that Penta-O-galloyl-β-D-glucose (PGG), a plant-derived gallotannin small molecule inhibitor, modulates ROR1-mediated oncogenic signaling and mitigates prostate cancer phenotypes. Results indicate that ROR1 protein levels were elevated in the highly aggressive ARneg-AI PC3 cancer cell line. PGG was selectively cytotoxic to PC3 cells and induced apoptosis of PC3 (IC50 of 31.64 µM) in comparison to normal prostate epithelial RWPE-1 cells (IC50 of 74.55 µM). PGG was found to suppress ROR1 and downstream oncogenic pathways in PC3 cells. These molecular phenomena were corroborated by reduced migration, invasion, and cell cycle progression of PC3 cells. PGG minimally and moderately affected RWPE-1 and ARneg-AI DU145, respectively, which may be due to these cells having lower levels of ROR1 expression in comparison to PC3 cells. Additionally, PGG acted synergistically with the standard chemotherapeutic agent docetaxel to lower the IC50 of both compounds about five-fold (combination index = 0.402) in PC3 cells. These results suggest that ROR1 is a key oncogenic driver and a promising target in aggressive prostate cancers that lack a targetable androgen receptor. Furthermore, PGG may be a selective and potent anti-cancer agent capable of treating ROR1-expressing prostate cancers.
Collapse
Affiliation(s)
- Vignesh Sivaganesh
- Department of Biology, Saint Joseph’s University, 600 S 43rd St, Philadelphia, PA 19104, USA; (V.S.); (T.M.T.)
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, 4170 City Ave, Philadelphia, PA 19131, USA
| | - Tram M. Ta
- Department of Biology, Saint Joseph’s University, 600 S 43rd St, Philadelphia, PA 19104, USA; (V.S.); (T.M.T.)
| | - Bela Peethambaran
- Department of Biology, Saint Joseph’s University, 600 S 43rd St, Philadelphia, PA 19104, USA; (V.S.); (T.M.T.)
| |
Collapse
|
7
|
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
|
8
|
Anselmino N, Labanca E, Shepherd PD, Dong J, Yang J, Song X, Nandakumar S, Kundra R, Lee C, Schultz N, Zhang J, Araujo JC, Aparicio AM, Subudhi SK, Corn PG, Pisters LL, Ward JF, Davis JW, Vazquez ES, Gueron G, Logothetis CJ, Futreal A, Troncoso P, Chen Y, Navone NM. Integrative Molecular Analyses of the MD Anderson Prostate Cancer Patient-derived Xenograft (MDA PCa PDX) Series. Clin Cancer Res 2024; 30:2272-2285. [PMID: 38488813 PMCID: PMC11094415 DOI: 10.1158/1078-0432.ccr-23-2438] [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: 08/15/2023] [Revised: 10/10/2023] [Accepted: 03/12/2024] [Indexed: 03/28/2024]
Abstract
PURPOSE Develop and deploy a robust discovery platform that encompasses heterogeneity, clinical annotation, and molecular characterization and overcomes the limited availability of prostate cancer models. This initiative builds on the rich MD Anderson (MDA) prostate cancer (PCa) patient-derived xenograft (PDX) resource to complement existing publicly available databases by addressing gaps in clinically annotated models reflecting the heterogeneity of potentially lethal and lethal prostate cancer. EXPERIMENTAL DESIGN We performed whole-genome, targeted, and RNA sequencing in representative samples of the same tumor from 44 PDXs derived from 38 patients linked to donor tumor metadata and corresponding organoids. The cohort includes models derived from different morphologic groups, disease states, and involved organ sites (including circulating tumor cells), as well as paired samples representing heterogeneity or stages before and after therapy. RESULTS The cohort recapitulates clinically reported alterations in prostate cancer genes, providing a data resource for clinical and molecular interrogation of suitable experimental models. Paired samples displayed conserved molecular alteration profiles, suggesting the relevance of other regulatory mechanisms (e.g., epigenomic) influenced by the microenvironment and/or treatment. Transcriptomically, models were grouped on the basis of morphologic classification. DNA damage response-associated mechanisms emerged as differentially regulated between adenocarcinoma and neuroendocrine prostate cancer in a cross-interrogation of PDX/patient datasets. CONCLUSIONS We addressed the gap in clinically relevant prostate cancer models through comprehensive molecular characterization of MDA PCa PDXs, providing a discovery platform that integrates with patient data and benchmarked to therapeutically relevant consensus clinical groupings. This unique resource supports robust hypothesis generation and testing from basic, translational, and clinical perspectives.
Collapse
Affiliation(s)
- Nicolas Anselmino
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Estefania Labanca
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Peter D.A. Shepherd
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiabin Dong
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jun Yang
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaofei Song
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Subhiksha Nandakumar
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ritika Kundra
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cindy Lee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Nikolaus Schultz
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John C. Araujo
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ana M. Aparicio
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sumit K. Subudhi
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paul G. Corn
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Louis L. Pisters
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John F. Ward
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John W. Davis
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elba S. Vazquez
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Inflamación y Cáncer, Buenos Aires, Argentina
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Geraldine Gueron
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Inflamación y Cáncer, Buenos Aires, Argentina
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Christopher J. Logothetis
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Nora M. Navone
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
9
|
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
|
10
|
Zheng X, Lu T, Wu S, Lin X, Bai J, Chen X, Miao Q, Yan J, Jiang K, Zhang L, Zheng X, Wang H, Xu Y, Xiao W, Li C, Peng W, Ding J, Zhong Q, Zou Z, Yang S, Li Y, Chen S, Zhang Q, Yan J, Tang G, Cai Y, kang M, Mok TSK, Lin G. A novel approach to evaluation of tumor response for advanced pulmonary adenocarcinoma using the intertumoral heterogeneity response score. MedComm (Beijing) 2024; 5:e493. [PMID: 38463396 PMCID: PMC10924640 DOI: 10.1002/mco2.493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 01/19/2024] [Accepted: 01/28/2024] [Indexed: 03/12/2024] Open
Abstract
Treatment response and prognosis estimation in advanced pulmonary adenocarcinoma are challenged by the significant heterogeneity of the disease. The current Response Evaluation Criteria in Solid Tumors (RECIST) criteria, despite providing a basis for solid tumor response evaluation, do not fully encompass this heterogeneity. To better represent these nuances, we introduce the intertumoral heterogeneity response score (THRscore), a measure built upon and expanding the RECIST criteria. This retrospective study included patients with 3-10 measurable advanced lung adenocarcinoma lesions who underwent first-line chemotherapy or targeted therapy. The THRscore, derived from the coefficient of variation in size for each measurable tumor before and 4-6 weeks posttreatment, unveiled a correlation with patient outcomes. Specifically, a high THRscore was associated with shorter progression-free survival, lower tumor response rate, and a higher tumor mutation burden. These associations were further validated in an external cohort, confirming THRscore's effectiveness in stratifying patients based on progression risk and treatment response, and enhancing the utility of RECIST in capturing complex tumor behaviors in lung adenocarcinoma. These findings affirm the promise of THRscore as an enhanced tool for tumor response assessment in advanced lung adenocarcinoma, extending the RECIST criteria's utility.
Collapse
Affiliation(s)
- Xinlong Zheng
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Tao Lu
- Department of RadiologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Shiwen Wu
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Xiaoyan Lin
- Department of OncologyFujian Medical University Union HospitalFuzhouChina
| | - Jing Bai
- Department of ResearchGeneplus‐Beijing InstituteBeijingChina
| | - Xiaohui Chen
- Department of Thoracic SurgeryClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Qian Miao
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Jianqun Yan
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Kan Jiang
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Longfeng Zhang
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Xiaobing Zheng
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Haibo Wang
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Yiquan Xu
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Weijin Xiao
- Department of PathologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Cao Li
- Department of PathologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Wenying Peng
- The Second Department of OncologyYunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer CenterKunmingChina
| | - Jianming Ding
- Department of Radiation OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Qiaofeng Zhong
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Zihua Zou
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Shanshan Yang
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Yujing Li
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Sihui Chen
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Qiuyu Zhang
- Institute of ImmunotherapyFujian Medical UniversityFuzhouChina
| | - Jianfeng Yan
- College of ChemistryFuzhou UniversityFuzhouChina
| | - Guofeng Tang
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Yuandong Cai
- College of ChemistryFuzhou UniversityFuzhouChina
| | - Miao kang
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
| | - Tony S. K. Mok
- Department of Clinical OncologyState Key Laboratory of Translational OncologyChinese University of Hong KongShatin, Hong Kong Special Administrative RegionChina
| | - Gen Lin
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouChina
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fujian Cancer HospitalFuzhouChina
- Interdisciplinary Institute for Medical EngineeringFuzhou UniversityFuzhouChina
| |
Collapse
|
11
|
Kouroukli O, Bravou V, Giannitsas K, Tzelepi V. Tissue-Based Diagnostic Biomarkers of Aggressive Variant Prostate Cancer: A Narrative Review. Cancers (Basel) 2024; 16:805. [PMID: 38398199 PMCID: PMC10887410 DOI: 10.3390/cancers16040805] [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: 01/12/2024] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Prostate cancer (PC) is a common malignancy among elderly men, characterized by great heterogeneity in its clinical course, ranging from an indolent to a highly aggressive disease. The aggressive variant of prostate cancer (AVPC) clinically shows an atypical pattern of disease progression, similar to that of small cell PC (SCPC), and also shares the chemo-responsiveness of SCPC. The term AVPC does not describe a specific histologic subtype of PC but rather the group of tumors that, irrespective of morphology, show an aggressive clinical course, dictated by androgen receptor (AR) indifference. AR indifference represents an adaptive response to androgen deprivation therapy (ADT), driven by epithelial plasticity, an inherent ability of tumor cells to adapt to their environment by changing their phenotypic characteristics in a bi-directional way. The molecular profile of AVPC entails combined alterations in the tumor suppressor genes retinoblastoma protein 1 (RB1), tumor protein 53 (TP53), and phosphatase and tensin homolog (PTEN). The understanding of the biologic heterogeneity of castration-resistant PC (CRPC) and the need to identify the subset of patients that would potentially benefit from specific therapies necessitate the development of prognostic and predictive biomarkers. This review aims to discuss the possible pathophysiologic mechanisms of AVPC development and the potential use of emerging tissue-based biomarkers in clinical practice.
Collapse
Affiliation(s)
- Olga Kouroukli
- Department of Pathology, Evaggelismos General Hospital, 10676 Athens, Greece
| | - Vasiliki Bravou
- Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, 26504 Patras, Greece;
| | | | - Vasiliki Tzelepi
- Department of Pathology, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
12
|
Waseem M, Wang BD. Organoids: An Emerging Precision Medicine Model for Prostate Cancer Research. Int J Mol Sci 2024; 25:1093. [PMID: 38256166 PMCID: PMC10816550 DOI: 10.3390/ijms25021093] [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: 12/08/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
Prostate cancer (PCa) has been known as the most prevalent cancer disease and the second leading cause of cancer mortality in men almost all over the globe. There is an urgent need for establishment of PCa models that can recapitulate the progress of genomic landscapes and molecular alterations during development and progression of this disease. Notably, several organoid models have been developed for assessing the complex interaction between PCa and its surrounding microenvironment. In recent years, PCa organoids have been emerged as powerful in vitro 3D model systems that recapitulate the molecular features (such as genomic/epigenomic changes and tumor microenvironment) of PCa metastatic tumors. In addition, application of organoid technology in mechanistic studies (i.e., for understanding cellular/subcellular and molecular alterations) and translational medicine has been recognized as a promising approach for facilitating the development of potential biomarkers and novel therapeutic strategies. In this review, we summarize the application of PCa organoids in the high-throughput screening and establishment of relevant xenografts for developing novel therapeutics for metastatic, castration resistant, and neuroendocrine PCa. These organoid-based studies are expected to expand our knowledge from basic research to clinical applications for PCa diseases. Furthermore, we also highlight the optimization of PCa cultures and establishment of promising 3D organoid models for in vitro and in vivo investigations, ultimately facilitating mechanistic studies and development of novel clinical diagnosis/prognosis and therapies for PCa.
Collapse
Affiliation(s)
- Mohammad Waseem
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA;
| | - Bi-Dar Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA;
- Hormone Related Cancers Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| |
Collapse
|
13
|
Siegelmann-Danieli N, Neiman V, Bareket-Samish A, Berger R, Peretz A, Alapi H, Tsur E, Patalon T, Beller D, Rimler G, Chodick G, Shohat M. Whole exome germline sequencing in early-onset prostate cancer patients: Genomic findings and clinical outcomes. Prostate 2024; 84:39-46. [PMID: 37842866 DOI: 10.1002/pros.24622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 08/13/2023] [Accepted: 08/23/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Whole exome sequencing (WES) furthered our understanding of various tumors. We assessed the occurrence of germline likely pathogenic/pathogenic (LP/P) variants, disease features, and clinical outcomes in early-onset prostate cancer. METHODS This retrospective study (N = 134) included consecutive prostate cancer patients who donated blood samples for research purposes to the Kahn-Sagol-Maccabi biobank. Patients diagnosed at >65 years were excluded. Clinical characteristics were extracted from the medical records. Germline WES was performed with analysis reporting on oncogenetic, two immunogenic, and a secondary minimum list panels (121, 468, 76, and 59 genes, respectively). RESULTS Median age at diagnosis was 61 (range 46-65) years; 131 (98%) were diagnosed with local disease. The median follow-up time from diagnosis was 14 (range <1-25) years. Of the patients with local disease, 32 (24%) and 10 (8%) had biochemical and distant recurrences, respectively. Twenty-five patients (19%) had ≥1 additional cancer (excluding non-melanoma skin cancer), most frequently bladder (6), colorectal (5), and lymphoma (5). Seven (5%) deaths were reported, with only one related to prostate cancer. LP/P variants were identified in 8 patients (6%), all in genes from the oncogenetic panel: ATM, BRCA1 (in two patients), BRCA2 (in two patients), HOXB13, MUTYH, and MYH7. Of these eight patients, with a median follow-up of 7 years (range <1-15), two (25%) had biochemical recurrences, one had (12.5%) distant recurrence, and no deaths were reported. CONCLUSIONS In this cohort of 134 early-onset prostate cancer patients, we identified germline LP/P variants in an oncogenetic panel in 6% of participants, with no unique clinical outcome.
Collapse
Affiliation(s)
- Nava Siegelmann-Danieli
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Professional Medicine, Maccabi Healthcare Services, Tel Aviv, Israel
- KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Victoria Neiman
- Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | | | - Racheli Berger
- Genetic Institute, Maccabi Healthcare Services, Rehovot, Israel
| | - Asaf Peretz
- KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
- Tipa Biobank, KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Hillel Alapi
- KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Erez Tsur
- Genetic Institute, Maccabi Healthcare Services, Rehovot, Israel
| | - Tal Patalon
- KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
- Tipa Biobank, KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Daniella Beller
- Tipa Biobank, KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Galit Rimler
- Tipa Biobank, KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Gabriel Chodick
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Mordechai Shohat
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Genetic Institute, Maccabi Healthcare Services, Rehovot, Israel
- The Genomics Unit, Sheba Cancer Research Center, Sheba Medical Center, Ramat Gan, Israel
| |
Collapse
|
14
|
Lin Y, Jin X. Effect of ubiquitin protease system on DNA damage response in prostate cancer (Review). Exp Ther Med 2024; 27:33. [PMID: 38125344 PMCID: PMC10731405 DOI: 10.3892/etm.2023.12321] [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: 08/02/2023] [Accepted: 10/26/2023] [Indexed: 12/23/2023] Open
Abstract
Genomic instability is an essential hallmark of cancer, and cellular DNA damage response (DDR) defects drive tumorigenesis by disrupting genomic stability. Several studies have identified abnormalities in DDR-associated genes, and a dysfunctional ubiquitin-proteasome system (UPS) is the most common molecular event in metastatic castration-resistant prostate cancer (PCa). For example, mutations in Speckle-type BTB/POZ protein-Ser119 result in DDR downstream target activation deficiency. Skp2 excessive upregulation inhibits homologous recombination repair and promotes cell growth and migration. Abnormally high expression of a deubiquitination enzyme, ubiquitin-specific protease 12, stabilizes E3 ligase MDM2, which further leads to p53 degradation, causing DDR interruption and genomic instability. In the present review, the basic pathways of DDR, UPS dysfunction, and its induced DDR alterations mediated by genomic instability, and especially the potential application of UPS and DDR alterations as biomarkers and therapeutic targets in PCa treatment, were described.
Collapse
Affiliation(s)
- Yan Lin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
15
|
Januskevicius T, Vaicekauskaite I, Sabaliauskaite R, Matulevicius A, Vezelis A, Ulys A, Jarmalaite S, Jankevicius F. Germline DNA Damage Response Gene Mutations in Localized Prostate Cancer. MEDICINA (KAUNAS, LITHUANIA) 2023; 60:73. [PMID: 38256334 PMCID: PMC10820233 DOI: 10.3390/medicina60010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024]
Abstract
Background and Objectives: Germline DNA damage response (DDR) gene mutations correlate with increased prostate cancer (PCa) risk and a more aggressive form of the disease. DDR mutation testing is recommended for metastatic PCa cases, while eligible information about the mutations' burden in the early-stage localized PCa is still limited. This study is aimed at the prospective detection of DDR pathway mutations in cases with localized PCa and correlation with clinical, histopathological, and radiological data. A comparison to the previously assessed cohort of the advanced PCa was performed. Materials and Methods: Germline DDR gene mutations were assessed prospectively in DNA samples from 139 patients, using a five-gene panel (BRCA1, BRCA2, ATM, CHEK2, and NBN) targeted next-generation sequencing. Results: This study revealed an almost three-fold higher risk of localized PCa among mutation carriers as compared to non-carriers (OR 2.84 and 95% CI: 0.75-20.23, p = 0.16). The prevalence of germline DDR gene mutations in PCa cases was 16.8% (18/107) and they were detected only in cases with PI-RADS 4/5 lesions. BRCA1/BRCA2/ATM mutation carriers were 2.6 times more likely to have a higher (>1) cISUP grade group compared to those with a CHEK2 mutation (p = 0.27). However, the number of cISUP > 1-grade patients with a CHEK2 mutation was significantly higher in advanced PCa than in localized PCa: 66.67% vs. 23.08% (p = 0.047). Conclusions: The results of our study suggest the potential of genetic screening for selected DDR gene mutations for early identification of cases at risk of aggressive PCa.
Collapse
Affiliation(s)
- Tomas Januskevicius
- Clinic of Gastroenterology, Nephro-Urology and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, M. K. Ciurlionio St. 21/27, LT-03101 Vilnius, Lithuania
| | - Ieva Vaicekauskaite
- Laboratory of Genetic Diagnostic, National Cancer Institute, Santariskiu St. 1, LT-08406 Vilnius, Lithuania
- Division of Human Genome Research Centre, Institute of Biomedical Sciences, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
| | - Rasa Sabaliauskaite
- Laboratory of Genetic Diagnostic, National Cancer Institute, Santariskiu St. 1, LT-08406 Vilnius, Lithuania
- Division of Human Genome Research Centre, Institute of Biomedical Sciences, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
| | - Augustinas Matulevicius
- Division of Human Genome Research Centre, Institute of Biomedical Sciences, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
- Urology Centre, Vilnius University Hospital Santaros Klinikos, Santariskiu St. 2, LT-08661 Vilnius, Lithuania
| | - Alvydas Vezelis
- Oncourology Department, National Cancer Institute, Santariskiu St. 1, LT-08660 Vilnius, Lithuania
| | - Albertas Ulys
- Oncourology Department, National Cancer Institute, Santariskiu St. 1, LT-08660 Vilnius, Lithuania
| | - Sonata Jarmalaite
- Laboratory of Genetic Diagnostic, National Cancer Institute, Santariskiu St. 1, LT-08406 Vilnius, Lithuania
- Division of Human Genome Research Centre, Institute of Biomedical Sciences, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
| | - Feliksas Jankevicius
- Clinic of Gastroenterology, Nephro-Urology and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, M. K. Ciurlionio St. 21/27, LT-03101 Vilnius, Lithuania
- Urology Centre, Vilnius University Hospital Santaros Klinikos, Santariskiu St. 2, LT-08661 Vilnius, Lithuania
| |
Collapse
|
16
|
Sivaganesh V, Peethambaran B. Receptor tyrosine kinase-like orphan receptor 1 inhibitor strictinin exhibits anti-cancer properties against highly aggressive androgen-independent prostate cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:1188-1209. [PMID: 38213538 PMCID: PMC10784114 DOI: 10.37349/etat.2023.00192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/07/2023] [Indexed: 01/13/2024] Open
Abstract
Aim It is important to identify anti-cancer compounds that can inhibit specific molecular targets to eradicate androgen-receptor negative (ARneg), androgen-independent (AI) prostate cancer, which is an aggressive form of prostate cancer with limited treatment options. The goal of this study was to selectively target prostate cancer cells that have high levels of oncogenic protein Receptor tyrosine kinase-like orphan receptor 1 (ROR1) by using strictinin, a small molecule ROR1 inhibitor. Methods The methods performed in this study include western blots, methyl thiazolyl tetrazolium (MTT) proliferation assays, phosphatidylserine apoptosis assays, apoptosis flow cytometry (Annexin V, caspase 3/7), migration scratch assays, Boyden chamber invasion assays, and cell cycle flow cytometry. Results Strictinin was most lethal against PC3 [half-maximal drug inhibitory concentration (IC50) of 277.2 µmol/L], an ARneg-AI cell type that expresses the highest levels of ROR1. Strictinin inhibited ROR1 expression, downstream phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT)-glycogen synthase kinase 3beta (GSK3β) pro-survival signaling, and epithelial-to-mesenchymal transition markers in PC3 cells. Additionally, strictinin decreased PC3 cell migration and invasion, while increasing S-phase cell cycle arrest. In ARneg-AI DU145 cells, strictinin inhibited ROR1 expression and modulated downstream AKT-GSK3β signaling. Furthermore, strictinin exhibited anti-migratory, anti-invasive, but minimal pro-apoptotic effects in DU145 cells likely due to DU145 having less ROR1 expression in comparison to PC3 cells. Throughout the study, strictinin minimally impacted the phenotype of normal prostatic epithelial cells RWPE-1 (IC50 of 658.5 µmol/L). Strictinin was further identified as synergistic with docetaxel [combination index (CI) = 0.311] and the combination therapy was found to reduce the IC50 of strictinin to 38.71 µmol/L in PC3 cells. Conclusions ROR1 is an emerging molecular target that can be utilized for treating prostate cancer. The data from this study establishes strictinin as a potential therapeutic agent that targets ARneg-AI prostate cancer with elevated ROR1 expression to reduce the migration, invasion, cell cycle progression, and survival of prostate cancer.
Collapse
Affiliation(s)
- Vignesh Sivaganesh
- Department of Biology, Saint Joseph’s University, Philadelphia, PA 19104, USA
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA 19131, USA
| | - Bela Peethambaran
- Department of Biology, Saint Joseph’s University, Philadelphia, PA 19104, USA
| |
Collapse
|
17
|
Xia Z, Wu J, Li Y, Yuan X, Sun J, Lv C, Huang P. LncRNA TYMSOS is a novel prognostic biomarker associated with immune infiltration in prostate cancer. Am J Cancer Res 2023; 13:4531-4546. [PMID: 37970350 PMCID: PMC10636683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 08/21/2023] [Indexed: 11/17/2023] Open
Abstract
The long noncoding RNA thymidylate synthetase opposite strand (lncRNA TYMSOS) plays an important role in cancers; however, its impact on prostate cancer (PCa) is still unclear. By analyzing the online data, we found that lncRNA TYMSOS was highly expressed in PCa and associated with T stage, Gleason score, age, and primary therapy outcome. The results of the ROC curve showed that lncRNA TYMSOS has a significant diagnostic ability. Furthermore, Kaplan-Meier analyses suggested that lncRNA TYMSOS plays an important role in progression-free survival (PFS). Increased lncRNA TYMSOS expression was an independent risk factor correlated with PFS in PCa patients. GSEA and GSVA indicated that the lncRNA TYMSOS was involved in the cell cycle, neurodegenerative diseases, oxidative phosphorylation, spliceosomes, and adaptive immune system pathways. Additionally, lncRNA TYMSOS expression was also associated with immune cell infiltrates and tumor mutational burden in PCa. Functional experiments were further conducted, and we verified that lncRNA TYMSOS played an oncogenic role in regulating PCa aggressiveness. Specifically, silencing of lncRNA TYMSOS suppressed cell proliferation, division and epithelial-mesenchymal transition (EMT) but promoted cell apoptosis in PCa cells, and conversely, lncRNA TYMSOS overexpression had the opposite effects. In summary, our study revealed that the lncRNA TYMSOS could be a biomarker and therapeutic target in PCa and participate in tumor-immune cell infiltration.
Collapse
Affiliation(s)
- Zhongyou Xia
- Department of Urology, Nanchong Central Hospital, The Second Clinical College, North Sichuan Medical College (University)Nanchong 637000, Sichuan, China
| | - Ji Wu
- Department of Urology, Nanchong Central Hospital, The Second Clinical College, North Sichuan Medical College (University)Nanchong 637000, Sichuan, China
| | - Yunxiang Li
- Department of Urology, Nanchong Central Hospital, The Second Clinical College, North Sichuan Medical College (University)Nanchong 637000, Sichuan, China
| | - Xinzhu Yuan
- Department of Nephrology, Blood Purification Center, Nanchong Central Hospital, The Second Clinical College, North Sichuan College (University)Nanchong 637000, Sichuan, China
| | - Jing Sun
- Department of Urology, Nanchong Central Hospital, The Second Clinical College, North Sichuan Medical College (University)Nanchong 637000, Sichuan, China
| | - Chen Lv
- Department of Urology, Nanchong Central Hospital, The Second Clinical College, North Sichuan Medical College (University)Nanchong 637000, Sichuan, China
| | - Peng Huang
- Department of Urology, Guizhou Provincial People’s HospitalGuiyang 550002, Guizhou, China
| |
Collapse
|
18
|
Lawrence MG, Taylor RA, Cuffe GB, Ang LS, Clark AK, Goode DL, Porter LH, Le Magnen C, Navone NM, Schalken JA, Wang Y, van Weerden WM, Corey E, Isaacs JT, Nelson PS, Risbridger GP. The future of patient-derived xenografts in prostate cancer research. Nat Rev Urol 2023; 20:371-384. [PMID: 36650259 PMCID: PMC10789487 DOI: 10.1038/s41585-022-00706-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 01/19/2023]
Abstract
Patient-derived xenografts (PDXs) are generated by engrafting human tumours into mice. Serially transplantable PDXs are used to study tumour biology and test therapeutics, linking the laboratory to the clinic. Although few prostate cancer PDXs are available in large repositories, over 330 prostate cancer PDXs have been established, spanning broad clinical stages, genotypes and phenotypes. Nevertheless, more PDXs are needed to reflect patient diversity, and to study new treatments and emerging mechanisms of resistance. We can maximize the use of PDXs by exchanging models and datasets, and by depositing PDXs into biorepositories, but we must address the impediments to accessing PDXs, such as institutional, ethical and legal agreements. Through collaboration, researchers will gain greater access to PDXs representing diverse features of prostate cancer.
Collapse
Affiliation(s)
- Mitchell G Lawrence
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
- Melbourne Urological Research Alliance, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia.
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
- Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia.
| | - Renea A Taylor
- Melbourne Urological Research Alliance, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Georgia B Cuffe
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Lisa S Ang
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ashlee K Clark
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| | - David L Goode
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Laura H Porter
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Clémentine Le Magnen
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Department of Urology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Nora M Navone
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jack A Schalken
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | | | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - John T Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter S Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Gail P Risbridger
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
- Melbourne Urological Research Alliance, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia.
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
- Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia.
| |
Collapse
|
19
|
Kiełb P, Kowalczyk K, Gurwin A, Nowak Ł, Krajewski W, Sosnowski R, Szydełko T, Małkiewicz B. Novel Histopathological Biomarkers in Prostate Cancer: Implications and Perspectives. Biomedicines 2023; 11:1552. [PMID: 37371647 DOI: 10.3390/biomedicines11061552] [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: 03/29/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Prostate cancer (PCa) is the second most frequently diagnosed cancer in men. Despite the significant progress in cancer diagnosis and treatment over the last few years, the approach to disease detection and therapy still does not include histopathological biomarkers. The dissemination of PCa is strictly related to the creation of a premetastatic niche, which can be detected by altered levels of specific biomarkers. To date, the risk factors for biochemical recurrence include lymph node status, prostate-specific antigen (PSA), PSA density (PSAD), body mass index (BMI), pathological Gleason score, seminal vesicle invasion, extraprostatic extension, and intraductal carcinoma. In the future, biomarkers might represent another prognostic factor, as discussed in many studies. In this review, we focus on histopathological biomarkers (particularly CD169 macrophages, neuropilin-1, cofilin-1, interleukin-17, signal transducer and activator of transcription protein 3 (STAT3), LIM domain kinase 1 (LIMK1), CD15, AMACR, prostate-specific membrane antigen (PSMA), Appl1, Sortilin, Syndecan-1, and p63) and their potential application in decision making regarding the prognosis and treatment of PCa patients. We refer to studies that found a correlation between the levels of biomarkers and tumor characteristics as well as clinical outcomes. We also hypothesize about the potential use of histopathological markers as a target for novel immunotherapeutic drugs or targeted radionuclide therapy, which may be used as adjuvant therapy in the future.
Collapse
Affiliation(s)
- Paweł Kiełb
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Kamil Kowalczyk
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Adam Gurwin
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Łukasz Nowak
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Wojciech Krajewski
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Roman Sosnowski
- Department of Urogenital Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Tomasz Szydełko
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Bartosz Małkiewicz
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
20
|
Sorrentino C, Di Carlo E. Molecular Targeted Therapies in Metastatic Prostate Cancer: Recent Advances and Future Challenges. Cancers (Basel) 2023; 15:2885. [PMID: 37296848 PMCID: PMC10251915 DOI: 10.3390/cancers15112885] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Prostate cancer is the most frequent malignant tumor in men, and, despite the great improvements in survival in patients with localized cancer, the prognosis for metastatic disease remains poor. Novel molecular targeted therapies, which block specific molecules or signaling pathways in tumor cells or in their microenvironment, have shown encouraging results in metastatic castration-resistant prostate cancer. Among these therapeutic approaches, prostate-specific membrane antigen-targeted radionuclide therapies and DNA repair inhibitors represent the most promising ones, with some therapeutic protocols already approved by the FDA, whereas therapies targeting tumor neovascularization and immune checkpoint inhibitors have not yet demonstrated clear clinical benefits. In this review, the most relevant studies and clinical trials on this topic are illustrated and discussed, together with future research directions and challenges.
Collapse
Affiliation(s)
- Carlo Sorrentino
- Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Emma Di Carlo
- Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
21
|
Chang T, Lian Z, Ma S, Liang Z, Ma X, Wen X, Wang Y, Liu R. Combination with vorinostat enhances the antitumor activity of cisplatin in castration-resistant prostate cancer by inhibiting DNA damage repair pathway and detoxification of GSH. Prostate 2023; 83:470-486. [PMID: 36576015 DOI: 10.1002/pros.24479] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 11/25/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Like DNA methylation, histone modifications are considered important processes for epigenetic alterations in gene function, and abnormally high expression of histone deacetylases (HDACs) plays a key role in many human diseases. In addition to regulating the acetylation levels of histone and non-histone proteins and gene transcription, HDAC inhibitors as antitumor drugs can also affect the DNA damage repair (DDR) pathway in tumor cells. Prostate cancer (PCa) is one of the most heritable malignancies in which DDR pathway defects can be detected in a considerable proportion of cases. Such defects are more prevalent in castration-resistant prostate cancer (CRPC) and are highly enriched in metastatic lesions. There is currently evidence that DDR pathway-deficient PCa is associated with high-risk biological behaviors and response sensitivity to platinum-based chemotherapy. Platinum-based drugs have been used in multiple clinical trials as monotherapy or in combination with other chemotherapeutic agents for the treatment of CRPC. METHODS This study evaluated the combined anticancer effect of (cisplatin) CDDP and the HDAC inhibitors vorinostat (SAHA) on three androgen-dependent cell lines PC-3, DU-145, and C4-2B in vitro. The efficacy and safety of SAHA combined with CDDP in the treatment of CRPC were further verified through animal experiments. RESULTS The combination of the two drugs increases cytotoxic effects by increasing DNA damage. Our results showed that the SAHA could not only reduce the expression of homologous recombinant repair proteins BRCA2, BRCA1, PARP1, and RAD51, but also decrease enzymes that Reduce the key enzymes of GSH biosynthesis, GSS and GCLC, and GSTP1 which can catalyze the binding of GSH to cisplatin. The intracellular GSH level also decreased with the increase of SAHA concentration, at the same time, the content of intracellular Pt element. CONCLUSION The combination of CDDP and SAHA can produce synergistic anticancer effects in androgen-independent PCa cells in vitro and in vivo. Our results open up a new avenue for the effective treatment of CRPC. To optimize the chemotherapy regimen for patients with advanced PCa, it is necessary to further study the molecular mechanism of platinum drugs, HDAC inhibitors, and their combined action.
Collapse
Affiliation(s)
- Taihao Chang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhenpeng Lian
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Shenfei Ma
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhengxin Liang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xudong Ma
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xiaodong Wen
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yanming Wang
- Tianjin Key Laboratory of Molecular Drug Research, College of Pharmacy, Nankai University, Tianjin, China
| | - Ranlu Liu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
22
|
Lin J, Zhuo Y, Zhang Y, Liu R, Zhong W. Molecular predictors of metastasis in patients with prostate cancer. Expert Rev Mol Diagn 2023; 23:199-215. [PMID: 36860119 DOI: 10.1080/14737159.2023.2187289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
INTRODUCTION Prostate cancer is a serious threat to the health of older adults worldwide. The quality of life and survival time of patients sharply decline once metastasis occurs. Thus, early screening for prostate cancer is very advanced in developed countries. The detection methods used include Prostate-specific antigen (PSA) detection and digital rectal examination. However, the lack of universal access to early screening in some developing countries has resulted in an increased number of patients presenting with metastatic prostate cancer. In addition, the treatment methods for metastatic and localized prostate cancer are considerably different. In many patients, early-stage prostate cancer cells often metastasize due to delayed observation, negative PSA results, and delay in treatment time. Therefore, the identification of patients who are prone to metastasis is important for future clinical studies. AREAS COVERED this review introduced a large number of predictive molecules related to prostate cancer metastasis. These molecules involve the mutation and regulation of tumor cell genes, changes in the tumor microenvironment, and the liquid biopsy. EXPERT OPINION In next decade, PSMA PET/CT and liquid biopsy will be the excellent predicting tools, while 177 Lu- PSMA-RLT will be showed excellent anti-tumor efficacy in mPCa patients.
Collapse
Affiliation(s)
- Jundong Lin
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yangjia Zhuo
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yixun Zhang
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Ren Liu
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Weide Zhong
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
23
|
Catalano M, Generali D, Gatti M, Riboli B, Paganini L, Nesi G, Roviello G. DNA repair deficiency as circulating biomarker in prostate cancer. Front Oncol 2023; 13:1115241. [PMID: 36793600 PMCID: PMC9922904 DOI: 10.3389/fonc.2023.1115241] [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: 12/03/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
Deleterious aberrations in DNA repair genes are actionable in approximately 25% of metastatic castration-resistant prostate cancers (mCRPC) patients. Homology recombination repair (HRR) is the DNA damage repair (DDR) mechanism most frequently altered in prostate cancer; of note BRCA2 is the most frequently altered DDR gene in this tumor. Poly ADP-ribose polymerase inhibitors showed antitumor activity with a improvement in overall survival in mCRPC carrying somatic and/or germline alterations of HHR. Germline mutations are tested on peripheral blood samples using DNA extracted from peripheral blood leukocytes, while the somatic alterations are assessed by extracting DNA from a tumor tissue sample. However, each of these genetic tests have some limitations: the somatic tests are related to the sample availability and tumor heterogeneity, while the germline testing are mainly related to the inability to detect somatic HRR mutations. Therefore, the liquid biopsy, a non-invasive and easily repeatable test compared to tissue test, could identified somatic mutation detected on the circulating tumor DNA (ctDNA) extracted from a plasma. This approach should better represent the heterogeneity of the tumor compared to the primary biopsy and maybe helpful in monitoring the onset of potential mutations involved in treatment resistance. Furthermore, ctDNA may inform about timing and potential cooperation of multiple driver genes aberration guiding the treatment options in patients with mCRPC. However, the clinical use of ctDNA test in prostate cancer compared to blood and tissue testing are currently very limited. In this review, we summarize the current therapeutic indications in prostate cancer patients with DDR deficiency, the recommendation for germline and somatic-genomic testing in advanced PC and the advantages of the use liquid biopsy in clinical routine for mCRPC.
Collapse
Affiliation(s)
- Martina Catalano
- School of Human Health Sciences, University of Florence, Florence, Italy
| | - Daniele Generali
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital Trieste, Trieste, Italy
| | - Marta Gatti
- Servizio di Citogenetica e Genetica - Azienda Socio-Sanitaria Territoriale (ASST) di Cremona, Cremona, Italy
| | - Barbara Riboli
- Servizio di Citogenetica e Genetica - Azienda Socio-Sanitaria Territoriale (ASST) di Cremona, Cremona, Italy
| | - Leda Paganini
- Servizio di Citogenetica e Genetica - Azienda Socio-Sanitaria Territoriale (ASST) di Cremona, Cremona, Italy
| | - Gabriella Nesi
- Department of Health Sciences, University of Florence, Florence, Italy
| | | |
Collapse
|
24
|
DNA Damage Repair Defects and Targeted Radionuclide Therapies for Prostate Cancer: Does Mutation Really Matter? A Systematic Review. Life (Basel) 2022; 13:life13010055. [PMID: 36676004 PMCID: PMC9860912 DOI: 10.3390/life13010055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
The aim of the present review was to assess the impact of DNA damage repair (DDR) mutations on response and outcome of patients (pts) affected by advanced prostate cancer (PCa) submitted to radionuclide therapies with [223Ra]RaCl2 (223Ra-therapy) or prostate specific membrane antigen (PSMA) ligands. A systematic literature search according to PRISMA criteria was made by using two main databases. Only studies published up until to October 2022 in the English language with ≥10 enrolled patients were selected. Seven studies including 326 pts, of whom 201 (61.6%) harboring DDR defects, were selected. The majority of selected papers were retrospective and four out of seven (57.1%) had small sample size (<50 pts). Three out of seven (42.8%) studies reported a more favorable outcome (overall or progression free survival) after therapy with alpha emitters (223Ra-therapy or [225Ac]Ac-PSMA-617) in subjects with DDR defects with respect to those without mutations. In two studies employing alpha or beta emitters ([177Lu]/[225Ac]-PMSA), no significant benefit was registered in pts harboring DDR defects. In all but one paper, no significant difference in response rate was reported among pts with or without DDR mutations. Although preliminary and biased by the retrospective design, preliminary data suggest a trend towards a longer survival in PCa pts harboring DDR defects submitted to radionuclide targeted therapy with alpha emitters.
Collapse
|
25
|
Cui X, Yu H, Yao J, Li J, Li Z, Jiang Z. ncRNA-mediated overexpression of ubiquitin-specific proteinase 13 contributes to the progression of prostate cancer via modulating AR signaling, DNA damage repair and immune infiltration. BMC Cancer 2022; 22:1350. [PMID: 36564767 PMCID: PMC9784269 DOI: 10.1186/s12885-022-10424-7] [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: 06/18/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is a lethal form of prostate cancer, and the molecular mechanism driving mCRPC progression has not yet been fully elucidated. Immunotherapies such as chimeric antigen receptor, T-cell therapy and immune checkpoint blockade have exerted promising antitumor effects in hematological and solid tumor malignancies; however, no encouraging responses have been observed against mCRPC. The deubiquitinase USP13 functions as a tumor suppressor in many human cancers, as it sustains the protein stability of PTEN and TP53; however, its role in prostate cancer (PCa) and involvement in DNA damage and AR signaling remain unclear. In the current study, we explored the prognostic value of USP13 in PCa based on the TCGA database, and we analyzed the expression of USP13 in PCa tissues and adjacent normal tissues based on TCGA and our cohort. The results suggested that USP13 is overexpressed in PCa tumors and has the potential to be an independent biomarker for the overall survival of PCa patients. Additionally, enrichment analysis indicated that USP13 may participate in the AR pathway and PI3k/Wnt signaling, which are closely related to PCa progression. We also observed a significant correlation between the expression of USP13 and AR-related genes, DDR genes and mismatch repair genes based on the TCGA_PRAD dataset, which further supported the critical role of USP13 in AR activation and the DNA damage response of PCa. USP13 was also found to be enriched in protein neddylation, and expression of USP13 was significantly associated with infiltration of immune cells and expression of immunomodulators. Taken together, our study revealed a key role of USP13 in contributing to PCa progression by participating in multiple oncogenic signaling pathways, the DNA damage response and the immunosuppressive tumor microenvironment. Targeting USP13 may inhibit tumor growth and provide additional benefits in cooperation with DDR inhibitors and immunotherapy.
Collapse
Affiliation(s)
- Xiaolu Cui
- grid.412636.40000 0004 1757 9485Department of Urology, First hospital of China Medical University, Shenyang, 110001 China
| | - Hongyuan Yu
- grid.412636.40000 0004 1757 9485Department of Urology, First hospital of China Medical University, Shenyang, 110001 China
| | - Jinlong Yao
- grid.412636.40000 0004 1757 9485Department of Urology, First hospital of China Medical University, Shenyang, 110001 China
| | - Jinling Li
- grid.412636.40000 0004 1757 9485Department of Urology, First hospital of China Medical University, Shenyang, 110001 China
| | - Zhenhua Li
- grid.412467.20000 0004 1806 3501Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004 China
| | - Zhenming Jiang
- grid.412636.40000 0004 1757 9485Department of Urology, First hospital of China Medical University, Shenyang, 110001 China
| |
Collapse
|
26
|
Boopathi E, Birbe R, Shoyele SA, Den RB, Thangavel C. Bone Health Management in the Continuum of Prostate Cancer Disease. Cancers (Basel) 2022; 14:4305. [PMID: 36077840 PMCID: PMC9455007 DOI: 10.3390/cancers14174305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
Prostate cancer (PCa) is the second-leading cause of cancer-related deaths in men. PCa cells require androgen receptor (AR) signaling for their growth and survival. Androgen deprivation therapy (ADT) is the preferred treatment for patients with locally advanced and metastatic PCa disease. Despite their initial response to androgen blockade, most patients eventually will develop metastatic castration-resistant prostate cancer (mCRPC). Bone metastases are common in men with mCRPC, occurring in 30% of patients within 2 years of castration resistance and in >90% of patients over the course of the disease. Patients with mCRPC-induced bone metastasis develop lesions throughout their skeleton; the 5-year survival rate for these patients is 47%. Bone-metastasis-induced early changes in the bone that proceed the osteoblastic response in the bone matrix are monitored and detected via modern magnetic resonance and PET/CT imaging technologies. Various treatment options, such as targeting osteolytic metastasis with bisphosphonates, prednisone, dexamethasone, denosumab, immunotherapy, external beam radiation therapy, radiopharmaceuticals, surgery, and pain medications are employed to treat prostate-cancer-induced bone metastasis and manage bone health. However, these diagnostics and treatment options are not very accurate nor efficient enough to treat bone metastases and manage bone health. In this review, we present the pathogenesis of PCa-induced bone metastasis, its deleterious impacts on vital organs, the impact of metastatic PCa on bone health, treatment interventions for bone metastasis and management of bone- and skeletal-related events, and possible current and future therapeutic options for bone management in the continuum of prostate cancer disease.
Collapse
Affiliation(s)
- Ettickan Boopathi
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ruth Birbe
- Laboratory Medicine, Department of Pathology, Cooper University Health Care, Camden, NJ 08103, USA
| | - Sunday A. Shoyele
- Department of Pharmaceutical Sciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Robert B. Den
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Chellappagounder Thangavel
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Department of Dermatology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Department of Interdisciplinary Oncology, Department of Biochemistry & Molecular Biology, LSUHSC Stanley S. Scott Cancer Center, 1700 Tulane Ave, New Orleans, LA 70112, USA
| |
Collapse
|
27
|
White JA, Kaninjing ET, Adeniji KA, Jibrin P, Obafunwa JO, Ogo CN, Mohammed F, Popoola A, Fatiregun OA, Oluwole OP, Karanam B, Elhussin I, Ambs S, Tang W, Davis M, Polak P, Campbell MJ, Brignole KR, Rotimi SO, Dean-Colomb W, Odedina FT, Martin DN, Yates C. Whole-exome Sequencing of Nigerian Prostate Tumors from the Prostate Cancer Transatlantic Consortium (CaPTC) Reveals DNA Repair Genes Associated with African Ancestry. CANCER RESEARCH COMMUNICATIONS 2022; 2:1005-1016. [PMID: 36922933 PMCID: PMC10010347 DOI: 10.1158/2767-9764.crc-22-0136] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/23/2022] [Accepted: 08/08/2022] [Indexed: 12/30/2022]
Abstract
In this study, we used whole-exome sequencing of a cohort of 45 advanced-stage, treatment-naïve Nigerian (NG) primary prostate cancer tumors and 11 unmatched nontumor tissues to compare genomic mutations with African American (AA) and European American (EA) The Cancer Genome Atlas (TCGA) prostate cancer. NG samples were collected from six sites in central and southwest Nigeria. After whole-exome sequencing, samples were processed using GATK best practices. BRCA1 (100%), BARD1 (45%), BRCA2 (27%), and PMS2(18%) had germline alterations in at least two NG nontumor samples. Across 111 germline variants, the AA cohort reflected a pattern [BRCA1 (68%), BARD1 (34%), BRCA2 (28%), and PMS2 (16%)] similar to NG samples. Of the most frequently mutated genes, BRCA1 showed a statistically (P ≤ 0.05) higher germline mutation frequency in men of African ancestry (MAA) and increasing variant frequency with increased African ancestry. Disaggregating gene-level mutation frequencies by variants revealed both ancestry-linked and NG-specific germline variant patterns. Driven by rs799917 (T>C), BRCA1 showed an increasing mutation frequency as African ancestry increased. BRCA2_rs11571831 was present only in MAA, and BRCA2_rs766173 was elevated in NG men. A total of 133 somatic variants were present in 26 prostate cancer-associated genes within the NG tumor cohort. BRCA2 (27%), APC (20%), ATM (20%), BRCA1 (13%), DNAJC6 (13%), EGFR (13%), MAD1L1 (13%), MLH1 (11%), and PMS2 (11%) showed mutation frequencies >10%. Compared with TCGA cohorts, NG tumors showed statistically significant elevated frequencies of BRCA2, APC, and BRCA1. The NG cohort variant pattern shared similarities (cosign similarities ≥0.734) with Catalogue of Somatic Mutations in Cancer signatures 5 and 6, and mutated genes showed significant (q < 0.001) gene ontology (GO) and functional enrichment in mismatch repair and non-homologous repair deficiency pathways. Here, we showed that mutations in DNA damage response genes were higher in NG prostate cancer samples and that a portion of those mutations correlate with African ancestry. Moreover, we identified variants of unknown significance that may contribute to population-specific routes of tumorigenesis and treatment. These results present the most comprehensive characterization of the NG prostate cancer exome to date and highlight the need to increase diversity of study populations. Significance MAA have higher rates of prostate cancer incidence and mortality, however, are severely underrepresented in genomic studies. This is the first study utilizing whole-exome sequencing in NG men to identify West African ancestry-linked variant patterns that impact DNA damage repair pathways.
Collapse
Affiliation(s)
- Jason A White
- Tuskegee University, Center for Cancer Research, Tuskegee, Alabama
| | | | | | | | - John O Obafunwa
- Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | | | | | | | | | | | | | - Isra Elhussin
- Tuskegee University, Center for Cancer Research, Tuskegee, Alabama
| | - Stefan Ambs
- Molecular Epidemiology Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Wei Tang
- Molecular Epidemiology Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Melissa Davis
- Department of Surgery, New York Presbyterian - Weill Cornell Medicine, New York, New York
| | | | - Moray J Campbell
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | | | | | - Windy Dean-Colomb
- Tuskegee University, Center for Cancer Research, Tuskegee, Alabama.,Piedmont Medical Oncology - Newnan, Newnan, Georgia
| | - Folake T Odedina
- Center for Health Equity and Community Engagement Research, Mayo Clinic, Jacksonville, Florida
| | - Damali N Martin
- Division of Cancer Control and Population Sciences, NCI, Rockville, Maryland
| | - Clayton Yates
- Tuskegee University, Center for Cancer Research, Tuskegee, Alabama
| |
Collapse
|
28
|
Congregado B, Rivero I, Osmán I, Sáez C, Medina López R. PARP Inhibitors: A New Horizon for Patients with Prostate Cancer. Biomedicines 2022; 10:1416. [PMID: 35740437 PMCID: PMC9220343 DOI: 10.3390/biomedicines10061416] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/16/2022] [Accepted: 05/23/2022] [Indexed: 11/23/2022] Open
Abstract
The introduction of PARP inhibitors (PARPi) in prostate cancer is a milestone and provides a pathway to hope in fighting this disease. It is the first time that drugs, based on the concept of synthetic lethality, have been approved for prostate cancer. In addition, it is also the first time that genetic mutation tests have been included in the therapeutic algorithm of this disease, representing a significant step forward for precision and personalized treatment of prostate cancer. The objectives of this review are: (1) understanding the mechanism of action of PARPi in monotherapy and combinations; (2) gaining insights on patient selection for PARPi; (3) exposing the pivotal studies that have allowed its approval, and; (4) offering an overview of the ongoing trials. Nevertheless, many unsolved questions remain, such as the number of patients who could potentially benefit from PARPi, whether to use PARPi in monotherapy or in combination, and when is the best time to use them in advanced or localized disease. To answer these and other questions, many clinical trials are underway. Some of them have recently demonstrated promising results that may favor the introduction of new combinations in metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Belén Congregado
- Urology and Nephrology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain; (I.R.); (I.O.); (R.M.L.)
| | - Inés Rivero
- Urology and Nephrology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain; (I.R.); (I.O.); (R.M.L.)
| | - Ignacio Osmán
- Urology and Nephrology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain; (I.R.); (I.O.); (R.M.L.)
| | - Carmen Sáez
- Department of Pathology, Biomedical Institute of Seville (IBIS), 41013 Seville, Spain;
| | - Rafael Medina López
- Urology and Nephrology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain; (I.R.); (I.O.); (R.M.L.)
| |
Collapse
|
29
|
Rizzo A, Mollica V, Merler S, Morelli F, Sorgentoni G, Oderda M, Santoni M, Massari F. Incidence of grade 3-4 adverse events, dose reduction, and treatment discontinuation in castration-resistant prostate cancer patients receiving PARP inhibitors: a meta-analysis. Expert Opin Drug Metab Toxicol 2022; 18:235-240. [PMID: 35485878 DOI: 10.1080/17425255.2022.2072727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND PARP inhibitors (PARPi) have recently emerged as a new treatment option for several solid tumors, including metastatic castration-resistant prostate cancer (mCRPC). However, several grade 3-4 adverse events have been reported during PARPi administration, leading to limitations in treatment adherence. METHODS Herein, we conducted a meta-analysis aimed at analyzing the incidence rate of commonly reported grade 3-4 adverse events, dose reduction, and treatment discontinuation in mCRPC patients treated with PARPi monotherapy. RESULTS Incidence rate with 95% confidence intervals (CIs) of grade 3-4 toxicities, dose reduction, and treatment discontinuation were calculated. Six trials involving 752 mCRPC patients were available for the meta-analysis. According to our results, anemia was the most frequently observed grade 3-4 toxicity (24.1%), and dose reduction (26.9%) and treatment discontinuation (14.1%) were common events during PARPi treatment. CONCLUSIONS Clinicians should carefully consider these risks, especially taking into account that the use of PARPi in mCRPC patients is expected to rise in the near future.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico Don Tonino Bello, I.R.C.C.S. Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Sara Merler
- U.O.C. Oncology, Azienda Ospedaliera Universitaria Integrata, University and Hospital Trust of Verona, Verona, Italy
| | - Franco Morelli
- Medical Oncology Unit, Gemelli Molise Hospital, Università Cattolica del Sacro Cuore, Campobasso, Italy
| | | | - Marco Oderda
- della Scienza, University of TurinDepartment of Urology, San Giovanni Battista Hospital, Città della Salute e , Turin, Italy
| | | | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
30
|
Yang F, Long N, Anekpuritanang T, Bottomly D, Savage JC, Lee T, Solis-Ruiz J, Borate U, Wilmot B, Tognon C, Bock AM, Pollyea DA, Radhakrishnan S, Radhakrishnan S, Patel P, Collins RH, Tantravahi S, Deininger MW, Fan G, Druker B, Shinde U, Tyner JW, Press RD, McWeeney S, Agarwal A. Identification and prioritization of myeloid malignancy germline variants in a large cohort of adult patients with AML. Blood 2022; 139:1208-1221. [PMID: 34482403 PMCID: PMC9211447 DOI: 10.1182/blood.2021011354] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/12/2021] [Indexed: 11/20/2022] Open
Abstract
Inherited predisposition to myeloid malignancies is more common than previously appreciated. We analyzed the whole-exome sequencing data of paired leukemia and skin biopsy samples from 391 adult patients from the Beat AML 1.0 consortium. Using the 2015 American College of Medical Genetics and Genomics (ACMG) guidelines for variant interpretation, we curated 1547 unique variants from 228 genes. The pathogenic/likely pathogenic (P/LP) germline variants were identified in 53 acute myeloid leukemia (AML) patients (13.6%) in 34 genes, including 6.39% (25/391) of patients harboring P/LP variants in genes considered clinically actionable (tier 1). 41.5% of the 53 patients with P/LP variants were in genes associated with the DNA damage response. The most frequently mutated genes were CHEK2 (8 patients) and DDX41 (7 patients). Pathogenic germline variants were also found in new candidate genes (DNAH5, DNAH9, DNMT3A, and SUZ12). No strong correlation was found between the germline mutational rate and age of AML onset. Among 49 patients who have a reported history of at least one family member affected with hematological malignancies, 6 patients harbored known P/LP germline variants and the remaining patients had at least one variant of uncertain significance, suggesting a need for further functional validation studies. Using CHEK2 as an example, we show that three-dimensional protein modeling can be one of the effective methodologies to prioritize variants of unknown significance for functional studies. Further, we evaluated an in silico approach that applies ACMG curation in an automated manner using the tool for assessment and (TAPES) prioritization in exome studies, which can minimize manual curation time for variants. Overall, our findings suggest a need to comprehensively understand the predisposition potential of many germline variants in order to enable closer monitoring for disease management and treatment interventions for affected patients and families.
Collapse
Affiliation(s)
- Fei Yang
- Department of Pathology and Laboratory Medicine and
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Nicola Long
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Tauangtham Anekpuritanang
- Department of Pathology and Laboratory Medicine and
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok
| | - Daniel Bottomly
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Division of Bioinformatics & Computational Biology and
| | - Jonathan C Savage
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR
| | - Tiffany Lee
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Jose Solis-Ruiz
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Uma Borate
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Beth Wilmot
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Division of Bioinformatics & Computational Biology and
| | - Cristina Tognon
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Allison M Bock
- Department of Medicine, University of Colorado, Aurora, CO
| | | | | | | | - Prapti Patel
- University of Texas Southwestern Medical Center, Dallas, TX
| | | | | | | | - Guang Fan
- Department of Pathology and Laboratory Medicine and
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Brian Druker
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Ujwal Shinde
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR
| | - Jeffrey W Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Department of Cell, Developmental & Cancer Biology
| | - Richard D Press
- Department of Pathology and Laboratory Medicine and
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Shannon McWeeney
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Division of Bioinformatics & Computational Biology and
| | - Anupriya Agarwal
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Department of Cell, Developmental & Cancer Biology
- Division of Hematology and Oncology, and
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR
| |
Collapse
|
31
|
PARP Inhibitors and Radiometabolic Approaches in Metastatic Castration-Resistant Prostate Cancer: What’s Now, What’s New, and What’s Coming? Cancers (Basel) 2022; 14:cancers14040907. [PMID: 35205654 PMCID: PMC8869833 DOI: 10.3390/cancers14040907] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Prostate cancer still represents an important health problem in men, considering its high frequency. Over the last decade, novel treatment options have emerged, leading to notable clinical benefits. These recent scientific acquisitions are creating the basis to widen the treatment scenario of this tumor, evolving from targeting the androgen receptor axis or the traditional chemotherapy approach. Abstract In recent years, the advances in the knowledge on the molecular characteristics of prostate cancer is allowing to explore novel treatment scenarios. Furthermore, technological discoveries are widening diagnostic and treatment weapons at the clinician disposal. Among these, great relevance is being gained by PARP inhibitors and radiometabolic approaches. The result is that DNA repair genes need to be altered in a high percentage of patients with metastatic prostate cancer, making these patients optimal candidates for PARP inhibitors. These compounds have already been proved to be active in pretreated patients and are currently being investigated in other settings. Radiometabolic approaches combine specific prostate cancer cell ligands to radioactive particles, thus allowing to deliver cytotoxic radiations in cancer cells. Among these, radium-223 and lutetium-177 have shown promising activity in metastatic pretreated prostate cancer patients and further studies are ongoing to expand the applications of this therapeutic approach. In addition, nuclear medicine techniques also have an important diagnostic role in prostate cancer. Herein, we report the state of the art on the knowledge on PARP inhibitors and radiometabolic approaches in advanced prostate cancer and present ongoing clinical trials that will hopefully expand these two treatment fields.
Collapse
|
32
|
Leith A, Ribbands A, Kim J, Last M, Barlow S, Yang L, Ghate SR. Real-world homologous recombination repair mutation testing in metastatic castration-resistant prostate cancer in the USA, Europe and Japan. Future Oncol 2022; 18:937-951. [DOI: 10.2217/fon-2021-1113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: To assess homologous recombination repair mutation (HRRm) testing patterns in metastatic castration-resistant prostate cancer. Methods: A point-in-time, international survey conducted January–August 2020. Results: Three-quarters of physicians (oncologists, urologists, specialist surgeons) globally reported access to genetic/genomic testing and just over half were HRRm testers. Surveyed physicians reported HRRm testing and positivity rates for 1913 patients, which were 18.1% and 33.7%, respectively. Of patients tested (n = 347), the most common HRR genes tested were BRCA (91.6%) and ATM (47.3%). Conclusion: Overall testing rates were low, with physicians mostly testing patients they considered higher risk. Increased awareness and education are needed to encourage broader testing, to understand familial risk and to identify patients with worse outcomes or those eligible for life-prolonging treatments.
Collapse
Affiliation(s)
| | | | - Jeri Kim
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | - Lingfeng Yang
- Merck & Co., Inc., Kenilworth, NJ, USA
- Employee at the time the study was conducted
| | | |
Collapse
|
33
|
Chi C, Liu J, Fan L, Zhu Y, Wang Y, Sha J, Huang Y, Dong B, Pan J, Xue W. Efficacy of neoadjuvant docetaxel + cisplatin chemo-hormonal therapy versus docetaxel chemo-hormonal therapy in patients with locally advanced prostate cancer with germline DNA damage repair gene alterations. Ther Adv Med Oncol 2022; 14:17588359221128356. [PMID: 36199621 PMCID: PMC9527989 DOI: 10.1177/17588359221128356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Purpose: To assess the efficacy and safety of neoadjuvant docetaxel + cisplatin chemotherapy with androgen deprivation therapy for the treatment of locally advanced prostate cancer (PCa) in patients harboring germline DNA damage repair genes (gDDR) defects. Methods: We conducted a prospective observational study in patients with locally advanced PCa confirmed with gDDR defects through next-generation sequencing. All patients received either docetaxel + cisplatin (platinum-group) or docetaxel chemo-hormonal therapy (docetaxel group) followed by radical prostatectomy with extended lymphadenectomy. The primary end point was biochemical progression-free survival (bPFS) and secondary end points include postoperative pathological response and safety assessment during the study period. Results: A total of 36 patients were included in the study, among whom 14 and 22 patients received docetaxel + cisplatin and docetaxel treatment, respectively. Down-staging of Tumor (T), Nodes (N), and Metastasis (M) stages was observed in 11 (78.57%) and 9 (40.9%) patients ( p = 0.041), respectively, in the docetaxel + cisplatin group and docetaxel group. The median bPFS was 7.76 months (95% CI 0.770–14.748) and not reached in the docetaxel group and docetaxel + cisplatin group, respectively. bPFS was significantly longer in the docetaxel + cisplatin group ( p = 0.039) with a hazard ratio of 0.386 (95% CI 0.151–0.987, p < 0.05). Furthermore, one patient discontinued docetaxel + cisplatin after second cycle due to severe liver insufficiency which was confirmed as viral hepatitis A and no significant perioperative complications was observed in either group. Conclusion: This study suggests that cisplatin may increase docetaxel anticancer activity with tolerable safety profile in patients with locally advanced PCa carrying gDDR defects in the neoadjuvant setting, a hypothesis which will require prospective, randomized confirmation.
Collapse
Affiliation(s)
- Chenfei Chi
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiazhou Liu
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liancheng Fan
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yinjie Zhu
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanqing Wang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Sha
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiran Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Baijun Dong
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Rd Pudong New Area, Shanghai, 200127, China
| | - Jiahua Pan
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Rd Pudong New Area, Shanghai, 200127, China
| | - Wei Xue
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Rd Pudong New Area, Shanghai, 200127, China
| |
Collapse
|
34
|
Ruiz de Porras V, Font A, Aytes A. Chemotherapy in metastatic castration-resistant prostate cancer: Current scenario and future perspectives. Cancer Lett 2021; 523:162-169. [PMID: 34517086 DOI: 10.1016/j.canlet.2021.08.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/03/2021] [Accepted: 08/25/2021] [Indexed: 01/04/2023]
Abstract
Taxanes - docetaxel and cabazitaxel - are the most active chemotherapy drugs currently used for the treatment of metastatic castration-resistant prostate cancer (mCRPC). However, despite a good initial response and survival benefit, nearly all patients eventually develop resistance, which is an important barrier to long-term survival. Resistance to taxanes is also associated with cross-resistance to androgen receptor signaling inhibitors (ARSIs). Unfortunately, other than platinum-based treatments, which have demonstrated some benefit in a subset of patients with Aggressive Variant Prostate Cancer (AVPC), few therapeutic options are available to patients progressing to taxanes. Hence, more research is required to determine whether platinum-based chemotherapy will confer a survival benefit in mCRPC, and the identification of predictive biomarkers and the clinical evaluation of platinum compounds in molecularly selected patients is an urgent but unmet clinical need. The present review focuses on the current status of chemotherapy treatments in mCRPC, interactions with androgen deprivation therapy (ADT) and novel ARSIs, and the main mechanisms of resistance. We will examine the impact of platinum-based treatments in mCRPC and summarize the known predictive biomarkers of platinum response. Finally, future approaches and avenues will be discussed.
Collapse
Affiliation(s)
- Vicenç Ruiz de Porras
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain; Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (BARGO), Badalona, Spain.
| | - Albert Font
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (BARGO), Badalona, Spain; Department of Medical Oncology, Catalan Institute of Oncology, Badalona, Spain
| | - Alvaro Aytes
- Program of Molecular Mechanisms and Experimental Therapeutics in Oncology (ONCOBELL), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de Llobregat, Gran Via de L'Hospitalet, Barcelona, Spain; Program Against Cancer Therapeutics Resistance (ProCURE), Catalan Institute of Oncology, Gran Via de L'Hospitalet, Barcelona, Spain.
| |
Collapse
|
35
|
Abstract
ABSTRACT In May 2020, the poly(ADP-ribose) polymerase (PARP) inhibitors rucaparib and olaparib were Food and Drug Administration approved for the management of metastatic castration-resistant prostate cancers. Rucaparib was approved for tumors that harbor alterations in BRCA1 and BRCA2 following progression on chemotherapy and androgen receptor-directed therapy, whereas olaparib was approved for tumors that harbor alterations in a broader range of DNA damage repair genes following progression on androgen receptor-directed therapy. Loss-of-function mutations in genes such as BRCA1 and BRCA2 increase reliance on PARP-mediated mechanisms of DNA repair, and inhibition of this pathway results in the accumulation of lethal levels of DNA damage. This dependence is advantageous in the management of prostate cancer, as mutations in DNA damage repair genes are frequent. This review summarizes the role of PARP in cell homeostasis, methods of targeting PARP in cancer cells, and current clinical trials in the management of advanced prostate cancer with PARP inhibitors.
Collapse
|
36
|
Ottini A, Sepe P, Beninato T, Claps M, Guadalupi V, Verzoni E, Giannatempo P, Baciarello G, de Braud F, Procopio G. Biomarker-driven immunotherapy for precision medicine in prostate cancer. Per Med 2021; 19:51-66. [PMID: 34873959 DOI: 10.2217/pme-2021-0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Although immunotherapy has recently revolutionized standard of care in different cancer types, prostate cancer has generally failed to show dramatic responses to immune checkpoint inhibitors. As in other tumors, the goal in prostate cancer is now to target treatments more precisely on patient's individual characteristics through precision medicine. Defects in mismatch repair, mutations in the exonuclease domain of the DNA polymerase epsilon (POLE), high tumor mutational burden and the presence of biallelic loss of CDK12 among others, are predictive biomarkers of response to immunotherapy. In the present review, we summarize the evolving landscape of immunotherapy in prostate cancer, including precision approaches and strategies to define classes of responsive patients and scale up resistance to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Arianna Ottini
- Department of Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Pierangela Sepe
- Department of Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Teresa Beninato
- Department of Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Mélanie Claps
- Department of Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Valentina Guadalupi
- Department of Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Verzoni
- Department of Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Patrizia Giannatempo
- Department of Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giulia Baciarello
- Department of Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppe Procopio
- Department of Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
37
|
Shah S, Rachmat R, Enyioma S, Ghose A, Revythis A, Boussios S. BRCA Mutations in Prostate Cancer: Assessment, Implications and Treatment Considerations. Int J Mol Sci 2021; 22:12628. [PMID: 34884434 PMCID: PMC8657599 DOI: 10.3390/ijms222312628] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/20/2021] [Accepted: 11/21/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer ranks fifth in cancer-related mortality in men worldwide. DNA damage is implicated in cancer and DNA damage response (DDR) pathways are in place against this to maintain genomic stability. Impaired DDR pathways play a role in prostate carcinogenesis and germline or somatic mutations in DDR genes have been found in both primary and metastatic prostate cancer. Among these, BRCA mutations have been found to be especially clinically relevant with a role for germline or somatic testing. Prostate cancer with DDR defects may be sensitive to poly(ADP-ribose) polymerase (PARP) inhibitors which target proteins in a process called PARylation. Initially they were used to target BRCA-mutated tumor cells in a process of synthetic lethality. However, recent studies have found potential for PARP inhibitors in a variety of other genetic settings. In this review, we explore the mechanisms of DNA repair, potential for genomic analysis of prostate cancer and therapeutics of PARP inhibitors along with their safety profile.
Collapse
Affiliation(s)
- Sidrah Shah
- Department of Palliative Care, Guy’s and St Thomas’ Hospital, Great Maze Pond, London SE1 9RT, UK;
| | - Rachelle Rachmat
- Department of Radiology, Guy’s and St Thomas’ Hospital, Great Maze Pond, London SE1 9RT, UK;
| | - Synthia Enyioma
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.E.); (A.R.)
| | - Aruni Ghose
- Department of Medical Oncology, Barts Cancer Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, W Smithfield, London EC1A 7BE, UK;
- Faculty of Life Sciences & Medicine, King’s College London, London WC2R 2LS, UK
| | - Antonios Revythis
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.E.); (A.R.)
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.E.); (A.R.)
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 9RT, UK
- AELIA Organization, 9th Km Thessaloniki-Thermi, 57001 Thessaloniki, Greece
| |
Collapse
|
38
|
Lawrence MG, Porter LH, Choo N, Pook D, Grummet JP, Pezaro CJ, Sandhu S, Ramm S, Luu J, Bakshi A, Goode DL, Sanij E, Pearson RB, Hannan RD, Simpson KJ, Taylor RA, Risbridger GP, Furic L. CX-5461 Sensitizes DNA Damage Repair-proficient Castrate-resistant Prostate Cancer to PARP Inhibition. Mol Cancer Ther 2021; 20:2140-2150. [PMID: 34413130 DOI: 10.1158/1535-7163.mct-20-0932] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/19/2021] [Accepted: 06/30/2021] [Indexed: 11/16/2022]
Abstract
Monotherapy with PARP inhibitors is effective for the subset of castrate-resistant prostate cancer (CRPC) with defects in homologous recombination (HR) DNA repair. New treatments are required for the remaining tumors, and an emerging strategy is to combine PARP inhibitors with other therapies that induce DNA damage. Here we tested whether PARP inhibitors are effective for HR-proficient CRPC, including androgen receptor (AR)-null tumors, when used in combination with CX-5461, a small molecule that inhibits RNA polymerase I transcription and activates the DNA damage response, and has antitumor activity in early phase I trials. The combination of CX-5461 and talazoparib significantly decreased in vivo growth of patient-derived xenografts of HR-proficient CRPC, including AR-positive, AR-null, and neuroendocrine tumors. CX-5461 and talazoparib synergistically inhibited the growth of organoids and cell lines, and significantly increased the levels of DNA damage. Decreased tumor growth after combination therapy was maintained for 2 weeks without treatment, significantly increasing host survival. Therefore, combination treatment with CX-5461 and talazoparib is effective for HR-proficient tumors that are not suitable for monotherapy with PARP inhibitors, including AR-null CRPC. This expands the spectrum of CRPC that is sensitive to PARP inhibition.
Collapse
Affiliation(s)
- Mitchell G Lawrence
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.,Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Laura H Porter
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Nicholas Choo
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - David Pook
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.,Medical Oncology, Monash Health, Clayton, Victoria, Australia
| | - Jeremy P Grummet
- Epworth Healthcare, Melbourne, Victoria, Australia.,Department of Surgery, Central Clinical School, Monash University, Clayton, Victoria, Australia.,Australian Urology Associates, Melbourne, VIC, Australia
| | - Carmel J Pezaro
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.,Eastern Health and Monash University Eastern Health Clinical School, Victoria, Australia.,University of Sheffield and Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Shahneen Sandhu
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Cancer Tissue Collection After Death (CASCADE) Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Susanne Ramm
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jennii Luu
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Andrew Bakshi
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - David L Goode
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Elaine Sanij
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Clinical Pathology, University of Melbourne, Parkville, Victoria, Australia.,St Vincent's Institute, Fitzroy, VIC, Australia
| | - Richard B Pearson
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Ross D Hannan
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia.,ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University, Australian Capital Territory, Australia.,School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Kaylene J Simpson
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Renea A Taylor
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Gail P Risbridger
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia. .,Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Luc Furic
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia. .,Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
39
|
Risbridger GP, Clark AK, Porter LH, Toivanen R, Bakshi A, Lister NL, Pook D, Pezaro CJ, Sandhu S, Keerthikumar S, Quezada Urban R, Papargiris M, Kraska J, Madsen HB, Wang H, Richards MG, Niranjan B, O'Dea S, Teng L, Wheelahan W, Li Z, Choo N, Ouyang JF, Thorne H, Devereux L, Hicks RJ, Sengupta S, Harewood L, Iddawala M, Azad AA, Goad J, Grummet J, Kourambas J, Kwan EM, Moon D, Murphy DG, Pedersen J, Clouston D, Norden S, Ryan A, Furic L, Goode DL, Frydenberg M, Lawrence MG, Taylor RA. The MURAL collection of prostate cancer patient-derived xenografts enables discovery through preclinical models of uro-oncology. Nat Commun 2021; 12:5049. [PMID: 34413304 PMCID: PMC8376965 DOI: 10.1038/s41467-021-25175-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Preclinical testing is a crucial step in evaluating cancer therapeutics. We aimed to establish a significant resource of patient-derived xenografts (PDXs) of prostate cancer for rapid and systematic evaluation of candidate therapies. The PDX collection comprises 59 tumors collected from 30 patients between 2012-2020, coinciding with availability of abiraterone and enzalutamide. The PDXs represent the clinico-pathological and genomic spectrum of prostate cancer, from treatment-naïve primary tumors to castration-resistant metastases. Inter- and intra-tumor heterogeneity in adenocarcinoma and neuroendocrine phenotypes is evident from bulk and single-cell RNA sequencing data. Organoids can be cultured from PDXs, providing further capabilities for preclinical studies. Using a 1 x 1 x 1 design, we rapidly identify tumors with exceptional responses to combination treatments. To govern the distribution of PDXs, we formed the Melbourne Urological Research Alliance (MURAL). This PDX collection is a substantial resource, expanding the capacity to test and prioritize effective treatments for prospective clinical trials in prostate cancer.
Collapse
Affiliation(s)
- Gail P Risbridger
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia. .,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
| | - Ashlee K Clark
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Laura H Porter
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Roxanne Toivanen
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew Bakshi
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Natalie L Lister
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - David Pook
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.,Department of Medical Oncology, Monash Health, Clayton, VIC, Australia
| | - Carmel J Pezaro
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Eastern Health and Monash University Eastern Health Clinical School, Box Hill, VIC, Australia.,Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, England
| | - Shahneen Sandhu
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Cancer Tissue Collection After Death (CASCADE) Program, Melbourne, VIC, Australia
| | - Shivakumar Keerthikumar
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Rosalia Quezada Urban
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Melissa Papargiris
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Australian Prostate Cancer Bioresource, VIC Node, Monash University, Clayton, VIC, Australia
| | - Jenna Kraska
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Australian Prostate Cancer Bioresource, VIC Node, Monash University, Clayton, VIC, Australia
| | - Heather B Madsen
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Australian Prostate Cancer Bioresource, VIC Node, Monash University, Clayton, VIC, Australia
| | - Hong Wang
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Michelle G Richards
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Birunthi Niranjan
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Samantha O'Dea
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Linda Teng
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - William Wheelahan
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Zhuoer Li
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Nicholas Choo
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - John F Ouyang
- Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Heather Thorne
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Lisa Devereux
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Rodney J Hicks
- Center for Molecular Imaging, Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | - Shomik Sengupta
- Eastern Health and Monash University Eastern Health Clinical School, Box Hill, VIC, Australia.,Department of Urology, Austin Hospital, The University of Melbourne, Heidelberg, VIC, Australia.,Department of Surgery, Austin Health, The University of Melbourne, Heidelberg, VIC, Australia.,Epworth Healthcare, Melbourne, VIC, Australia.,Epworth Freemasons, Epworth Health, East Melbourne, VIC, Australia
| | - Laurence Harewood
- Epworth Healthcare, Melbourne, VIC, Australia.,Department of Surgery, The University of Melbourne, Parkville, VIC, Australia
| | - Mahesh Iddawala
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Arun A Azad
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jeremy Goad
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Epworth Healthcare, Melbourne, VIC, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, VIC, Australia
| | - Jeremy Grummet
- Epworth Healthcare, Melbourne, VIC, Australia.,Department of Surgery, Central Clinical School, Monash University, Clayton, VIC, Australia.,Australian Urology Associates, Melbourne, VIC, Australia
| | - John Kourambas
- Department of Medicine, Monash Health, Casey Hospital, Berwick, VIC, Australia
| | - Edmond M Kwan
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.,Department of Medical Oncology, Monash Health, Clayton, VIC, Australia
| | - Daniel Moon
- Epworth Healthcare, Melbourne, VIC, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, VIC, Australia.,Australian Urology Associates, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Clayton, VIC, Australia.,The Epworth Prostate Centre, Epworth Hospital, Richmond, VIC, Australia
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Epworth Healthcare, Melbourne, VIC, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, VIC, Australia
| | - John Pedersen
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,TissuPath, Mount Waverley, VIC, Australia
| | | | - Sam Norden
- TissuPath, Mount Waverley, VIC, Australia
| | | | - Luc Furic
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - David L Goode
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Mark Frydenberg
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Epworth Healthcare, Melbourne, VIC, Australia.,Australian Urology Associates, Melbourne, VIC, Australia.,Department of Surgery, Monash University, Clayton, VIC, Australia.,Department of Urology, Cabrini Institute, Cabrini Health, Melbourne, VIC, Australia
| | - Mitchell G Lawrence
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Renea A Taylor
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia. .,Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Physiology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
40
|
Babasaki T, Sentani K, Sekino Y, Kobayashi G, Thang Pham Q, Katsuya N, Akabane S, Taniyama D, Hayashi T, Shiota M, Oue N, Teishima J, Matsubara A, Yasui W. Overexpression of claspin promotes docetaxel resistance and is associated with prostate-specific antigen recurrence in prostate cancer. Cancer Med 2021; 10:5574-5588. [PMID: 34240817 PMCID: PMC8366092 DOI: 10.1002/cam4.4113] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 12/18/2022] Open
Abstract
Although docetaxel (DTX) confers significant survival benefits in patients with castration‐resistant prostate cancer (CRPC), resistance to DTX inevitably occurs. Therefore, clarifying the mechanisms of DTX resistance may improve survival in patients with CRPC. Claspin plays a pivotal role in DNA replication stress and damage responses and is an essential regulator for the S‐phase checkpoint. CLSPN is an oncogenic gene that contributes to tumor proliferation in several human solid tumors. However, the clinical significance of claspin in prostate cancer (PCa) has not been examined. The present study aimed to elucidate the role of claspin and its relationship with DTX resistance in PCa. We immunohistochemically analyzed the expression of claspin in 89 PCa cases, of which 31 (35%) were positive for claspin. Claspin‐positive cases were associated with higher Gleason score, venous invasion, and perineural invasion. Kaplan–Meier analysis showed that high claspin expression was related to poor prostate‐specific antigen (PSA) relapse‐free prognosis. In a public database, high CLSPN expression was associated with poor PSA relapse‐free prognosis, Gleason score, T stage, lymph node metastasis, CRPC, and metastatic PCa. Claspin knockdown by siRNA decreased cell proliferation, upregulated DTX sensitivity, and suppressed the expression of Akt, Erk1/2, and CHK1 phosphorylation in DU145 and PC3 cell lines. Furthermore, claspin expression was much more upregulated in DTX‐resistant DU145 (DU145‐DR) than in parental DU145 cells. Claspin knockdown significantly upregulated the sensitivity to DTX in DU145‐DR cells. These results suggest that claspin plays an important role in PCa tumor progression and DTX resistance.
Collapse
Affiliation(s)
- Takashi Babasaki
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhiro Sentani
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yohei Sekino
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Go Kobayashi
- Department of Pathology, Kure Kyosai Hospital, Federation of National Public Service Personnel Mutual Aid Associations, Hiroshima, Japan
| | - Quoc Thang Pham
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Narutaka Katsuya
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shintaro Akabane
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Daiki Taniyama
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tetsutaro Hayashi
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naohide Oue
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Jun Teishima
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akio Matsubara
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Wataru Yasui
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
41
|
Ghosh S, Hazra J, Pal K, Nelson VK, Pal M. Prostate cancer: Therapeutic prospect with herbal medicine. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100034. [PMID: 34909665 PMCID: PMC8663990 DOI: 10.1016/j.crphar.2021.100034] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is a major cause of morbidity and mortality in men worldwide. A geographic variation on the burden of the disease suggested that the environment, genetic makeup, lifestyle, and food habits modulate one's susceptibility to the disease. Although it has been generally thought to be an older age disease, and awareness and timely execution of screening programs have managed to contain the disease in the older population over the last decades, the incidence is still increasing in the population younger than 50. Existing treatment is efficient for PCa that is localized and responsive to androgen. However, the androgen resistant and metastatic PCa are challenging to treat. Conventional radiation and chemotherapies are associated with severe side effects in addition to being exorbitantly expensive. Many isolated phytochemicals and extracts of plants used in traditional medicine are known for their safety and diverse healing properties, including many with varying levels of anti-PCa activities. Many of the phytochemicals discussed here, as shown by many laboratories, inhibit tumor cell growth and proliferation by interfering with the components in the pathways responsible for the enhanced proliferation, metabolism, angiogenesis, invasion, and metastasis in the prostate cells while upregulating the mechanisms of cell death and cell cycle arrest. Notably, many of these agents simultaneously target multiple cellular pathways. We analyzed the available literature and provided an update on this issue in this review article.
Collapse
Affiliation(s)
- Suvranil Ghosh
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, India
| | - Joyita Hazra
- Department of Biotechnology, Indian Institute of Technology Madras, Tamil Nadu, India
| | | | - Vinod K. Nelson
- Department of Pharmacology, Raghavendra Institute of Pharmaceutical Education and Research, Andhra Pradesh, India
| | - Mahadeb Pal
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, India
| |
Collapse
|
42
|
Decreased ATM Protein Expression Is Substantiated with PTEN Loss in Defining Aggressive Phenotype of Prostate Cancer Associated with Lethal Disease. EUR UROL SUPPL 2021; 29:93-101. [PMID: 34337539 PMCID: PMC8317877 DOI: 10.1016/j.euros.2021.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2021] [Indexed: 11/24/2022] Open
Abstract
Background Ataxia Telangiectasia Mutated (ATM) serine/threonine protein kinase is a known tumor suppressor, involved in DNA damage repair. It has prognostic and predictive therapeutic implications and is associated with aggressive prostate cancer (PCa). Objective To investigate the prognostic value of ATM protein expression in PCa patients and assessed the combined value of ATM, ERG, and PTEN status. Design, setting, and participants This study consisted of 303 patients with incidental, locally advanced, and castrate-resistant PCa by transurethral resection of the prostate (TURP). Outcome measurements and statistical analysis TURP samples from 303 PCa patients were assessed by immunohistochemistry (IHC for ATM, ERG, and PTEN. Individual and combined marker status were correlated with International Society of Urological Pathology Gleason grade group, overall survival (OS), and PCa-specific mortality (PCSM). Results and limitations Decreased ATM expression (negative/weak intensity) occurred in 164/303 (54.1%) patients, and was associated with shorter OS and higher PCSM (p = 0.015 and p = 0.001, respectively). Negative/weak ATM expression was significantly associated with PCSM with a hazard ratio of 2.09 (95% confidence interval 1.34–3.27, p = 0.001). Assessment of Combined ATM/PTEN expression showed improved prognostic power to predict OS and PCSM, independent of Gleason grade groups. Conclusions Decreased ATM protein expression is associated with poor outcomes in advanced PCa patients. Patients with combined low ATM/PTEN negative expression are at the highest risk for reduced OS and PCSM. Assessing the combined status of ATM/PTEN by IHC in PCa patients may aid in risk stratification relative to OS and PCSM. Moreover, since ATM plays an integral role in DNA damage response pathways, future studies will enhance our understanding of how outcomes of patients with altered ATM and PTEN expression can be improved further with poly-ADP ribose polymerase inhibitors (PARPi), combinations of PARPi and androgen receptor–targeted therapies, as well as platinum-based chemotherapies. Patient summary Lower ATM intensity is associated with increased cancer-specific mortality in prostate cancer patients. Patients with lower ATM and PTEN negative expression showed decreased overall survival and increased cancer mortality compared with controls.
Collapse
|
43
|
King B, McHugh J, Snape K. A Case-Based Clinical Approach to the Investigation, Management and Screening of Families with BRCA2 Related Prostate Cancer. APPLICATION OF CLINICAL GENETICS 2021; 14:255-266. [PMID: 34295175 PMCID: PMC8290889 DOI: 10.2147/tacg.s261737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/02/2021] [Indexed: 12/02/2022]
Abstract
BRCA2 is the most commonly implicated DNA damage repair gene associated with inherited prostate cancer. BRCA2 deficient prostate cancer typically presents at a younger age, is more poorly differentiated, and is associated with worse survival outcomes than non-BRCA2 associated prostate cancer. Despite these unfavourable prognostic implications, poly-ADP ribose polymerase inhibitors and platinum-based chemotherapy have been identified as potent targeted therapeutic agents towards BRCA1/2 deficient cancer cells. This review article explores the literature surrounding BRCA2-related prostate cancer through a familial clinical scenario. The investigation, diagnosis and management of BRCA2 deficient prostate cancer will be explored, alongside the implications of the identification of a germline pathogenic BRCA2 variant within a family, cascade screening and prostate cancer surveillance in unaffected male BRCA2 carriers. A greater understanding of the molecular pathogenesis of DNA damage repair gene deficient prostate cancer, coupled with new treatment paradigms and widened access to both somatic and germline genetic analysis for prostate cancer patients and their families will hopefully enable the robust implementation of high quality evidence-based clinical pathways for both the management and identification of BRCA2 deficient prostate cancer and improved screening, early detection and prevention strategies for individuals at increased genetic risk of prostate cancer.
Collapse
Affiliation(s)
- Bradley King
- Institute of Medical and Biomedical Education, St. George's, University of London, London, UK
| | - Jana McHugh
- Department of Oncogenomics, Institute of Cancer Research, London, UK
| | - Katie Snape
- Department of Clinical Genetics, St George's University Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
44
|
Creemers JHA, van der Doelen MJ, van Wilpe S, Hermsen R, Duiveman-de Boer T, Somford DM, Janssen MJR, Sedelaar JPM, Mehra N, Textor J, Westdorp H. Immunophenotyping Reveals Longitudinal Changes in Circulating Immune Cells During Radium-223 Therapy in Patients With Metastatic Castration-Resistant Prostate Cancer. Front Oncol 2021; 11:667658. [PMID: 34084750 PMCID: PMC8167220 DOI: 10.3389/fonc.2021.667658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/15/2021] [Indexed: 11/13/2022] Open
Abstract
Background Radium-223 improves overall survival (OS) in men with bone metastatic castration-resistant prostate cancer (mCRPC). While the exact mechanism behind this survival benefit remains unclear, radium-induced immunological mechanisms might contribute to the OS advantage. We performed a comprehensive evaluation of the immunological changes in mCRPC patients by phenotyping the peripheral blood mononuclear cells (PBMCs) during radium-223 therapy. Materials and Methods In this prospective, single-arm, exploratory study, PBMCs of 30 mCRPC patients were collected before, during, and after treatment with radium-223. Lymphocyte and monocyte counts were analyzed to get insight into general immune cell trends. Next, we analyzed changes in T cell subsets, myeloid-derived suppressor cells (MDSCs), and immune checkpoint expression using linear regression models. Per subset, the 6-month change (% of baseline) was determined. Bootstrapped 95% confidence intervals were used to measure the degree of uncertainty of our findings. Results We observed a substantial decrease in absolute lymphocyte counts (-0.12 * 10^9 cells/L per injection, 95% CI: -0.143 - -0.102). Simultaneously, an increase was observed in the proportion of T cells that expressed costimulatory (ICOS) or inhibitory (TIM-3, PD-L1, and PD-1) checkpoint molecules. Moreover, the fraction of two immunosuppressive subsets – the regulatory T cells and the monocytic MDSCs – increased throughout treatment. These findings were not more pronounced in patients with an alkaline phosphatase response during therapy. Conclusion Immune cell subsets in patients with mCRPC changed during radium-223 therapy, which warrants further research into the possible immunological consequences of these changes.
Collapse
Affiliation(s)
- Jeroen H A Creemers
- Department of Tumor Immunology, Radboudumc, Nijmegen, Netherlands.,Oncode Institute, Nijmegen, Netherlands
| | - Maarten J van der Doelen
- Department of Medical Oncology, Radboudumc, Nijmegen, Netherlands.,Department of Urology, Radboudumc, Nijmegen, Netherlands
| | - Sandra van Wilpe
- Department of Tumor Immunology, Radboudumc, Nijmegen, Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, Netherlands
| | - Rick Hermsen
- Department of Nuclear Medicine, Canisius-Wilhelmina Hospital, Nijmegen, Netherlands
| | | | - Diederik M Somford
- Department of Urology, Canisius-Wilhelmina Hospital, Nijmegen, Netherlands
| | - Marcel J R Janssen
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, Netherlands
| | | | - Niven Mehra
- Department of Tumor Immunology, Radboudumc, Nijmegen, Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, Netherlands
| | - Johannes Textor
- Department of Tumor Immunology, Radboudumc, Nijmegen, Netherlands.,Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen, Netherlands
| | - Harm Westdorp
- Department of Tumor Immunology, Radboudumc, Nijmegen, Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, Netherlands
| |
Collapse
|
45
|
Sabol RA, Ledet EM, Jaeger E, Hatton W, Moses M, Lankford A, Zaheria A, Barata P, Layton JL, Lewis BE, Sartor O. Family history and pathogenic/likely pathogenic germline variants in prostate cancer patients. Prostate 2021; 81:427-432. [PMID: 33760238 DOI: 10.1002/pros.24120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Recent literature highlights the importance of germline genetic testing in prostate cancer (PCa) patients. Surprisingly, a literature review indicates that family history (FH) records are incomplete in the major published studies from prostate cancer patients. METHODS Prospective family history data were gathered from 496 men in a single institution with a personal history of PCa who underwent germline genetic testing using a panel of at least 79 genes. Comprehensive first degree FH were obtained in all PCa of patients and analysis of prevalent FH was assessed at the time of sample collection. RESULTS Pathogenic/likely pathogenic variants (PV/LPVs) were not associated with age at diagnosis, race, or presence of metastasis. One or more first degree relatives (FDR) with any cancer was not predictive for germline PV/LPVs for men with PCa (p = .96). Separate analysis of patients with one or more FDR with breast, prostate, ovarian, or pancreatic cancer revealed that only FDR with breast or ovarian cancer was predictive for PV/LPVs (p = .028, p = .015 respectively). Patients with a FDR with breast cancer had 1.8 increased risk of PV/LPVs, and patients with a FDR with ovarian cancer had 2.9 increased risk of PV/LPV. CONCLUSION In men with a personal history of PCa, germline PV/LPVs were associated with a FDR with breast or ovarian cancer. Notably having FDRs with PCa does not predict for PV/LPVs. These data emphasize the contribution of FH in a data set with complete ascertainment of FH.
Collapse
Affiliation(s)
- Rachel A Sabol
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Elisa M Ledet
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Ellen Jaeger
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Whitley Hatton
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Marcus Moses
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Anjali Lankford
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Alexa Zaheria
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Pedro Barata
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jodi L Layton
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Brian E Lewis
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Oliver Sartor
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
46
|
Ruiz de Porras V, Pardo JC, Notario L, Etxaniz O, Font A. Immune Checkpoint Inhibitors: A Promising Treatment Option for Metastatic Castration-Resistant Prostate Cancer? Int J Mol Sci 2021; 22:ijms22094712. [PMID: 33946818 PMCID: PMC8124759 DOI: 10.3390/ijms22094712] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023] Open
Abstract
Since 2010, several treatment options have been available for men with metastatic castration-resistant prostate cancer (mCRPC), including immunotherapeutic agents, although the clinical benefit of these agents remains inconclusive in unselected mCRPC patients. In recent years, however, immunotherapy has re-emerged as a promising therapeutic option to stimulate antitumor immunity, particularly with the use of immune checkpoint inhibitors (ICIs), such as PD-1/PD-L1 and CTLA-4 inhibitors. There is increasing evidence that ICIs may be especially beneficial in specific subgroups of patients with high PD-L1 tumor expression, high tumor mutational burden, or tumors with high microsatellite instability/mismatch repair deficiency. If we are to improve the efficacy of ICIs, it is crucial to have a better understanding of the mechanisms of resistance to ICIs and to identify predictive biomarkers to determine which patients are most likely to benefit. This review focuses on the current status of ICIs for the treatment of mCRPC (either as monotherapy or in combination with other drugs), mechanisms of resistance, potential predictive biomarkers, and future challenges in the management of mCRPC.
Collapse
Affiliation(s)
- Vicenç Ruiz de Porras
- Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
- Badalona Applied Research Group in Oncology (B·ARGO), Catalan Institute of Oncology, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.C.P.); (L.N.); (O.E.)
- Correspondence: (V.R.d.P.); (A.F.); Tel.: +34-93-554-6301 (V.R.d.P.); +34-93-497-8925 (A.F.); Fax: +34-93-497-8950 (A.F.)
| | - Juan Carlos Pardo
- Badalona Applied Research Group in Oncology (B·ARGO), Catalan Institute of Oncology, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.C.P.); (L.N.); (O.E.)
- Department of Medical Oncology, Catalan Institute of Oncology, University Hospital Germans Trias i Pujol, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
| | - Lucia Notario
- Badalona Applied Research Group in Oncology (B·ARGO), Catalan Institute of Oncology, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.C.P.); (L.N.); (O.E.)
- Department of Medical Oncology, Catalan Institute of Oncology, University Hospital Germans Trias i Pujol, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
| | - Olatz Etxaniz
- Badalona Applied Research Group in Oncology (B·ARGO), Catalan Institute of Oncology, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.C.P.); (L.N.); (O.E.)
- Department of Medical Oncology, Catalan Institute of Oncology, University Hospital Germans Trias i Pujol, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
| | - Albert Font
- Badalona Applied Research Group in Oncology (B·ARGO), Catalan Institute of Oncology, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.C.P.); (L.N.); (O.E.)
- Department of Medical Oncology, Catalan Institute of Oncology, University Hospital Germans Trias i Pujol, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
- Correspondence: (V.R.d.P.); (A.F.); Tel.: +34-93-554-6301 (V.R.d.P.); +34-93-497-8925 (A.F.); Fax: +34-93-497-8950 (A.F.)
| |
Collapse
|
47
|
The Screen Project: Guided Direct-To-Consumer Genetic Testing for Breast Cancer Susceptibility in Canada. Cancers (Basel) 2021; 13:cancers13081894. [PMID: 33920818 PMCID: PMC8071125 DOI: 10.3390/cancers13081894] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 11/17/2022] Open
Abstract
There is limited information of the outcomes of direct-to-consumer testing for BRCA1 and BRCA2 mutations. The Screen Project was initiated in 2017 to offer BRCA1 and BRCA2 genetic screening to all Canadians over the age of 18 who wish to know their mutation status. Patients enrolled in the study from 2017 to 2019 and were followed for one year after the receipt of a genetic test result. Study subjects registered online and were sent a saliva sample kit, which was shipped to the reference laboratory. Pre-test genetic counselling and counselling for mutation-negative subjects was optional and at the individual's discretion. There were 1269 tested individuals between March 2017 and January 2019. A total of 1157 (93%) were women and 87 (7%) were men. Sixty-six percent had a first- or second-degree relative with breast or ovarian cancer. Of the 1269 tested individuals, 30 (2.4%) had a pathogenic mutation in BRCA1 or BRCA2 (20 women and 10 men). Seventy-five percent of the female mutation carriers underwent a bilateral mastectomy and/or salpingo-oophorectomy within a year of receiving a positive result. Genetic counselling was available at no cost to all participants but was requested by only 5% of the non-carriers. The study subjects expressed a high degree of satisfaction with the process.
Collapse
|
48
|
Vietri MT, D’Elia G, Caliendo G, Resse M, Casamassimi A, Passariello L, Albanese L, Cioffi M, Molinari AM. Hereditary Prostate Cancer: Genes Related, Target Therapy and Prevention. Int J Mol Sci 2021; 22:ijms22073753. [PMID: 33916521 PMCID: PMC8038462 DOI: 10.3390/ijms22073753] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/27/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) is globally the second most diagnosed cancer type and the most common cause of cancer-related deaths in men. Family history of PCa, hereditary breast and ovarian cancer (HBOC) and Lynch syndromes (LS), are among the most important risk factors compared to age, race, ethnicity and environmental factors for PCa development. Hereditary prostate cancer (HPCa) has the highest heritability of any major cancer in men. The proportion of PCa attributable to hereditary factors has been estimated in the range of 5–15%. To date, the genes more consistently associated to HPCa susceptibility include mismatch repair (MMR) genes (MLH1, MSH2, MSH6, and PMS2) and homologous recombination genes (BRCA1/2, ATM, PALB2, CHEK2). Additional genes are also recommended to be integrated into specific research, including HOXB13, BRP1 and NSB1. Importantly, BRCA1/BRCA2 and ATM mutated patients potentially benefit from Poly (ADP-ribose) polymerase PARP inhibitors, through a mechanism of synthetic lethality, causing selective tumor cell cytotoxicity in cell lines. Moreover, the detection of germline alterations in MMR genes has therapeutic implications, as it may help to predict immunotherapy benefits. Here, we discuss the current knowledge of the genetic basis for inherited predisposition to PCa, the potential target therapy, and the role of active surveillance as a management strategy for patients with low-risk PCa. Finally, the current PCa guideline recommendations are reviewed.
Collapse
Affiliation(s)
- Maria Teresa Vietri
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy; (A.C.); (A.M.M.)
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
- Correspondence: ; Tel.: +39-081-566-7639; Fax: +39-081-450-169
| | - Giovanna D’Elia
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| | - Gemma Caliendo
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| | - Marianna Resse
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| | - Amelia Casamassimi
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy; (A.C.); (A.M.M.)
| | - Luana Passariello
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| | - Luisa Albanese
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| | - Michele Cioffi
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| | - Anna Maria Molinari
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy; (A.C.); (A.M.M.)
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| |
Collapse
|
49
|
Lozano R, Salles DC, Sandhu S, Aragón IM, Thorne H, López-Campos F, Rubio-Briones J, Gutierrez-Pecharroman AM, Maldonado L, di Domenico T, Sanz A, Prieto JD, García I, Pacheco MI, Garcés T, Llacer C, Romero-Laorden N, Zambrana F, López-Casas PP, Lorente D, Mateo J, Pritchard CC, Antonarakis ES, Olmos D, Lotan TL, Castro E. Association between BRCA2 alterations and intraductal and cribriform histologies in prostate cancer. Eur J Cancer 2021; 147:74-83. [PMID: 33626496 DOI: 10.1016/j.ejca.2021.01.027] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/29/2020] [Accepted: 01/16/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Intraductal (IDC) and cribriform (CRIB) histologies in prostate cancer have been associated with germline BRCA2 (gBRCA2) mutations in small retrospective series, leading to the recommendation of genetic testing for patients with IDC in the primary tumour. PATIENTS AND METHODS To examine the association of gBRCA2 mutations and other tumour molecular features with IDC and/or cribriform (CRIB) histologies, we conducted a case-control study in which primary prostate tumours from 58 gBRCA2 carriers were matched (1:2) by Gleason Grade Group and specimen type to 116 non-carriers. Presence/absence of IDC and CRIB morphologies was established by two expert uropathologists blinded to gBRCA2 status. Fluorescent in-situ hybridization (FISH) and next-generation sequencing (NGS) were used to detect BRCA2 alterations, PTEN deletions and TMPRSS2-ERG fusions. Chi-squared tests were used to compare the frequency of IDC and CRIB in gBRCA2 carriers and controls and to assess associations with other variables. Logistic regression models were constructed to identify independent factors associated with both histology patterns. RESULTS No significant differences between gBRCA2 carriers and non-carriers were observed in the prevalence of IDC (36% gBRCA2 versus 50% non-carriers, p = 0.085) or CRIB (53% gBRCA2 versus 43% non-carriers p = 0.197) patterns. However, IDC histology was independently associated with bi-allelic BRCA2 alterations (OR 4.3, 95%CI 1.1-16.2) and PTEN homozygous loss (OR 5.2, 95%CI 2.1-13.1). CRIB morphology was also independently associated with bi-allelic BRCA2 alterations (OR 5.6, 95%CI 1.7-19.3). CONCLUSIONS While we found no association between gBRCA2 mutations and IDC or CRIB histologies, bi-allelic BRCA2 loss in primary prostate tumours was significantly associated with both variant morphologies, independently of other clinical-pathologic factors.
Collapse
Affiliation(s)
- Rebeca Lozano
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Genitourinary Cancer Translational Research Group, Instituto de Investigación Biomédica de Málaga, Spain
| | - Daniela C Salles
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Shahneen Sandhu
- Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Isabel M Aragón
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Genitourinary Cancer Translational Research Group, Instituto de Investigación Biomédica de Málaga, Spain
| | - Heather Thorne
- Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Fernando López-Campos
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Radiation Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - José Rubio-Briones
- Urology Department, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Ana M Gutierrez-Pecharroman
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Department of Pathology, Hospital de Getafe, Getafe, Spain
| | - Laneisha Maldonado
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Tomas di Domenico
- Bioinformatics Unit, Spanish National Cancer Research Center, Madrid, Spain
| | - Alejandro Sanz
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Juan D Prieto
- Department of Pathology, Hospital Universitarios Virgen de la Victoria Málaga, Spain
| | - Isabel García
- Department of Pathology, Hospital Universitarios Virgen de la Victoria Málaga, Spain
| | - María I Pacheco
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Teresa Garcés
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Genitourinary Cancer Translational Research Group, Instituto de Investigación Biomédica de Málaga, Spain
| | - Casilda Llacer
- Genitourinary Cancer Translational Research Group, Instituto de Investigación Biomédica de Málaga, Spain; Medical Oncology, UGCI, Hospitales Universitarios Virgen de la Victoria y Regional de Málaga, Málaga, Spain
| | | | | | - Pedro P López-Casas
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - David Lorente
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Medical Oncology, Hospital Provincial de Castellón, Castellón de la Plana, Spain
| | - Joaquin Mateo
- Prostate Cancer Traslational Research Unit, Vall'Hebron Institute of Oncology, Spain
| | | | - Emmanuel S Antonarakis
- Medical Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - David Olmos
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Genitourinary Cancer Translational Research Group, Instituto de Investigación Biomédica de Málaga, Spain
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA.
| | - Elena Castro
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Genitourinary Cancer Translational Research Group, Instituto de Investigación Biomédica de Málaga, Spain; Medical Oncology, UGCI, Hospitales Universitarios Virgen de la Victoria y Regional de Málaga, Málaga, Spain.
| |
Collapse
|
50
|
Clinical and genomic insights into circulating tumor DNA-based alterations across the spectrum of metastatic hormone-sensitive and castrate-resistant prostate cancer. EBioMedicine 2021; 54:102728. [PMID: 32268276 PMCID: PMC7186589 DOI: 10.1016/j.ebiom.2020.102728] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/26/2020] [Accepted: 03/04/2020] [Indexed: 01/10/2023] Open
Abstract
Background Metastatic prostate cancer is a clonally heterogeneous disease state characterized by progressive somatic perturbations. The aim of this study was to identify cell free DNA- (cfDNA-) based alterations and their associations with outcomes in progressive metastatic prostate cancer. Methods In this longitudinal prospective cohort study plasma cfDNA/circulating tumor DNA (ctDNA) was analyzed before, during, and after androgen deprivation therapy (ADT) in 4 independent patient groups ranging from untreated metastatic hormone sensitive prostate cancer (mHSPC) to metastatic castrate resistant prostate cancer (mCRPC). Next generation sequencing was performed on ctDNA and germline DNA to characterize alterations and associations with clinical outcomes were determined for each group. Findings cfDNA yields were different in progressive mHSPC and mCRPC states (P < .001). In mHSPC, a higher than median ctDNA fraction was predictive of shorter time to ADT failure (HR, 2.29 [95% CI, 1.13–4.65]; Log-Rank P = .02). cfDNA, ctDNA taken with volume of metastatic disease in mHSPC and with alkaline phosphatase levels prognosticated survival better than clinical factors alone in mHSPC and mCRPC states (Log Rank P = 0.03). ctDNA-based AR, APC mutations were increased in mCRPC compared to mHSPC (P < ·05).TP53 mutations, RB1 loss, and AR gene amplifications correlated with poorer survival in mCRPC. Mutations in multiple DNA repair genes (ATM, BRCA1, BRCA2, CHEK2) were associated with time to ADT treatment failure and survival in mHSPC. Interpretation ctDNA fraction can further refine clinical prognostic factors in metastatic prostate cancer. Somatic ctDNA alterations have potential prognostic, predictive, and therapeutic implications in metastatic prostate cancer management. Funding Several funding sources have supported this study. A full list is provided in the Acknowledgments. No funding was received from Predicine, Inc. during the conduct of the study.
Collapse
|