401
|
PROMISE: a real-world clinical-genomic database to address knowledge gaps in prostate cancer. Prostate Cancer Prostatic Dis 2021; 25:388-396. [PMID: 34363009 PMCID: PMC9385488 DOI: 10.1038/s41391-021-00433-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/24/2021] [Accepted: 07/21/2021] [Indexed: 01/23/2023]
Abstract
PURPOSE Prostate cancer is a heterogeneous disease with variable clinical outcomes. Despite numerous recent approvals of novel therapies, castration-resistant prostate cancer remains lethal. A "real-world" clinical-genomic database is urgently needed to enhance our characterization of advanced prostate cancer and further enable precision oncology. METHODS The Prostate Cancer Precision Medicine Multi-Institutional Collaborative Effort (PROMISE) is a consortium whose aims are to establish a repository of de-identified clinical and genomic patient data that are linked to patient outcomes. The consortium structure includes a (1) bio-informatics committee to standardize genomic data and provide quality control, (2) biostatistics committee to independently perform statistical analyses, (3) executive committee to review and select proposals of relevant questions for the consortium to address, (4) diversity/inclusion committee to address important clinical questions pertaining to racial disparities, and (5) patient advocacy committee to understand patient perspectives to improve patients' quality of care. RESULTS The PROMISE consortium was formed by 16 academic institutions in early 2020 and a secure RedCap database was created. The first patient record was entered into the database in April 2020 and over 1000 records have been entered as of early 2021. Data entry is proceeding as planned with the goal to have over 2500 patient records by the end of 2021. CONCLUSIONS The PROMISE consortium provides a powerful clinical-genomic platform to interrogate and address data gaps that have arisen with increased genomic testing in the clinical management of prostate cancer. The dataset incorporates data from patient populations that are often underrepresented in clinical trials, generates new hypotheses to direct further research, and addresses important clinical questions that are otherwise difficult to investigate in prospective studies.
Collapse
|
402
|
Maughan BL, Antonarakis ES. Olaparib and rucaparib for the treatment of DNA repair-deficient metastatic castration-resistant prostate cancer. Expert Opin Pharmacother 2021; 22:1625-1632. [PMID: 33827356 PMCID: PMC8419006 DOI: 10.1080/14656566.2021.1912015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/29/2021] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Metastatic prostate cancer is a heterogeneous disease characterized by clinical and genomic heterogeneity. Many prostate cancers harbor mutations causing DNA repair deficiency, specifically homologous recombination deficiency, sensitizing to drugs that inhibit poly ADP-ribose polymerase (PARP). PARP is an enzyme that is involved in single-stranded DNA repair and is the target of newly approved treatments for metastatic prostate cancer. AREAS COVERED Here, the authors' review the clinical trials leading to the recent approvals of two PARP inhibitors (PARPi), olaparib and rucaparib, specifically TOPARP-A, TOPARP-B, PROfound and TRITON-2. They also compare the different FDA approvals for both of these medications and outline the safety of this class of drugs in prostate cancer. EXPERT OPINION Because PARPi are particularly effective in men with somatic or germline alterations in BRCA1 and BRCA2, we recommend that all men be tested for DNA alterations with next-generation sequencing in tumor cells obtained from either tissue or blood. We also recommend that olaparib or rucaparib be considered relatively early in the treatment sequence in metastatic castration-resistant prostate cancer patients with BRCA1 or BRCA2 mutations. Other DNA alterations might also sensitize to PARPi though the response rates are lower, so other standard therapies should be prioritized first.
Collapse
Affiliation(s)
| | - Emmanuel S. Antonarakis
- Department of Oncology and Urology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
403
|
Tomlins SA, Hovelson DH, Suga JM, Anderson DM, Koh HA, Dees EC, McNulty B, Burkard ME, Guarino M, Khatri J, Safa MM, Matrana MR, Yang ES, Menter AR, Parsons BM, Slim JN, Thompson MA, Hwang L, Edenfield WJ, Nair S, Onitilo A, Siegel R, Miller A, Wassenaar T, Irvin WJ, Schulz W, Padmanabhan A, Harish V, Gonzalez A, Mansoor AH, Kellum A, Harms P, Drewery S, Falkner J, Fischer A, Hipp J, Kwiatkowski K, Lazo de la Vega L, Mitchell K, Reeder T, Siddiqui J, Vakil H, Johnson DB, Rhodes DR. Real-World Performance of a Comprehensive Genomic Profiling Test Optimized for Small Tumor Samples. JCO Precis Oncol 2021; 5:PO.20.00472. [PMID: 34476329 PMCID: PMC8384401 DOI: 10.1200/po.20.00472] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/18/2021] [Accepted: 07/09/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Tissue-based comprehensive genomic profiling (CGP) is increasingly used for treatment selection in patients with advanced cancer; however, tissue availability may limit widespread implementation. Here, we established real-world CGP tissue availability and assessed CGP performance on consecutively received samples. MATERIALS AND METHODS We conducted a post hoc, nonprespecified analysis of 32,048 consecutive tumor tissue samples received for StrataNGS, a multiplex polymerase chain reaction (PCR)-based comprehensive genomic profiling (PCR-CGP) test, as part of an ongoing observational trial (NCT03061305). Sample characteristics and PCR-CGP performance were assessed across all tested samples, including exception samples not meeting minimum input quality control (QC) requirements (< 20% tumor content [TC], < 2 mm2 tumor surface area [TSA], DNA or RNA yield < 1 ng/µL, or specimen age > 5 years). Tests reporting ≥ 1 prioritized alteration or meeting TC and sequencing QC were considered successful. For prostate carcinoma and lung adenocarcinoma, tests reporting ≥ 1 actionable or informative alteration or meeting TC and sequencing QC were considered actionable. RESULTS Among 31,165 (97.2%) samples where PCR-CGP was attempted, 10.7% had < 20% TC and 59.2% were small (< 25 mm2 tumor surface area). Of 31,101 samples evaluable for input requirements, 8,089 (26.0%) were exceptions not meeting requirements. However, 94.2% of the 31,101 tested samples were successfully reported, including 80.5% of exception samples. Positive predictive value of PCR-CGP for ERBB2 amplification in exceptions and/or sequencing QC-failure breast cancer samples was 96.7%. Importantly, 84.0% of tested prostate carcinomas and 87.9% of lung adenocarcinomas yielded results informing treatment selection. CONCLUSION Most real-world tissue samples from patients with advanced cancer desiring CGP are limited, requiring optimized CGP approaches to produce meaningful results. An optimized PCR-CGP test, coupled with an inclusive exception testing policy, delivered reportable results for > 94% of samples, potentially expanding the proportion of CGP-testable patients and impact of biomarker-guided therapies.
Collapse
Affiliation(s)
| | | | | | - Daniel M. Anderson
- Metro-Minnesota Community Oncology Research Consortium (MMCORC), St Louis Park, MN
| | | | - Elizabeth C. Dees
- The University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | | | | | - Michael Guarino
- ChristianaCare's Helen F. Graham Cancer Center & Research Institute, Newark, DE
| | - Jamil Khatri
- ChristianaCare's Helen F. Graham Cancer Center & Research Institute, Newark, DE
| | | | | | - Eddy S. Yang
- University of Alabama at Birmingham, Birmingham, AL
| | | | | | | | | | - Leon Hwang
- Kaiser Permanente Mid Atlantic, Rockville, MD
| | | | | | | | - Robert Siegel
- Bon Secours St Francis Cancer Center, Greenville, SC
| | | | | | - William J. Irvin
- Bon Secours St Francis Medical Center Midlothian, Midlothian, VA
| | | | | | | | | | | | | | - Paul Harms
- University of Michigan Health Systems, Ann Arbor, MI
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
404
|
Loeb S, Li R, Sanchez Nolasco T, Byrne N, Cheng HH, Becker D, Leader AE, Giri VN. Barriers and facilitators of germline genetic evaluation for prostate cancer. Prostate 2021; 81:754-764. [PMID: 34057231 DOI: 10.1002/pros.24172] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Genetic counseling and germline testing have an increasingly important role for patients with prostate cancer (PCa); however, recent data suggests they are underutilized. Our objective was to perform a qualitative study of the barriers and facilitators of germline genetic evaluation among physicians who manage PCa. METHODS We conducted semi-structured interviews with medical oncologists, radiation oncologists, and urologists from different U.S. practice settings until thematic saturation was achieved at n = 14. The interview guide was based on the Tailored Implementation in Chronic Diseases Framework to identify key determinants of practice. Interview transcripts were independently coded by ≥2 investigators using a constant comparative method. RESULTS The decision to perform or refer for germline genetic evaluation is affected by factors at multiple levels. Although patient factors sometimes play a role, the dominant themes in the decision to conduct germline genetic evaluation were at the physician and organizational level. Physician knowledge, coordination of care, perceptions of the guidelines, and concerns about cost were most frequently discussed as the main factors affecting utilization of germline genetic evaluation. CONCLUSIONS There are currently numerous barriers to implementation of germline genetic evaluation for PCa. Efforts to expand physician education, to develop tools to enhance genetics in practice, and to facilitate coordination of care surrounding genetic evaluation are important to promote guideline-concordant care.
Collapse
Affiliation(s)
- Stacy Loeb
- Department of Urology, New York University, New York, New York, USA
- Department of Population Health, New York University, New York, New York, USA
- Department of Surgery/Urology, Manhattan Veterans Affairs, New York, New York, USA
| | - Randall Li
- Department of Urology, New York University, New York, New York, USA
| | - Tatiana Sanchez Nolasco
- Department of Urology, New York University, New York, New York, USA
- Department of Population Health, New York University, New York, New York, USA
| | - Nataliya Byrne
- Department of Urology, New York University, New York, New York, USA
- Department of Population Health, New York University, New York, New York, USA
| | - Heather H Cheng
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, Washington State, USA
| | - Daniel Becker
- Department of Surgery/Urology, Manhattan Veterans Affairs, New York, New York, USA
- Department of Medicine, New York University, New York, NY, USA
| | - Amy E Leader
- Division of Population Science, Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Veda N Giri
- Division of Population Science, Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
405
|
Shore ND, Laliberté F, Ionescu-Ittu R, Yang L, Mahendran M, Lejeune D, Yu LH, Burgents J, Duh MS, Ghate SR. Real-World Treatment Patterns and Overall Survival of Patients with Metastatic Castration-Resistant Prostate Cancer in the US Prior to PARP Inhibitors. Adv Ther 2021; 38:4520-4540. [PMID: 34282527 PMCID: PMC8342357 DOI: 10.1007/s12325-021-01823-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/08/2021] [Indexed: 12/19/2022]
Abstract
Introduction Therapeutic options for metastatic castration-resistant prostate cancer (mCRPC) patients are continuously advancing. We described mCRPC treatment patterns in the US from 2013 to 2019. Methods Patients with a confirmed mCRPC diagnosis and adenocarcinoma histology were included in the US Flatiron Health Electronic Health Record-derived de-identified database. Treatment patterns [including treatment per lines of therapies (LOTs), LOT sequences, and time on treatment] and overall survival (OS) have been described in mCRPC settings. Results Of 5213 patients (mean age: 72.6 years), 4374 (83.9%) were treated with ≥ 1 LOT post-mCRPC diagnosis (among those with ≥ 1 LOT, 55.3%, 29.5%, 14.7%, and 6.7% had ≥ 2, 3, 4, and 5 LOTs, respectively). In first line (1L), the main treatment class was next-generation hormonal agents (NHA; 62.5% of patients with ≥ 1 LOT), while the shortest and longest time on 1L were observed for chemotherapy (median 2.8 months) and NHA (median 5.1 months), respectively. The most common LOT sequences were NHA → NHA (29.4% of patients with ≥ 2 LOTs) and NHA → NHA → chemotherapy (16.7% of patients with ≥ 3 LOTs). In Kaplan–Meier analyses, the median OS was 19.4, 14.6, and 11.1 months post-1L, 2L, and 3L start, respectively. Patients who moved rapidly through LOTs had an increased risk of death. Conclusions NHA were widely used as 1L therapy in mCRPC patients from 2013 to 2019, but time on 1L NHA treatment was on average < 6 months. While NHA → NHA was the most observed 1L → 2L LOT sequence, a plethora of other LOT sequences were observed. OS was poor, highlighting an unmet need for life-prolonging treatments. Supplementary Information The online version contains supplementary material available at 10.1007/s12325-021-01823-6.
Collapse
Affiliation(s)
- Neal D Shore
- Carolina Urologic Research Center, 823 82nd Pkwy Suite b, Myrtle Beach, SC, 29572, USA
| | - François Laliberté
- Analysis Group, Inc., 1190 Avenue des Canadiens-de-Montréal, Suite 1500, Montréal, QC, H3B 0G7, Canada
| | - Raluca Ionescu-Ittu
- Analysis Group, Inc., 1190 Avenue des Canadiens-de-Montréal, Suite 1500, Montréal, QC, H3B 0G7, Canada.
| | - Lingfeng Yang
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Malena Mahendran
- Analysis Group, Inc., 1190 Avenue des Canadiens-de-Montréal, Suite 1500, Montréal, QC, H3B 0G7, Canada
| | - Dominique Lejeune
- Analysis Group, Inc., 1190 Avenue des Canadiens-de-Montréal, Suite 1500, Montréal, QC, H3B 0G7, Canada
| | - Louise H Yu
- Analysis Group, Inc, 111 Huntington Ave 14th Floor, Boston, MA, 02199, USA
| | - Joseph Burgents
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Mei Sheng Duh
- Analysis Group, Inc, 111 Huntington Ave 14th Floor, Boston, MA, 02199, USA
| | - Sameer R Ghate
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| |
Collapse
|
406
|
Carvalho RL, de Miranda AS, Nunes MP, Gomes RS, Jardim GAM, Júnior ENDS. On the application of 3d metals for C-H activation toward bioactive compounds: The key step for the synthesis of silver bullets. Beilstein J Org Chem 2021; 17:1849-1938. [PMID: 34386103 PMCID: PMC8329403 DOI: 10.3762/bjoc.17.126] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/28/2021] [Indexed: 01/24/2023] Open
Abstract
Several valuable biologically active molecules can be obtained through C-H activation processes. However, the use of expensive and not readily accessible catalysts complicates the process of pharmacological application of these compounds. A plausible way to overcome this issue is developing and using cheaper, more accessible, and equally effective catalysts. First-row transition (3d) metals have shown to be important catalysts in this matter. This review summarizes the use of 3d metal catalysts in C-H activation processes to obtain potentially (or proved) biologically active compounds.
Collapse
Affiliation(s)
- Renato L Carvalho
- Institute of Exact Sciences, Department of Chemistry, Federal University of Minas Gerais - UFMG, CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Amanda S de Miranda
- Institute of Exact Sciences, Department of Chemistry, Federal University of Minas Gerais - UFMG, CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Mateus P Nunes
- Institute of Exact Sciences, Department of Chemistry, Federal University of Minas Gerais - UFMG, CEP 31270-901, Belo Horizonte, MG, Brazil
| | - Roberto S Gomes
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, United States
| | - Guilherme A M Jardim
- Institute of Exact Sciences, Department of Chemistry, Federal University of Minas Gerais - UFMG, CEP 31270-901, Belo Horizonte, MG, Brazil
- Centre for Excellence for Research in Sustainable Chemistry (CERSusChem), Department of Chemistry, Federal University of São Carlos – UFSCar, CEP 13565-905, São Carlos, SP, Brazil
| | - Eufrânio N da Silva Júnior
- Institute of Exact Sciences, Department of Chemistry, Federal University of Minas Gerais - UFMG, CEP 31270-901, Belo Horizonte, MG, Brazil
| |
Collapse
|
407
|
Liu YL, Stadler ZK. The Future of Parallel Tumor and Germline Genetic Testing: Is There a Role for All Patients With Cancer? J Natl Compr Canc Netw 2021; 19:871-878. [PMID: 34340209 PMCID: PMC11123333 DOI: 10.6004/jnccn.2021.7044] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 04/09/2021] [Indexed: 11/17/2022]
Abstract
Under the traditional paradigm of genetic testing in cancer, the role of germline testing was to assess for the inherited risk of cancer, whereas the role of tumor testing was to determine therapeutic selection. Parallel tumor-normal genetic testing uses simultaneous genetic testing of the tumor and normal tissue to identify mutations and allows their classification as either germline or somatic. The increasing adoption of parallel testing has revealed a greater number of germline findings in patients who otherwise would not have met clinical criteria for testing. This result has widespread implications for the screening and further testing of at-risk relatives and for gene discovery. It has also revealed the importance of germline testing in therapeutic actionability. Herein, we describe the pros and cons of tumor-only versus parallel tumor-normal testing and summarize the data on the prevalence of incidental actionable germline findings. Because germline testing in patients with cancer continues to expand, it is imperative that systems be in place for the proper interpretation, dissemination, and counseling for patients and at-risk relatives. We also review new therapeutic approvals with germline indications and highlight the increasing importance of germline testing in selecting therapies. Because recommendations for universal genetic testing are increasing in multiple cancer types and the number of approved therapies with germline indications is also increasing, a gradual transition toward parallel tumor-normal genetic testing in all patients with cancer is foreseeable.
Collapse
Affiliation(s)
- Ying L. Liu
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zsofia K. Stadler
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
408
|
Nientiedt C, Budczies J, Endris V, Kirchner M, Schwab C, Jurcic C, Behnisch R, Hoveida S, Lantwin P, Kaczorowski A, Geisler C, Dieffenbacher S, Falkenbach F, Franke D, Görtz M, Heller M, Himmelsbach R, Pecqueux C, Rath M, Reimold P, Schütz V, Simunovic I, Walter E, Hofer L, Gasch C, Schönberg G, Pursche L, Hatiboglu G, Nyarangi-Dix J, Sültmann H, Zschäbitz S, Koerber SA, Jäger D, Debus J, Duensing A, Schirmacher P, Hohenfellner M, Stenzinger A, Duensing S. Mutations in TP53 or DNA damage repair genes define poor prognostic subgroups in primary prostate cancer. Urol Oncol 2021; 40:8.e11-8.e18. [PMID: 34325986 DOI: 10.1016/j.urolonc.2021.06.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/11/2021] [Accepted: 06/27/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Mutations in DNA damage repair genes, in particular genes involved in homology-directed repair, define a subgroup of men with prostate cancer with a more unfavorable prognosis but a therapeutic vulnerability to PARP inhibition. In current practice, mutational testing of prostate cancer patients is commonly done late i.e., when the tumor is castration resistant. In addition, most sequencing panels do not include TP53, one of the most crucial tumor suppressor genes in human cancer. In this proof-of-concept study, we sought to extend the clinical use of these molecular markers by exploring the early prognostic impact of mutations in TP53 and DNA damage repair genes in men with primary, nonmetastatic prostate cancer undergoing radical prostatectomy (RPX). METHODS Tumor specimens from a cohort of 68 RPX patients with intermediate (n = 11, 16.2%) or high-risk (n = 57, 83.8%) disease were analyzed by targeted next generation sequencing using a 37 DNA damage repair and checkpoint gene panel including TP53. Sequencing results were correlated to clinicopathologic variables as well as PSA persistence or time to PSA failure. In addition, the distribution of TP53 and DNA damage repair gene mutations was analyzed in three large publicly available datasets (TCGA, MSKCC and SU2C). RESULTS Of 68 primary prostate cancers analyzed, 23 (33.8%) were found to harbor a mutation in either TP53 (n = 12, 17.6%) or a DNA damage repair gene (n = 11, 16.2%). The vast majority of these mutations (22 of 23, 95.7%) were detected in primary tumors from patients with high-risk features. These mutations were mutually exclusive in our cohort and additional data mining suggests an enrichment of DNA damage repair gene mutations in TP53 wild-type tumors. Mutations in either TP53 or a DNA damage repair gene were associated with a significantly worse prognosis after RPX. Importantly, the presence of TP53/DNA damage repair gene mutations was an independent risk factor for PSA failure or PSA persistence in multivariate Cox regression models. CONCLUSION TP53 or DNA damage repair gene mutations are frequently detected in primary prostate cancer with high-risk features and define a subgroup of patients with an increased risk for PSA failure or persistence after RPX. The significant adverse impact of these alterations on patient prognosis may be exploited to identify men with prostate cancer who may benefit from a more intensified treatment.
Collapse
Affiliation(s)
- Cathleen Nientiedt
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Im Neuenheimer Feld 460, Heidelberg, Germany
| | - Jan Budczies
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, Heidelberg, Germany
| | - Volker Endris
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, Heidelberg, Germany
| | - Martina Kirchner
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, Heidelberg, Germany
| | - Constantin Schwab
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, Heidelberg, Germany
| | - Christina Jurcic
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, Heidelberg, Germany
| | - Rouven Behnisch
- Institute of Medical Biometry and Informatics, University of Heidelberg, Im Neuenheimer Feld 130, Heidelberg, Germany
| | - Shirin Hoveida
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, Heidelberg, Germany
| | - Philippa Lantwin
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, Heidelberg, Germany
| | - Adam Kaczorowski
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, Heidelberg, Germany
| | - Christine Geisler
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Svenja Dieffenbacher
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Fabian Falkenbach
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Desiree Franke
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Magdalena Görtz
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Martina Heller
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Ruth Himmelsbach
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Carine Pecqueux
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Mathias Rath
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Philipp Reimold
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Viktoria Schütz
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Iva Simunovic
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Elena Walter
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Luisa Hofer
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Claudia Gasch
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Gita Schönberg
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Lars Pursche
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Gencay Hatiboglu
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Joanne Nyarangi-Dix
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Holger Sültmann
- Cancer Genome Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 460, Heidelberg, Germany
| | - Stefanie Zschäbitz
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Im Neuenheimer Feld 460, Heidelberg, Germany
| | - Stefan A Koerber
- Department of Radiation Oncology, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Im Neuenheimer Feld 460, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
| | - Anette Duensing
- Cancer Therapeutics Program and Department of Pathology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, USA; Precision Oncology of Urological Malignancies, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, Heidelberg, Germany
| | - Markus Hohenfellner
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, Heidelberg, Germany.
| | - Stefan Duensing
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, Heidelberg, Germany.
| |
Collapse
|
409
|
Lutetium-177 Labelled PSMA Targeted Therapy in Advanced Prostate Cancer: Current Status and Future Perspectives. Cancers (Basel) 2021; 13:cancers13153715. [PMID: 34359614 PMCID: PMC8371469 DOI: 10.3390/cancers13153715] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/09/2021] [Accepted: 07/21/2021] [Indexed: 11/17/2022] Open
Abstract
Patients suffering from metastatic castration-resistant prostate cancer (mCRPC) have a poor prognosis. As a further treatment option 177Lutetium (Lu) prostate-specific membrane antigen (PSMA) radioligand therapy gained a significant interest of many investigators. Several publications showed great response and prolonged survival with limited adverse events. However, to this point, it still remains unclear which patients benefit the most from 177Lu-PSMA therapy, and how to improve the treatment regimen to achieve best outcome while minimizing potential adverse events. The efficacy for mCRPC patients is a given fact, and with the newly published results of the VISION trial its approval is only a matter of time. Recently, investigators started to focus on treating prostate cancer patients in earlier disease stages and in combination with other compounds. This review gives a brief overview of the current state and the future perspectives of 177Lu labelled PSMA radioligand therapy.
Collapse
|
410
|
Vanacker H, Harter P, Labidi-Galy SI, Banerjee S, Oaknin A, Lorusso D, Ray-Coquard I. PARP-inhibitors in epithelial ovarian cancer: Actual positioning and future expectations. Cancer Treat Rev 2021; 99:102255. [PMID: 34332292 DOI: 10.1016/j.ctrv.2021.102255] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022]
Abstract
Poly-(ADP)-ribose polymerase inhibitors (PARPi) are a class of oral anticancer drugs first developed as "synthetically lethal" in cancers harboring BRCA1/BRCA2 inactivating mutations. In high-grade serous or endometrioid ovarian cancers (HGOC), PARPi demonstrated benefit as maintenance therapy in relapsing BRCA-mutated and non-mutated tumors. Recently, they extended their indications to frontline maintenance therapy. This review summarizes the current place of PARPi (i) as maintenance or single agent in recurrent disease and (ii) frontline maintenance with different settings. We reviewed the course of biomarker identification, the challenge of overcoming resistance to PARPi and future combinations with targeted therapies, including anti-angiogenic, immune checkpoint inhibitors and DNA damage response inhibitors.
Collapse
Affiliation(s)
- Hélène Vanacker
- Centre Léon Bérard, Lyon, France; University Claude Bernard Lyon 1, France.
| | - Philipp Harter
- Department of Gynecology & Gynecologic Oncology, Ev. Kliniken Essen-Mitte, Essen, Germany.
| | - Sana Intidhar Labidi-Galy
- Department of Oncology, Hôpitaux Universitaires de Genève, Switzerland; Faculty of Medicine, Swiss Cancer Center Leman, Geneva, Switzerland.
| | - Susana Banerjee
- Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, United Kingdom.
| | - Ana Oaknin
- Vall d'Hebron Institute of Oncology, Barcelona, Spain.
| | | | | |
Collapse
|
411
|
Dorff TB, O'Neil B, Hoffman KE, Lin DW, Loughlin KR, Dall'Era M. 25-year perspective on prostate cancer: Conquering frontiers and understanding tumor biology. Urol Oncol 2021; 39:521-527. [PMID: 34266741 DOI: 10.1016/j.urolonc.2021.04.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 10/20/2022]
Abstract
Major changes in the field of prostate cancer over the last 25 years include the implementation of prostate specific antigen screening and the recognition that BRCA confers hereditary risk of prostate cancer. Quality of life and survivorship have driven risk stratification for localized prostate cancer, facilitated by molecular signatures and leading to increased acceptance of active surveillance as a mainstream treatment option. Advances in technology have improved efficacy and reduced toxicity in both radical prostatectomy and radiation therapy for localized prostate cancer. Improved understanding of the androgen receptor has yielded substantially more effective therapies. Future growth areas include personalized treatment based on genomic and genetic information, theranostics radiopharmaceuticals, and more aggressive treatment of metastatic disease to include focal therapy. Multidisciplinary management between specialized urologists, radiation oncologists, and medical oncologists remains central to maximizing patient outcomes.
Collapse
Affiliation(s)
- Tanya B Dorff
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center. Duarte, CA.
| | - Brock O'Neil
- Department of Urology, University of Utah Huntsman Comprehensive Cancer Center. Salt Lake City, UT
| | - Karen E Hoffman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center. Houston, TX
| | - Daniel W Lin
- Department of Urology, University of Washington, Seattle Cancer Care Alliance. Seattle, WA
| | - Kevin R Loughlin
- Vascular biology research laboratory, Boston Children's Hospital. Boston, MA
| | - Marc Dall'Era
- Department of Urology, University of California Davis Comprehensive Cancer Center. Davis, CA
| |
Collapse
|
412
|
Chandrasekar T, Kelly WK, Gomella LG. Overview of Prostate Cancer Genetic Testing. Urol Clin North Am 2021; 48:279-282. [PMID: 34210484 DOI: 10.1016/j.ucl.2021.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Thenappan Chandrasekar
- Department of Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1025 Walnut Street, Suite 1100, Philadelphia, PA 19107, USA.
| | - William K Kelly
- Medical Oncology and Urology, Division of Solid Tumor Oncology, Department of Medical Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Sidney Kimmel Cancer Center, 1025 Walnut Street, Suite 700, Philadelphia, PA 19107, USA
| | - Leonard G Gomella
- Department of Urology, Thomas Jefferson University and Hospital, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1025 Walnut Street, Suite 1100, Philadelphia, PA 19107, USA. https://twitter.com/LeonardGomella
| |
Collapse
|
413
|
Setton J, Zinda M, Riaz N, Durocher D, Zimmermann M, Koehler M, Reis-Filho JS, Powell SN. Synthetic Lethality in Cancer Therapeutics: The Next Generation. Cancer Discov 2021; 11:1626-1635. [PMID: 33795234 PMCID: PMC8295179 DOI: 10.1158/2159-8290.cd-20-1503] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/01/2021] [Accepted: 02/23/2021] [Indexed: 12/25/2022]
Abstract
Synthetic lethality (SL) provides a conceptual framework for tackling targets that are not classically "druggable," including loss-of-function mutations in tumor suppressor genes required for carcinogenesis. Recent technological advances have led to an inflection point in our understanding of genetic interaction networks and ability to identify a wide array of novel SL drug targets. Here, we review concepts and lessons emerging from first-generation trials aimed at testing SL drugs, discuss how the nature of the targeted lesion can influence therapeutic outcomes, and highlight the need to develop clinical biomarkers distinct from those based on the paradigms developed to target activated oncogenes. SIGNIFICANCE: SL offers an approach for the targeting of loss of function of tumor suppressor and DNA repair genes, as well as of amplification and/or overexpression of genes that cannot be targeted directly. A next generation of tumor-specific alterations targetable through SL has emerged from high-throughput CRISPR technology, heralding not only new opportunities for drug development, but also important challenges in the development of optimal predictive biomarkers.
Collapse
Affiliation(s)
- Jeremy Setton
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Nadeem Riaz
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniel Durocher
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | - Simon N Powell
- Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
414
|
Technical and biological constraints on ctDNA-based genotyping. Trends Cancer 2021; 7:995-1009. [PMID: 34219051 DOI: 10.1016/j.trecan.2021.06.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022]
Abstract
Circulating tumor DNA (ctDNA) enables real-time genomic profiling of cancer without the need for tissue biopsy. ctDNA-based technology is seeing rapid uptake in clinical practice due to the potential to inform patient management from diagnosis to advanced disease. In metastatic disease, ctDNA can identify somatic mutations, copy-number variants (CNVs), and structural rearrangements that are predictive of therapy response. However, the ctDNA fraction (ctDNA%) is unpredictable and confounds variant detection strategies, undermining confidence in liquid biopsy results. Assay design also influences which types of genomic alterations are identifiable. Here, we describe the relationships between ctDNA%, methodology, and sensitivity-specificity for major classes of genomic alterations in prostate cancer. We provide recommendations to navigate the technical complexities that constrain the detection of clinically relevant genomic alterations in ctDNA.
Collapse
|
415
|
Carthon B, Sibold HC, Blee S, D. Pentz R. Prostate Cancer: Community Education and Disparities in Diagnosis and Treatment. Oncologist 2021; 26:537-548. [PMID: 33683758 PMCID: PMC8265358 DOI: 10.1002/onco.13749] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/19/2021] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer remains the leading diagnosed cancer and the second leading cause of death among American men. Despite improvements in screening modalities, diagnostics, and treatment, disparities exist among Black men in this country. The primary objective of this systematic review is to describe the reported disparities in screening, diagnostics, and treatments as well as efforts to alleviate these disparities through community and educational outreach efforts. Critical review took place of retrospective, prospective, and socially descriptive data of English language publications in the PubMed database. Despite more advanced presentation, lower rates of screening and diagnostic procedures, and low rates of trial inclusion, subanalyses have shown that various modalities of therapy are quite effective in Black populations. Moreover, patients treated on prospective clinical trials and within equal-access care environments have shown similar outcomes regardless of race. Additional prospective studies and enhanced participation in screening, diagnostic and genetic testing, clinical trials, and community-based educational endeavors are important to ensure equitable progress in prostate cancer for all patients. IMPLICATIONS FOR PRACTICE: Notable progress has been made with therapeutic advances for prostate cancer, but racial disparities continue to exist. Differing rates in screening and utility in diagnostic procedures play a role in these disparities. Black patients often present with more advanced disease, higher prostate-specific antigen, and other adverse factors, but outcomes can be attenuated in trials or in equal-access care environments. Recent data have shown that multiple modalities of therapy are quite effective in Black populations. Novel and bold hypotheses to increase inclusion in clinical trial, enhance decentralized trial efforts, and enact successful models of patient navigation and community partnership are vital to ensure continued progress in prostate cancer disparities.
Collapse
Affiliation(s)
| | | | - Shannon Blee
- Winship Cancer Institute, Emory UniversityAtlantaGeorgiaUSA
| | | |
Collapse
|
416
|
Prostate Cancer Biomarkers: From diagnosis to prognosis and precision-guided therapeutics. Pharmacol Ther 2021; 228:107932. [PMID: 34174272 DOI: 10.1016/j.pharmthera.2021.107932] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/23/2022]
Abstract
Prostate cancer (PCa) is one of the most commonly diagnosed malignancies and among the leading causes of cancer-related death worldwide. It is a highly heterogeneous disease, ranging from remarkably slow progression or inertia to highly aggressive and fatal disease. As therapeutic decision-making, clinical trial design and outcome highly depend on the appropriate stratification of patients to risk groups, it is imperative to differentiate between benign versus more aggressive states. The incorporation of clinically valuable prognostic and predictive biomarkers is also potentially amenable in this process, in the timely prevention of metastatic disease and in the decision for therapy selection. This review summarizes the progress that has so far been made in the identification of the genomic events that can be used for the classification, prediction and prognostication of PCa, and as major targets for clinical intervention. We include an extensive list of emerging biomarkers for which there is enough preclinical evidence to suggest that they may constitute crucial targets for achieving significant advances in the management of the disease. Finally, we highlight the main challenges that are associated with the identification of clinically significant PCa biomarkers and recommend possible ways to overcome such limitations.
Collapse
|
417
|
Boilève A, Lavaud P, Caron O. Germline BRCA1 Mutation and Prostate Cancer: Be Careful on Causality. Eur Urol Oncol 2021; 4:674-675. [PMID: 34154978 DOI: 10.1016/j.euo.2021.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/29/2021] [Accepted: 06/10/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Alice Boilève
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France.
| | - Pernelle Lavaud
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Olivier Caron
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| |
Collapse
|
418
|
Schiewer MJ, Knudsen KE. Basic Science and Molecular Genetics of Prostate Cancer Aggressiveness. Urol Clin North Am 2021; 48:339-347. [PMID: 34210489 DOI: 10.1016/j.ucl.2021.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Androgen receptor function, tumor cell plasticity, loss of tumor suppressors, and defects in DNA repair genes affect aggressive features of prostate cancer. Prostate cancer development, progression, and aggressive behavior are often attributable to function of the androgen receptor. Tumor cell plasticity, neuroendocrine features, and loss of tumor suppressors lend aggressive behavior to prostate cancer cells. DNA repair defects have ramifications for prostate cancer cell behavior.
Collapse
Affiliation(s)
- Matthew J Schiewer
- Department of Urology, Urology Research Laboratory, Thomas Jefferson University, Sidney Kimmel Cancer Center, 233 South 10th Street BLSB 804, Philadelphia, PA 19107, USA; Department of Cancer Biology, Urology Research Laboratory, Thomas Jefferson University, Sidney Kimmel Cancer Center, 233 South 10th Street BLSB 804, Philadelphia, PA 19107, USA.
| | - Karen E Knudsen
- Department of Cancer Biology, Thomas Jefferson University, 233 South 10th Street BLSB 1050, Philadelphia, PA 19107, USA; Department of Urology, Thomas Jefferson University, 233 South 10th Street BLSB 1050, Philadelphia, PA 19107, USA; Department of Medical Oncology, Thomas Jefferson University, 233 South 10th Street BLSB 1050, Philadelphia, PA 19107, USA; Department of Radiation Oncology, Thomas Jefferson University, 233 South 10th Street BLSB 1050, Philadelphia, PA 19107, USA. https://twitter.com/SKCCDirector
| |
Collapse
|
419
|
Choo N, Ramm S, Luu J, Winter JM, Selth LA, Dwyer AR, Frydenberg M, Grummet J, Sandhu S, Hickey TE, Tilley WD, Taylor RA, Risbridger GP, Lawrence MG, Simpson KJ. High-Throughput Imaging Assay for Drug Screening of 3D Prostate Cancer Organoids. SLAS DISCOVERY 2021; 26:1107-1124. [PMID: 34111999 PMCID: PMC8458687 DOI: 10.1177/24725552211020668] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
New treatments are required for advanced prostate cancer; however, there are fewer preclinical models of prostate cancer than other common tumor types to test candidate therapeutics. One opportunity to increase the scope of preclinical studies is to grow tissue from patient-derived xenografts (PDXs) as organoid cultures. Here we report a scalable pipeline for automated seeding, treatment and an analysis of the drug responses of prostate cancer organoids. We established organoid cultures from 5 PDXs with diverse phenotypes of prostate cancer, including castrate-sensitive and castrate-resistant disease, as well as adenocarcinoma and neuroendocrine pathology. We robotically embedded organoids in Matrigel in 384-well plates and monitored growth via brightfield microscopy before treatment with poly ADP-ribose polymerase inhibitors or a compound library. Independent readouts including metabolic activity and live-cell imaging–based features provided robust measures of organoid growth and complementary ways of assessing drug efficacy. Single organoid analyses enabled in-depth assessment of morphological differences between patients and within organoid populations and revealed that larger organoids had more striking changes in morphology and composition after drug treatment. By increasing the scale and scope of organoid experiments, this automated assay complements other patient-derived models and will expedite preclinical testing of new treatments for prostate cancer.
Collapse
Affiliation(s)
- 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, VIC, Australia
| | - Susanne Ramm
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jennii Luu
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jean M Winter
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Freemason's Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA, Australia
| | - Luke A Selth
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Freemason's Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA, Australia.,Flinders Health and Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Amy R Dwyer
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Mark Frydenberg
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Australian Urology Associates, Melbourne, VIC, Australia.,Department of Urology, Cabrini Health, Malvern, VIC, Australia
| | - Jeremy Grummet
- Australian Urology Associates, Melbourne, VIC, Australia.,Epworth Healthcare, Melbourne, VIC, Australia.,Department of Surgery, Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Shahneen Sandhu
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Cancer Tissue Collection After Death (CASCADE) Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Freemason's Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA, Australia
| | - Renea A Taylor
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Physiology, Monash University, Clayton, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Melbourne Urological Research Alliance (MURAL), Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 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, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Melbourne Urological Research Alliance (MURAL), Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - 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, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Melbourne Urological Research Alliance (MURAL), Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Kaylene J Simpson
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
420
|
Abstract
Prostate cancer represents a significant health care burden in the United States due to its incidence, treatment-related morbidity, and cancer-specific mortality. The burden begins with prostate-specific antigen screening, which has been subject to controversy due to concerns of overdiagnosis and overtreatment. Advancements in molecular oncology have provided evidence for the inherited predisposition to prostate cancer, which could improve individualized, risk-adapted approaches to screening and mitigate the harms of routine screening. This review presents the current evidence for the genetic basis of prostate cancer and novel genetically informed, risk-adapted screening strategies for prostate cancer.
Collapse
|
421
|
Zhang J, Sun J, Bakht S, Hassan W. Recent Development and Future Prospects of Molecular Targeted Therapy in Prostate Cancer. Curr Mol Pharmacol 2021; 15:159-169. [PMID: 34102978 DOI: 10.2174/1874467214666210608141102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/25/2021] [Accepted: 04/05/2021] [Indexed: 11/22/2022]
Abstract
Prostate cancer (PC) is a rapidly increasing ailment worldwide. The previous decade has observed a rapid advancement in PC therapies that was evident from the number of FDA approvals during this phase. Androgen deprivation therapies (ADT) have traditionally remained a mainstay for the management of PCs, but the past decade has experienced the emergence of newer classes of drugs that can be used with or without the administration of ADT. FDA approved poly (ADP-ribose) polymerase inhibitors (PARPi), such as olaparib and rucaparib, after successful clinical trials against gene-mutated metastatic castration-resistant prostate cancer. Furthermore, drugs like apalutamide, darolutamide, and enzalutamide with an androgen-targeted mechanism of action have manifested superior results in non-metastatic castration-resistant prostate cancer (nmCRPC), metastatic castration-sensitive prostate cancer (mCSPC), and metastatic castration-resistant prostate cancer (mCRPC), respectively, with or without previously administered docetaxel. Relugolix, an oral gonadotropin-releasing hormone antagonist, and a combination of abiraterone acetate plus prednisone were also approved by FDA after a successful trial in advanced PC and mCRPC, respectively. This review aims to analyze the FDA-approved agents in PC during the last decade and provide a summary of their clinical trials. It also presents an overview of the ongoing progress of prospective molecules still under trial.
Collapse
Affiliation(s)
- Jinku Zhang
- Department of Pathology, First center Hospital of Baoding city, Hebei, 071000, China
| | - Jirui Sun
- Department of Pathology, First center Hospital of Baoding city, Hebei, 071000, China
| | - Sahar Bakht
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Waseem Hassan
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, 54000, Pakistan
| |
Collapse
|
422
|
Stein MK, Oluoha O, Patel K, VanderWalde A. Precision Medicine in Oncology: A Review of Multi-Tumor Actionable Molecular Targets with an Emphasis on Non-Small Cell Lung Cancer. J Pers Med 2021; 11:518. [PMID: 34198738 PMCID: PMC8226771 DOI: 10.3390/jpm11060518] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Precision medicine is essential for the modern care of a patient with cancer. Comprehensive molecular profiling of the tumor itself is necessary to determine the presence or absence of certain targetable abnormalities or biomarkers. In particular, lung cancer is a disease for which targetable genomic alterations will soon guide therapy in the majority of cases. In this comprehensive review of solid tumor-based biomarkers, we describe the genomic alterations for which targeted agents have been approved by the United States Food and Drug Administration (FDA). While focusing on alterations leading to approvals in a tumor-agnostic fashion (MSI-h, TMB-h, NTRK) and on those alterations with approvals in multiple malignancies (BRAF, ERBB2, RET, BRCA, PD-L1), we also describe several biomarkers or indications that are likely to lead to an approved drug in the near future (e.g., KRAS G12C, PD-L1 amplification, HER2 overexpression in colon cancer, HER2 mutations in lung cancer). Finally, we detail the current landscape of additional actionable alterations (EGFR, ALK, ROS1, MET) in lung cancer, a biomarker-rich malignancy that has greatly benefitted from the precision oncology revolution.
Collapse
Affiliation(s)
- Matthew K. Stein
- Missouri Baptist Medical Center, Heartland Cancer Research, NCI Community Oncology Research Program, St. Louis, MO 63131, USA;
| | - Oluchukwu Oluoha
- Division of Hematology and Oncology, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (O.O.); (K.P.)
| | - Kruti Patel
- Division of Hematology and Oncology, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (O.O.); (K.P.)
| | - Ari VanderWalde
- West Cancer Center and Research Institute, Germantown, TN 38138, USA
| |
Collapse
|
423
|
Schweizer MT, Sivakumar S, Tukachinsky H, Coleman I, De Sarkar N, Yu EY, Konnick EQ, Nelson PS, Pritchard CC, Montgomery B. Concordance of DNA Repair Gene Mutations in Paired Primary Prostate Cancer Samples and Metastatic Tissue or Cell-Free DNA. JAMA Oncol 2021; 7:2780857. [PMID: 34086042 PMCID: PMC8446811 DOI: 10.1001/jamaoncol.2021.2350] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/03/2021] [Indexed: 12/20/2022]
Abstract
IMPORTANCE DNA damage repair (DDR) gene mutations represent actionable alterations that can guide precision medicine strategies for advanced prostate cancer. However, acquisition of contemporary tissue samples for molecular testing can be a barrier to deploying precision medicine approaches. We hypothesized that most DDR alterations represent truncal events in prostate cancer and that primary tissue would faithfully reflect mutations found in cell-free circulating tumor DNA (ctDNA) and/or metastatic tissue. OBJECTIVE To assess concordance in DDR gene alterations between primary prostate cancer and metastases or ctDNA specimens. DESIGN, SETTING, AND PARTICIPANTS Patients were included if a DDR pathway mutation was detected in metastatic tissue or ctDNA and primary tissue sequencing was available for comparison. Sequencing data from 3 cohorts were analyzed: (1) FoundationOne, (2) University of Washington clinical cases (University of Washington-OncoPlex or Stand Up to Cancer-Prostate Cancer Foundation International Dream Team sequencing pipelines), and (3) University of Washington rapid autopsy series. Only pathogenic somatic mutations were included, and more than 30 days between primary tumor tissue and ctDNA and/or metastatic tissue acquisition was required. Clonal hematopoiesis of indeterminate potential (CHIP) and germline events were adjudicated by an expert molecular pathologist and excluded. MAIN OUTCOMES AND MEASURES The DDR gene alterations detected in primary prostate tissue matched with metastatic tissue and/or ctDNA findings. RESULTS A total of 72 men with known DDR alterations were included in the analysis, and primary samples with paired ctDNA and/or metastatic tissue were sequenced. After excluding patients with ctDNA where only CHIP and/or germline events (n = 21) were observed, 51 patients remained and were included in the final analysis. The median (range) time from acquisition of primary tissue to acquisition of ctDNA or tumor tissue was 55 (5-193) months. Concordance in DDR gene mutation status across samples was 84% (95% CI, 71%-92%). Rates of concordance between metastatic-primary and ctDNA-primary pairs were similar when patients with CHIP events were excluded. Multiclonal BRCA2 reversion mutations associated with resistance to PARP inhibitors and platinum chemotherapy were detected in ctDNA from 2 patients. CONCLUSIONS AND RELEVANCE In this genetic association study of 3 patient cohorts, primary prostate tissue accurately reflected the mutational status of actionable DDR genes in metastatic tissue, consistent with DDR alterations being truncal in most patients. After excluding likely CHIP events, ctDNA profiling accurately captured these DDR mutations while also detecting reversion alterations that may suggest resistance mechanisms.
Collapse
Affiliation(s)
- Michael T. Schweizer
- Division of Oncology, Department of Medicine, University of Washington, Seattle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | | | - Ilsa Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Navonil De Sarkar
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Evan Y. Yu
- Division of Oncology, Department of Medicine, University of Washington, Seattle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Eric Q. Konnick
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
| | - Peter S. Nelson
- Division of Oncology, Department of Medicine, University of Washington, Seattle
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Colin C. Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
- Brotman Baty Institute for Precision Medicine, Seattle, Washington
| | - Bruce Montgomery
- Division of Oncology, Department of Medicine, University of Washington, Seattle
- Prostate Cancer Foundation Precision Oncology Program for Cancer of the Prostate,VA Puget Sound Health Care System, Seattle, Washington
| |
Collapse
|
424
|
Homologous recombination deficiency (HRD) score in germline BRCA2- versus ATM-altered prostate cancer. Mod Pathol 2021; 34:1185-1193. [PMID: 33462368 PMCID: PMC8154637 DOI: 10.1038/s41379-020-00731-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/14/2020] [Accepted: 12/14/2020] [Indexed: 01/28/2023]
Abstract
The homologous recombination deficiency (HRD) score integrates three DNA-based measures of genomic instability, and has been understudied in prostate cancer. Given the recent FDA approval of two PARP inhibitors for prostate cancer, HRD score analysis could help to refine treatment selection. We assessed HRD score (defined as the sum of loss-of-heterozygosity, telomeric allelic imbalance, and large-scale state transitions) in three cohorts of primary prostate cancer, including a Johns Hopkins University (JHU) cohort with germline mutations in BRCA2, ATM, or CHEK2 (n = 64), the TCGA cohort (n = 391), and the PROGENE cohort (n = 102). In the JHU cohort, tumors with germline BRCA2 mutations had higher HRD scores (median = 27) than those with germline ATM or CHEK2 mutations (median = 16.5 [p = 0.029] and 9 [p < 0.001], respectively). For TCGA tumors without underlying HR pathway mutations, the median HRD score was 11, significantly lower than ovarian carcinoma lacking BRCA1/2 mutations (median = 28). In the absence of HR gene mutations, the median HRD score was unexpectedly higher among prostate cancers with TP53 mutations versus those without (17 vs. 11; p = 0.015); this finding was confirmed in the PROGENE cohort (24 vs. 16; p = 0.001). Finally, among eight BRCA2-altered patients who received olaparib, progression-free survival trended longer in those with HRD scores above versus below the median (14.9 vs. 9.9 months). We conclude that HRD scores are low in primary prostate cancer and higher in cases with germline BRCA2 or somatic TP53 mutations. Germline BRCA2-altered cases have significantly higher HRD scores than germline ATM-altered or CHEK2-altered cases, consistent with the lower efficacy of PARP inhibitors among the latter.
Collapse
|
425
|
Westaby D, Viscuse PV, Ravilla R, de la Maza MDLDF, Hahn A, Sharp A, de Bono J, Aparicio A, Fleming MT. Beyond the Androgen Receptor: The Sequence, the Mutants, and New Avengers in the Treatment of Castrate-Resistant Metastatic Prostate Cancer. Am Soc Clin Oncol Educ Book 2021; 41:e190-e202. [PMID: 34061561 DOI: 10.1200/edbk_321209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Targeting the androgen receptor by depriving testosterone with gonadotropin-releasing hormone agonists or antagonists, or surgical castration, has been the backbone of metastatic prostate cancer treatment. Although most prostate cancers initially respond to androgen deprivation, metastatic castration-resistant prostate cancer evolves into a heterogeneous disease with diverse drivers of progression and mechanisms of therapeutic resistance. Development of castrate resistance phenotype is associated with lethality despite the recent noteworthy strides gained via increase in therapeutic options. Identification of novel therapeutics to further improve survival and achieve durable responses in metastatic castration-resistant prostate cancer is a clinical necessity. In this review, we outline the existing avengers for treatment of metastatic castration-resistant prostate cancer by clinical presentation, placing into context the clinical state of the patient, such as burden of disease and symptoms. Doing so might aid in the ability to optimize the sequence of agents and allow for maximal exposure to life-prolonging therapeutics. Realizing the limitations of the androgen signaling inhibition, we explore the androgen-indifferent prostate cancer: the mutants. Classically, these subtypes have been associated with variant histology, but androgen-indifferent prostate cancer features are now frequently observed in association with heterogeneous morphologies, including double-negative prostate cancers, lacking both androgen receptor and neuroendocrine features, or clinicopathologic criteria, such as the aggressive variant prostate cancer criteria. The framework of new avengers against metastatic castration-resistant prostate cancer based on mechanism, including DNA repair, immune checkpoint inhibition, PTEN/PI3K/AKT pathway, prostate-specific membrane antigen targets, bispecific T-cell engagers, and radionuclide therapies, is summarized in this review.
Collapse
Affiliation(s)
- Daniel Westaby
- The Institute of Cancer Research and The Royal Marsden Hospital, London, United Kingdom
| | - Paul V Viscuse
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rahul Ravilla
- US Oncology Research, New York Oncology Hematology, Albany, NY
| | | | - Andrew Hahn
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Adam Sharp
- The Institute of Cancer Research and The Royal Marsden Hospital, London, United Kingdom
| | - Johann de Bono
- The Institute of Cancer Research and The Royal Marsden Hospital, London, United Kingdom
| | - Ana Aparicio
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mark T Fleming
- US Oncology Research, Virginia Oncology Associates, Norfolk, VA
| |
Collapse
|
426
|
Dabkara D, Mondal D, Ghosh J, Biswas B, Ganguly S. How I Treat Metastatic Hormone-Sensitive Prostate Cancer? Indian J Med Paediatr Oncol 2021. [DOI: 10.1055/s-0041-1729725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
AbstractAndrogen deprivation therapy (ADT) combined with docetaxel or antiandrogens (abiraterone, enzalutamide, or apalutamide) improved the outcomes in men with metastatic hormone-sensitive prostate cancer (mHSPC). When multiple options are available, the dilemma remains how to choose among these options. Similarly, issues of bone health, long-term side effects of therapies, and hereditary risk need to be discussed for comprehensive care. In the present article, we reviewed the relevant evidence for the treatment of mHSPC. ADT alone is not the current standard of care for most patients. In these times of plenty and price crisis, it is imperative to find the best option for treating these patients.
Collapse
Affiliation(s)
- Deepak Dabkara
- Department of Medical Oncology, Tata Medical Center, Kolkata, West Bengal, India
| | - Debapriya Mondal
- Department of Medical Oncology, Tata Medical Center, Kolkata, West Bengal, India
| | - Joydeep Ghosh
- Department of Medical Oncology, Tata Medical Center, Kolkata, West Bengal, India
| | - Bivas Biswas
- Department of Medical Oncology, Tata Medical Center, Kolkata, West Bengal, India
| | - Sandip Ganguly
- Department of Medical Oncology, Tata Medical Center, Kolkata, West Bengal, India
| |
Collapse
|
427
|
Zhou Q, Liu M, Shao T, Xie P, Zhu S, Wang W, Miao Q, Peng J, Zhang P. TPX2 Enhanced the Activation of the HGF/ETS-1 Pathway and Increased the Invasion of Endocrine-Independent Prostate Carcinoma Cells. Front Oncol 2021; 11:618540. [PMID: 34123781 PMCID: PMC8193931 DOI: 10.3389/fonc.2021.618540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 04/06/2021] [Indexed: 01/16/2023] Open
Abstract
The prognosis for endocrine-independent prostate carcinoma is still poor due to its highly metastatic feature. In the present work, TPX2 (the targeting protein for Xklp2), which is known as a micro-tubulin interacted protein, was identified as a novel coactivator of ETS-1, a transcription factor that plays a central role in mediating the metastasis of human malignancies. TPX2 enhanced the transcription factor activation of ETS-1 and increased the expression of ETS-1's downstream metastasis-related genes, such as mmp3 or mmp9, induced by HGF (hepatocyte growth factor), a typical agonist of the HGF/c-MET/ETS-1 pathway. The protein-interaction between TPX2 and ETS-1 was examined using immunoprecipitation (IP). TPX2 enhanced the accumulation of ETS-1 in the nuclear and the recruitment of its binding element (EST binding site, EBS) located in the promoter region of its downstream gene, mmp9. Moreover, TPX2 enhanced the in vitro or in vivo invasion of a typical endocrine-independent prostate carcinoma cell line, PC-3. Therefore, TPX2 enhanced the activation of the HGF/ETS-1 pathway to enhance the invasion of endocrine-independent prostate carcinoma cells and thus it would be a promising target for prostate carcinoma treatment.
Collapse
Affiliation(s)
- Qinghong Zhou
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Mingsheng Liu
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Tao Shao
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Pingbo Xie
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Shaojie Zhu
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Wei Wang
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Qiong Miao
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Jiaxi Peng
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Peng Zhang
- Department of Urology, Chinese People's Liberation Army (PLA) General Hospital/Chinese PLA Medical Academy, Beijing, China
| |
Collapse
|
428
|
Cimadamore A, Cheng L, Massari F, Santoni M, Pepi L, Franzese C, Scarpelli M, Lopez-Beltran A, Galosi AB, Montironi R. Circulating Tumor DNA Testing for Homology Recombination Repair Genes in Prostate Cancer: From the Lab to the Clinic. Int J Mol Sci 2021; 22:5522. [PMID: 34073818 PMCID: PMC8197269 DOI: 10.3390/ijms22115522] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/24/2022] Open
Abstract
Approximately 23% of metastatic castration-resistant prostate cancers (mCRPC) harbor deleterious aberrations in DNA repair genes. Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) therapy has shown improvements in overall survival in patients with mCRPC who harbor somatic and/or germline alterations of homology recombination repair (HRR) genes. Peripheral blood samples are typically used for the germline mutation analysis test using the DNA extracted from peripheral blood leucocytes. Somatic alterations can be assessed by extracting DNA from a tumor tissue sample or using circulating tumor DNA (ctDNA) extracted from a plasma sample. Each of these genetic tests has its own benefits and limitations. The main advantages compared to the tissue test are that liquid biopsy is a non-invasive and easily repeatable test with the value of better representing tumor heterogeneity than primary biopsy and of capturing changes and/or resistance mutations in the genetic tumor profile during disease progression. Furthermore, ctDNA can inform about mutation status and guide treatment options in patients with mCRPC. Clinical validation and test implementation into routine clinical practice are currently very limited. In this review, we discuss the state of the art of the ctDNA test in prostate cancer compared to blood and tissue testing. We also illustrate the ctDNA testing workflow, the available techniques for ctDNA extraction, sequencing, and analysis, describing advantages and limits of each techniques.
Collapse
Affiliation(s)
- Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (L.P.); (M.S.)
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA;
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy;
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | - Laura Pepi
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (L.P.); (M.S.)
| | - Carmine Franzese
- Department of Specialist Clinical Science and Odontostomatology, Urology Division, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (C.F.); (A.B.G.)
| | - Marina Scarpelli
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (L.P.); (M.S.)
| | - Antonio Lopez-Beltran
- Department of Morphological Sciences, Cordoba University Medical School, 14071 Cordoba, Spain;
| | - Andrea Benedetto Galosi
- Department of Specialist Clinical Science and Odontostomatology, Urology Division, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (C.F.); (A.B.G.)
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (L.P.); (M.S.)
| |
Collapse
|
429
|
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
|
430
|
BRCA Germline Mutations in Prostate Cancer: The Future Is Tailored. Diagnostics (Basel) 2021; 11:diagnostics11050908. [PMID: 34069669 PMCID: PMC8161324 DOI: 10.3390/diagnostics11050908] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/08/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer (PCa) is the second most common neoplasm in men and the fifth leading cause of death worldwide [...].
Collapse
|
431
|
Chi J, Chung SY, Parakrama R, Fayyaz F, Jose J, Saif MW. The role of PARP inhibitors in BRCA mutated pancreatic cancer. Therap Adv Gastroenterol 2021; 14:17562848211014818. [PMID: 34025781 PMCID: PMC8120537 DOI: 10.1177/17562848211014818] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/14/2021] [Indexed: 02/04/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) accounts for about 3% of all cancers in the United States and about 7% of all cancer deaths. Despite the lower prevalence relative to other solid tumors, it is one of the leading causes of cancer-related death in the US. PDAC is highly resistant to chemotherapy as well as radiation therapy. Current standard-of-care chemotherapeutic regimens provide transient disease control but eventually tumors develop chemoresistance. Tumors that are deficient in DNA damage repair mechanisms such as BRCA mutants respond better to platinum-based chemotherapies. However, these tumor cells can utilize the poly adenosine diphosphate (ADP)-ribose polymerase (PARP) as a salvage DNA repair pathway to prolong survival. Hence, in the presence of BRCA mutations, the inhibition of the PARP pathway can lead to tumor cell death. This provides the rationale for using PARP inhibitors in patients with BRCA mutated PDAC. The phase III POLO trial showed a near doubling of progression-free survival (PFS) compared with placebo in advanced PDAC when a PARP inhibitor, olaparib, was used as maintenance therapy. As a result, the US Food and Drug Administration (FDA) approved olaparib as a maintenance treatment for germline BRCA mutated advanced PDAC that has not progressed on platinum-based chemotherapy. The success of olaparib in treating advanced PDAC opened the new field for utilizing PARP inhibitors in patients with DNA damage repair (DDR) gene defects. Currently, many clinical trials with various PARP inhibitors are ongoing either as monotherapy or in combination with other agents. In addition to germline/somatic BRCA mutations, some trials are enrolling patients with defects in other DDR genes such as ATM, PALB2, and CHEK2. With many ongoing PARP inhibitor trials, it is hopeful that the management of PDAC will continuously evolve and eventually lead to improved patient outcomes.
Collapse
Affiliation(s)
- Jeffrey Chi
- Northwell Health Cancer Institute, Donald and Barbara Zucker School of Medicine at Hofstra, Feinstein Institute for Medical Research, Lake Success, NY, USA
| | - Su Yun Chung
- Northwell Health Cancer Institute, Donald and Barbara Zucker School of Medicine at Hofstra, Feinstein Institute for Medical Research, Lake Success, NY, USA
| | - Ruwan Parakrama
- Northwell Health Cancer Institute, Donald and Barbara Zucker School of Medicine at Hofstra, Feinstein Institute for Medical Research, Lake Success, NY, USA
| | - Fatima Fayyaz
- Northwell Health Cancer Institute, Donald and Barbara Zucker School of Medicine at Hofstra, Feinstein Institute for Medical Research, Lake Success, NY, USA
| | - Jyothi Jose
- Northwell Health Cancer Institute, Donald and Barbara Zucker School of Medicine at Hofstra, Feinstein Institute for Medical Research, Lake Success, NY, USA
| | - Muhammad Wasif Saif
- Northwell Health Cancer Institute, Donald and Barbara Zucker School of Medicine, Feinstein Institute of Research, Lake Success, NY 11042, USA
| |
Collapse
|
432
|
Doan DK, Schmidt KT, Chau CH, Figg WD. Germline Genetics of Prostate Cancer: Prevalence of Risk Variants and Clinical Implications for Disease Management. Cancers (Basel) 2021; 13:cancers13092154. [PMID: 33947030 PMCID: PMC8124444 DOI: 10.3390/cancers13092154] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/30/2022] Open
Abstract
Prostate cancer has entered into the era of precision medicine with the recent approvals of targeted therapeutics (olaparib and rucaparib). The presence of germline mutations has important hereditary cancer implications for patients with prostate cancer, and germline testing is increasingly important in cancer screening, risk assessment, and the overall treatment and management of the disease. In this review, we discuss germline variants associated with inherited predisposition, prostate cancer risk and outcomes. We review recommendations for germline testing, available testing platforms, genetic counseling as well as discuss the therapeutic implications of germline variants relevant to prostate cancer treatments. Understanding the role of germline (heritable) mutations that affect prostate cancer biology and risk as well as the subsequent effect of these alterations on potential therapies is critical as the treatment paradigm shifts towards precision medicine. Furthermore, enhancing patient education tactics and healthcare system infrastructure is essential for the utilization of relevant predictive biomarkers and the improvement of clinical outcomes of patients with prostate cancer or at high risk of developing the disease.
Collapse
Affiliation(s)
| | - Keith T. Schmidt
- Clinical Pharmacology Program, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA;
| | - Cindy H. Chau
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA;
| | - William D. Figg
- Clinical Pharmacology Program, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA;
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA;
- Correspondence: ; Tel.: +1-240-760-6179; Fax: +1-240-858-3020
| |
Collapse
|
433
|
Shore N, Ionescu-Ittu R, Yang L, Laliberté F, Mahendran M, Lejeune D, Yu L, Burgents J, Duh MS, Ghate SR. Real-world genetic testing patterns in metastatic castration-resistant prostate cancer. Future Oncol 2021; 17:2907-2921. [PMID: 33906368 DOI: 10.2217/fon-2021-0153] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To assess the patterns of genetic testing for homologous recombination repair mutations in patients with metastatic castration-resistant prostate cancer (mCRPC) pre-PARP inhibitors approval. Patients & methods: mCRPC patients were selected in an oncology electronic medical records database. Patterns and predictors of testing for ATM, BRCA1/2, CDK12, PALB2 and FANCA gene alterations were assessed. Results: Of 5213 mCRPC patients, 674 (13%) had a documented genetic test. The number of tested patients increased from 1 in 2013 to 313 in 2018 (out of 3161 and 3010 clinically active patients, respectively). Receiving care in an academic oncology center (versus a community-based center) strongly predicted genetic testing (hazard ratio = 2.41). Conclusion: The use of and access to genetic testing pre-PARP inhibitor approval was suboptimal.
Collapse
Affiliation(s)
- Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC 29572, USA
| | | | | | | | | | | | - Louise Yu
- Analysis Group, Inc., Boston, MA 02199, USA
| | | | | | | |
Collapse
|
434
|
Cabañas Morafraile E, Pérez-Peña J, Fuentes-Antrás J, Manzano A, Pérez-Segura P, Pandiella A, Galán-Moya EM, Ocaña A. Genomic Correlates of DNA Damage in Breast Cancer Subtypes. Cancers (Basel) 2021; 13:cancers13092117. [PMID: 33925616 PMCID: PMC8123819 DOI: 10.3390/cancers13092117] [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: 03/24/2021] [Revised: 04/20/2021] [Accepted: 04/24/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Breast cancer (BC) is the most common invasive tumor in women and the second leading cause of cancer-related death. Therefore, identification of druggable targets to improve current therapies and overcome resistance is a major goal. In this work, we performed an in silico analysis of transcriptomic datasets in breast cancer, and focused on those involved in DNA damage, as were clearly upregulated using gene set enrichment analyses (GSEA), particular the following pathways: ATM/ATR, BARD1 and Fanconi Anemia. BHLHE40, RFWD2, BRIP1, PRKDC, NBN, RNF8, FANCD2, RAD1, BLM, DCLRE1C, UBE2T, CSTF1, MCM7, RFC4, YWHAB, YWHAZ, CDC6, CCNE1, and FANCI genes were amplified/overexpressed in BC, and correlated with detrimental prognosis. Finally, we selected the best transcriptomic signature of genes within this function that associated with clinical outcome to identify functional genomic correlates of outcome. Abstract Among the described druggable vulnerabilities, acting on the DNA repair mechanism has gained momentum, with the approval of PARP inhibitors in several indications, including breast cancer. However, beyond the mere presence of BRCA1/BRCA2 mutations, the identification of additional biomarkers that would help to select tumors with an extreme dependence on DNA repair machinery would help to stratify therapeutic decisions. Gene set enrichment analyses (GSEA) using public datasets evaluating expression values between normal breast tissue and breast cancer identified a set of upregulated genes. Genes included in different pathways, such as ATM/ATR, BARD1, and Fanconi Anemia, which are involved in the DNA damage response, were selected and confirmed using molecular alterations data contained at cBioportal. Nineteen genes from these gene sets were identified to be amplified and upregulated in breast cancer but only five of them NBN, PRKDC, RFWD2, UBE2T, and YWHAZ meet criteria in all breast cancer molecular subtypes. Correlation of the selected genes with prognosis (relapse free survival, RFS, and overall survival, OS) was performed using the KM Plotter Online Tool. In last place, we selected the best signature of genes within this process whose upregulation can be indicative of a more aggressive phenotype and linked with worse outcome. In summary, we identify genomic correlates within DNA damage pathway associated with prognosis in breast cancer.
Collapse
Affiliation(s)
- Esther Cabañas Morafraile
- Experimental Therapeutics Unit, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC) and Centro de Investigación Biomédica en Red en Oncología (CIBERONC), 28040 Madrid, Spain; (E.C.M.); (J.F.-A.); (A.M.); (P.P.-S.)
| | - Javier Pérez-Peña
- Instituto de Biología Molecular y Celular del Cáncer del CSIC, IBSAL and CIBERONC, 37007 Salamanca, Spain; (J.P.-P.); (A.P.)
| | - Jesús Fuentes-Antrás
- Experimental Therapeutics Unit, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC) and Centro de Investigación Biomédica en Red en Oncología (CIBERONC), 28040 Madrid, Spain; (E.C.M.); (J.F.-A.); (A.M.); (P.P.-S.)
| | - Aránzazu Manzano
- Experimental Therapeutics Unit, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC) and Centro de Investigación Biomédica en Red en Oncología (CIBERONC), 28040 Madrid, Spain; (E.C.M.); (J.F.-A.); (A.M.); (P.P.-S.)
| | - Pedro Pérez-Segura
- Experimental Therapeutics Unit, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC) and Centro de Investigación Biomédica en Red en Oncología (CIBERONC), 28040 Madrid, Spain; (E.C.M.); (J.F.-A.); (A.M.); (P.P.-S.)
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer del CSIC, IBSAL and CIBERONC, 37007 Salamanca, Spain; (J.P.-P.); (A.P.)
| | - Eva M. Galán-Moya
- Translational Oncology Laboratory, Centro Regional de Investigaciones Biomédicas (CRIB) and Nursery School, Campus de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain;
| | - Alberto Ocaña
- Experimental Therapeutics Unit, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC) and Centro de Investigación Biomédica en Red en Oncología (CIBERONC), 28040 Madrid, Spain; (E.C.M.); (J.F.-A.); (A.M.); (P.P.-S.)
- Translational Oncology Laboratory, Centro Regional de Investigaciones Biomédicas (CRIB) and Nursery School, Campus de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain;
- Correspondence:
| |
Collapse
|
435
|
Kwan EM, Dai C, Fettke H, Hauser C, Docanto MM, Bukczynska P, Ng N, Foroughi S, Graham LJK, Mahon K, Tan W, Wang X, Zhao Z, Zheng T, Zhou K, Yu J, Du P, Horvath LG, Jia S, Kohli M, Azad AA. Plasma Cell-Free DNA Profiling of PTEN-PI3K-AKT Pathway Aberrations in Metastatic Castration-Resistant Prostate Cancer. JCO Precis Oncol 2021; 5:PO.20.00424. [PMID: 34250422 PMCID: PMC8232889 DOI: 10.1200/po.20.00424] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/10/2021] [Accepted: 02/24/2021] [Indexed: 12/13/2022] Open
Abstract
Tumor tissue from metastatic castration-resistant prostate cancer (mCRPC) harbors frequent copy number variations (CNVs) in the PTEN-PI3K-AKT pathway. However, identifying CNVs in plasma cell-free DNA (cfDNA) has proven to be challenging. With emerging data supporting Akt inhibition in PTEN-deficient mCRPC, we profiled PTEN-PI3K-AKT pathway aberrations in patients with mCRPC using a novel cfDNA assay optimized for CNV detection. METHODS A next-generation sequencing-based cfDNA assay was used to profile 231 patients with mCRPC from two independent cohorts (Australian, n = 78; United States, n = 153). PTEN-PI3K-AKT pathway genomic aberrations were correlated with clinical outcomes, including progression-free survival and overall survival (OS). RESULTS PTEN loss and PIK3CA gain were detected in 37% (85 of 231) and 17% (39 of 231) of patients, respectively. Poorer outcomes were observed in patients with PTEN-PI3K-AKT pathway aberrations, including those with dual PTEN loss and PIK3CA gain (hazard ratio 2.3, 95% CI 1.2 to 4.4). Cumulative CNV burden in the PTEN-PI3K-AKT and androgen receptor (AR) pathways was associated with significantly worse clinical outcomes (0 v 1 v ≥ 2 CNVs in Australian cohort: median OS 33.5 v 17.2 v 9.7 months, P < .001; 0 v 1 v ≥ 2 CNVs in US cohort: median OS 35.5 v 14.3 v 9.2 months, P < .001). Notably, 21% (31 of 146) of PTEN-neutral patients harbored alternative PTEN-PI3K-AKT pathway aberrations. CONCLUSION PTEN-PI3K-AKT pathway CNVs were readily detected using our cfDNA assay, with the prevalence of PTEN loss comparable with tissue-based studies. Additional PTEN-PI3K-AKT pathway aberrations were found in one fifth of PTEN-neutral cases. Concurrent CNVs in the PTEN-PI3K-AKT and AR pathways portended poor survival, and identifying this high-risk patient subset for dual AR/Akt inhibition may optimize precision treatment with Akt inhibitors in mCRPC.
Collapse
Affiliation(s)
- Edmond M. Kwan
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia
- Department of Medical Oncology, Monash Health, Melbourne, Australia
| | | | - Heidi Fettke
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia
| | | | - Maria M. Docanto
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia
| | | | - Nicole Ng
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Personalized Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - Siavash Foroughi
- Personalized Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | | | - Kate Mahon
- Medical Oncology, Chris O'Brien Lifehouse, Sydney, Australia
- University of Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Sydney, Australia
| | - Winston Tan
- Division of Medical Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL
| | | | | | | | | | | | - Pan Du
- Predicine Inc, Hayward, CA
| | - Lisa G. Horvath
- Medical Oncology, Chris O'Brien Lifehouse, Sydney, Australia
- University of Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Sydney, Australia
- Royal Prince Alfred Hospital, Sydney, Australia
| | | | - Manish Kohli
- Division of Medical Oncology, Department of Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Arun A. Azad
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
436
|
Teyssonneau D, Margot H, Cabart M, Anonnay M, Sargos P, Vuong NS, Soubeyran I, Sevenet N, Roubaud G. Prostate cancer and PARP inhibitors: progress and challenges. J Hematol Oncol 2021; 14:51. [PMID: 33781305 PMCID: PMC8008655 DOI: 10.1186/s13045-021-01061-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/10/2021] [Indexed: 12/22/2022] Open
Abstract
Despite survival improvements achieved over the last two decades, prostate cancer remains lethal at the metastatic castration-resistant stage (mCRPC) and new therapeutic approaches are needed. Germinal and/or somatic alterations of DNA-damage response pathway genes are found in a substantial number of patients with advanced prostate cancers, mainly of poor prognosis. Such alterations induce a dependency for single strand break reparation through the poly(adenosine diphosphate-ribose) polymerase (PARP) system, providing the rationale to develop PARP inhibitors. In solid tumors, the first demonstration of an improvement in overall survival was provided by olaparib in patients with mCRPC harboring homologous recombination repair deficiencies. Although this represents a major milestone, a number of issues relating to PARP inhibitors remain. This timely review synthesizes and discusses the rationale and development of PARP inhibitors, biomarker-based approaches associated and the future challenges related to their prescription as well as patient pathways.
Collapse
Affiliation(s)
- Diego Teyssonneau
- Department of Medical Oncology, Institut Bergonie, Bordeaux, France.
| | - Henri Margot
- Department of Genetic, Institut Bergonie, Bordeaux, France
| | - Mathilde Cabart
- Department of Medical Oncology, Institut Bergonie, Bordeaux, France
| | - Mylène Anonnay
- Department of Medical Oncology, Institut Bergonie, Bordeaux, France
| | - Paul Sargos
- Department of Radiotherapy, Institut Bergonie, Bordeaux, France
| | - Nam-Son Vuong
- Department of Urology, Clinique Saint-Augustin, Bordeaux, France
| | | | | | - Guilhem Roubaud
- Department of Medical Oncology, Institut Bergonie, Bordeaux, France
| |
Collapse
|
437
|
Dror CM, Wyatt AW, Chi KN. Olaparib for the treatment of metastatic prostate cancer. Future Oncol 2021; 17:2413-2429. [PMID: 33769071 DOI: 10.2217/fon-2020-1245] [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] Open
Abstract
Recent innovations in the treatment of metastatic prostate cancer have improved patient outcomes. Nonetheless, this disease remains fatal and additional treatment approaches are needed. Greater understanding of the molecular landscape of metastatic prostate cancer has revealed recurrent alterations in key pathways amenable to therapeutic targeting. One such pathway is DNA repair, particularly alterations in genes directly or indirectly associated with homologous recombination repair found in up to one-quarter of patients with metastatic castrate-resistant prostate cancer (mCRPC). Olaparib, an inhibitor of poly-ADP-ribose polymerase, has recently gained approval for the treatment of mCRPC harboring alterations in homologous recombination repair genes. This review will provide a summary of evidence regarding PARP inhibition in the treatment of mCRPC, with a specific focus on olaparib.
Collapse
Affiliation(s)
| | - Alexander W Wyatt
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V5Z 4S6, Canada.,Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 4S6, Canada
| | - Kim N Chi
- BC Cancer, Vancouver, Vancouver, BC, V5Z 4S6, Canada.,Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V5Z 4S6, Canada
| |
Collapse
|
438
|
Leung DKW, Chiu PKF, Ng CF, Teoh JYC. Novel Strategies for Treating Castration-Resistant Prostate Cancer. Biomedicines 2021; 9:biomedicines9040339. [PMID: 33801751 PMCID: PMC8066514 DOI: 10.3390/biomedicines9040339] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 01/06/2023] Open
Abstract
The development of castration resistance is an inevitable pathway for the vast majority of patients with advanced prostate cancer. Recently, there have been significant breakthroughs in the understanding and management options of castration-resistant prostate cancer. Three novel hormonal agents showed survival benefits in non-metastatic patients. As for metastatic disease, there was an even wider range of management options being investigated. This review summarized advances in the management of castration-resistant prostate cancer (CRPC) including emerging data on novel imaging techniques and treatment strategies.
Collapse
|
439
|
Robinson AG, Izard JP, Vera-Badillo FE. Treatment and Patient Selection for Patients with Metastatic Castration-resistant Prostate After Progression on Docetaxel and Abiraterone/Enzalutamide: When to Play Your CARD and When to Do Your PARP. Eur Urol 2021; 80:123-126. [PMID: 33773874 DOI: 10.1016/j.eururo.2021.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/02/2021] [Indexed: 10/21/2022]
Abstract
For patients with metastatic castration-resistant prostate cancer (mCRPC) progressing after an androgen receptor axis-targeted therapy and docetaxel, poly (ADP-ribose) polymerase (PARP) inhibitors and chemotherapy with cabazitaxel have shown promise. We address the trials for the two approaches and consider possible sequencing of these drugs. We suggest that only patients with a BRCA2 mutation should receive a PARP inhibitor, and docetaxel or cabazitaxel should be favored in the absence of BRCA2 alterations, provided the patient is naïve to these drugs.
Collapse
Affiliation(s)
- Andrew G Robinson
- Division of Medical Oncology, Department of Oncology, Queen's University, Kingston, ON, Canada
| | - Jason P Izard
- Department of Urology, Queen's University, Kingston, ON, Canada
| | | |
Collapse
|
440
|
Saad F, Chi KN, Shore ND, Graff JN, Posadas EM, Lattouf JB, Espina BM, Zhu E, Yu A, Hazra A, De Meulder M, Mamidi RNVS, Bradic B, Francis P, Hayreh V, Rezazadeh Kalebasty A. Niraparib with androgen receptor-axis-targeted therapy in patients with metastatic castration-resistant prostate cancer: safety and pharmacokinetic results from a phase 1b study (BEDIVERE). Cancer Chemother Pharmacol 2021; 88:25-37. [PMID: 33754187 PMCID: PMC8149334 DOI: 10.1007/s00280-021-04249-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/12/2021] [Indexed: 12/19/2022]
Abstract
Purpose To assess the safety and pharmacokinetics and determine the recommended phase 2 dose (RP2D) of niraparib with apalutamide or abiraterone acetate plus prednisone (AAP) in patients with metastatic castration-resistant prostate cancer (mCRPC). Methods BEDIVERE was a multicenter, open-label, phase 1b study of niraparib 200 or 300 mg/day with apalutamide 240 mg or AAP (abiraterone acetate 1000 mg; prednisone 10 mg). Patients with mCRPC were previously treated with ≥ 2 lines of systemic therapy, including ≥ 1 androgen receptor-axis-targeted therapy for prostate cancer. Results Thirty-three patients were enrolled (niraparib-apalutamide, 6; niraparib-AAP, 27). No dose-limiting toxicities (DLTs) were reported when combinations included niraparib 200 mg; five patients receiving niraparib 300 mg experienced DLTs [niraparib-apalutamide, 2/3 patients (66.7%); niraparib-AAP, 3/8 patients (37.5%)]. Although data are limited, niraparib exposures were lower when given with apalutamide compared with historical niraparib monotherapy exposures in patients with solid tumors. Because of the higher incidence of DLTs, the niraparib–apalutamide combination and niraparib 300 mg combination with AAP were not further evaluated. Niraparib 200 mg was selected as the RP2D with AAP. Of 19 patients receiving niraparib 200 mg with AAP, 12 (63.2%) had grade 3/4 treatment-emergent adverse events, the most common being thrombocytopenia (26.3%) and hypertension (21.1%). Five patients (26.3%) had adverse events leading to treatment discontinuation. Conclusions These results support the choice of niraparib 200 mg as the RP2D with AAP. The niraparib–AAP combination was tolerable in patients with mCRPC, with no new safety signals. An ongoing phase 3 study is further assessing this combination in patients with mCRPC. Trial registration no. NCT02924766 (ClinicalTrials.gov). Supplementary Information The online version contains supplementary material available at 10.1007/s00280-021-04249-7.
Collapse
Affiliation(s)
- Fred Saad
- Centre Hospitalier de l'Université de Montréal, Montréal, Canada.
| | | | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - Julie N Graff
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | | | | | | | - Eugene Zhu
- Janssen Research & Development, Raritan, NJ, USA
| | - Alex Yu
- Janssen Research & Development, Spring House, PA, USA
| | - Anasuya Hazra
- Janssen Research & Development, Spring House, PA, USA
| | | | | | | | | | - Vinny Hayreh
- Janssen Research & Development, Los Angeles, CA, USA
| | | |
Collapse
|
441
|
Caracciolo D, Riillo C, Di Martino MT, Tagliaferri P, Tassone P. Alternative Non-Homologous End-Joining: Error-Prone DNA Repair as Cancer's Achilles' Heel. Cancers (Basel) 2021; 13:cancers13061392. [PMID: 33808562 PMCID: PMC8003480 DOI: 10.3390/cancers13061392] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Cancer onset and progression lead to a high rate of DNA damage, due to replicative and metabolic stress. To survive in this dangerous condition, cancer cells switch the DNA repair machinery from faithful systems to error-prone pathways, strongly increasing the mutational rate that, in turn, supports the disease progression and drug resistance. Although DNA repair de-regulation boosts genomic instability, it represents, at the same time, a critical cancer vulnerability that can be exploited for synthetic lethality-based therapeutic intervention. We here discuss the role of the error-prone DNA repair, named Alternative Non-Homologous End Joining (Alt-NHEJ), as inducer of genomic instability and as a potential therapeutic target. We portray different strategies to drug Alt-NHEJ and discuss future challenges for selecting patients who could benefit from Alt-NHEJ inhibition, with the aim of precision oncology. Abstract Error-prone DNA repair pathways promote genomic instability which leads to the onset of cancer hallmarks by progressive genetic aberrations in tumor cells. The molecular mechanisms which foster this process remain mostly undefined, and breakthrough advancements are eagerly awaited. In this context, the alternative non-homologous end joining (Alt-NHEJ) pathway is considered a leading actor. Indeed, there is experimental evidence that up-regulation of major Alt-NHEJ components, such as LIG3, PolQ, and PARP1, occurs in different tumors, where they are often associated with disease progression and drug resistance. Moreover, the Alt-NHEJ addiction of cancer cells provides a promising target to be exploited by synthetic lethality approaches for the use of DNA damage response (DDR) inhibitors and even as a sensitizer to checkpoint-inhibitors immunotherapy by increasing the mutational load. In this review, we discuss recent findings highlighting the role of Alt-NHEJ as a promoter of genomic instability and, therefore, as new cancer’s Achilles’ heel to be therapeutically exploited in precision oncology.
Collapse
|
442
|
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: 37] [Impact Index Per Article: 12.3] [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
|
443
|
McNevin CS, Baird AM, McDermott R, Finn SP. Diagnostic Strategies for Treatment Selection in Advanced Prostate Cancer. Diagnostics (Basel) 2021; 11:345. [PMID: 33669657 PMCID: PMC7922176 DOI: 10.3390/diagnostics11020345] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/22/2022] Open
Abstract
Prostate Cancer (PCa) is a leading cause of morbidity and mortality among men worldwide. For most men with PCa, their disease will follow an indolent course. However, advanced PCa is associated with poor outcomes. There has been an advent of new therapeutic options with proven efficacy for advanced PCa in the last decade which has improved survival outcomes for men with this disease. Despite this, advanced PCa continues to be associated with a high rate of death. There is a lack of strong evidence guiding the timing and sequence of these novel treatment strategies. This paper focuses on a review of the strategies for diagnostic and the current evidence available for treatment selection in advanced PCa.
Collapse
Affiliation(s)
- Ciara S. McNevin
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland;
- Department of Medical Oncology, St. James Hospital, D08 NHY1 Dublin, Ireland
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D02 A440 Dublin, Ireland;
| | - Ray McDermott
- Department of Medical Oncology, Tallaght University Hospital, D24 NR0A Dublin, Ireland;
- Department of Medical Oncology, St. Vincent’s University Hospital, D04 YN26 Dublin, Ireland
| | - Stephen P. Finn
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland;
- Department of Histopathology, St. James’s Hospital, P.O. Box 580, James’s Street, D08 X4RX Dublin, Ireland
| |
Collapse
|
444
|
Rebello RJ, Oing C, Knudsen KE, Loeb S, Johnson DC, Reiter RE, Gillessen S, Van der Kwast T, Bristow RG. Prostate cancer. Nat Rev Dis Primers 2021. [PMID: 33542230 DOI: 10.1038/s41572-020-0024.3-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2023]
Abstract
Prostate cancer is a complex disease that affects millions of men globally, predominantly in high human development index regions. Patients with localized disease at a low to intermediate risk of recurrence generally have a favourable outcome of 99% overall survival for 10 years if the disease is detected and treated at an early stage. Key genetic alterations include fusions of TMPRSS2 with ETS family genes, amplification of the MYC oncogene, deletion and/or mutation of PTEN and TP53 and, in advanced disease, amplification and/or mutation of the androgen receptor (AR). Prostate cancer is usually diagnosed by prostate biopsy prompted by a blood test to measure prostate-specific antigen levels and/or digital rectal examination. Treatment for localized disease includes active surveillance, radical prostatectomy or ablative radiotherapy as curative approaches. Men whose disease relapses after prostatectomy are treated with salvage radiotherapy and/or androgen deprivation therapy (ADT) for local relapse, or with ADT combined with chemotherapy or novel androgen signalling-targeted agents for systemic relapse. Advanced prostate cancer often progresses despite androgen ablation and is then considered castration-resistant and incurable. Current treatment options include AR-targeted agents, chemotherapy, radionuclides and the poly(ADP-ribose) inhibitor olaparib. Current research aims to improve prostate cancer detection, management and outcomes, including understanding the fundamental biology at all stages of the disease.
Collapse
Affiliation(s)
- Richard J Rebello
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK
| | - Christoph Oing
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK
- Department of Oncology, Haematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Centre Eppendorf, Hamburg, Germany
| | - Karen E Knudsen
- Sidney Kimmel Cancer Center at Jefferson Health and Thomas Jefferson University, Philadelphia, PA, USA
| | - Stacy Loeb
- Department of Urology and Population Health, New York University and Manhattan Veterans Affairs, Manhattan, NY, USA
| | - David C Johnson
- Department of Urology, University of North Carolina, Chapel Hill, NC, USA
| | - Robert E Reiter
- Department of Urology, Jonssen Comprehensive Cancer Center UCLA, Los Angeles, CA, USA
| | | | - Theodorus Van der Kwast
- Laboratory Medicine Program, Princess Margaret Cancer Center, University Health Network, Toronto, Canada
| | - Robert G Bristow
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK.
| |
Collapse
|
445
|
Abstract
Prostate cancer is a complex disease that affects millions of men globally, predominantly in high human development index regions. Patients with localized disease at a low to intermediate risk of recurrence generally have a favourable outcome of 99% overall survival for 10 years if the disease is detected and treated at an early stage. Key genetic alterations include fusions of TMPRSS2 with ETS family genes, amplification of the MYC oncogene, deletion and/or mutation of PTEN and TP53 and, in advanced disease, amplification and/or mutation of the androgen receptor (AR). Prostate cancer is usually diagnosed by prostate biopsy prompted by a blood test to measure prostate-specific antigen levels and/or digital rectal examination. Treatment for localized disease includes active surveillance, radical prostatectomy or ablative radiotherapy as curative approaches. Men whose disease relapses after prostatectomy are treated with salvage radiotherapy and/or androgen deprivation therapy (ADT) for local relapse, or with ADT combined with chemotherapy or novel androgen signalling-targeted agents for systemic relapse. Advanced prostate cancer often progresses despite androgen ablation and is then considered castration-resistant and incurable. Current treatment options include AR-targeted agents, chemotherapy, radionuclides and the poly(ADP-ribose) inhibitor olaparib. Current research aims to improve prostate cancer detection, management and outcomes, including understanding the fundamental biology at all stages of the disease.
Collapse
|
446
|
Schaeffer E, Srinivas S, Antonarakis ES, Armstrong AJ, Bekelman JE, Cheng H, D’Amico AV, Davis BJ, Desai N, Dorff T, Eastham JA, Farrington TA, Gao X, Horwitz EM, Ippolito JE, Kuettel MR, Lang JM, McKay R, McKenney J, Netto G, Penson DF, Pow-Sang JM, Reiter R, Richey S, Roach, III M, Rosenfeld S, Shabsigh A, Spratt DE, Teply BA, Tward J, Shead DA, Freedman-Cass DA. NCCN Guidelines Insights: Prostate Cancer, Version 1.2021. J Natl Compr Canc Netw 2021; 19:134-143. [DOI: 10.6004/jnccn.2021.0008] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The NCCN Guidelines for Prostate Cancer address staging and risk assessment after a prostate cancer diagnosis and include management options for localized, regional, and metastatic disease. Recommendations for disease monitoring and treatment of recurrent disease are also included. The NCCN Prostate Cancer Panel meets annually to reevaluate and update their recommendations based on new clinical data and input from within NCCN Member Institutions and from external entities. This article summarizes the panel’s discussions for the 2021 update of the guidelines with regard to systemic therapy for metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Edward Schaeffer
- 1Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | | | - Heather Cheng
- 6Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | - Anthony Victor D’Amico
- 7Dana-Farber/Brigham and Women’s Cancer Center
- Massachusetts General Hospital Cancer Center
| | | | - Neil Desai
- 9UT Southwestern Simmons Comprehensive Cancer Center
| | | | | | | | - Xin Gao
- 7Dana-Farber/Brigham and Women’s Cancer Center
- Massachusetts General Hospital Cancer Center
| | | | - Joseph E. Ippolito
- 14Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | - Jesse McKenney
- 18Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | | | - Sylvia Richey
- 23St. Jude Children’s Research Hospital/The University of Tennessee Health Science Center
| | | | - Stan Rosenfeld
- 25University of California San Francisco Patient Services Committee Chair
| | - Ahmad Shabsigh
- 26The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | | | | | - Jonathan Tward
- 29Huntsman Cancer Institute at the University of Utah; and
| | | | | |
Collapse
|
447
|
Lozano R, Castro E, Aragón IM, Cendón Y, Cattrini C, López-Casas PP, Olmos D. Genetic aberrations in DNA repair pathways: a cornerstone of precision oncology in prostate cancer. Br J Cancer 2021; 124:552-563. [PMID: 33106584 PMCID: PMC7851123 DOI: 10.1038/s41416-020-01114-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 08/03/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022] Open
Abstract
Over the past years, several studies have demonstrated that defects in DNA damage response and repair (DDR) genes are present in a significant proportion of patients with prostate cancer. These alterations, particularly mutations in BRCA2, are known to be associated with an increased risk of developing prostate cancer and more aggressive forms of the disease. There is growing evidence that certain DDR gene aberrations confer sensitivity to poly-(ADP ribose) polymerase inhibitors and/or platinum chemotherapy, while other defects might identify cases that are more likely to benefit from immune checkpoint inhibition. The potential prognostic impact and relevance for treatment selection together with the decreasing costs and broader accessibility to next-generation sequencing have already resulted in the increased frequency of genetic profiling of prostate tumours. Remarkably, almost half of all DDR genetic defects can occur in the germline, and prostate cancer patients identified as mutation carriers, as well as their families, will require appropriate genetic counselling. In this review, we summarise the current knowledge regarding the biology and clinical implications of DDR defects in prostate cancer, and outline how this evidence is prompting a change in the treatment landscape of the disease.
Collapse
Affiliation(s)
- Rebeca Lozano
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Genitourinary Cancer Translational Research Group, The Institute of Biomedical Research in Málaga (IBIMA), Málaga, Spain
| | - Elena Castro
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Genitourinary Cancer Translational Research Group, The Institute of Biomedical Research in Málaga (IBIMA), Málaga, Spain
- UGCI Oncología Médica, Hospitales Universitarios Virgen de la Victoria y Regional de Málaga, Málaga, Spain
| | - Isabel M Aragón
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Genitourinary Cancer Translational Research Group, The Institute of Biomedical Research in Málaga (IBIMA), Málaga, Spain
| | - Ylenia Cendón
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Carlo Cattrini
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Academic Unit of Medical Oncology, IRCCS San Martino Polyclinic Hospital, Genoa, Italy
| | - Pedro P López-Casas
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Genitourinary Cancer Translational Research Group, The Institute of Biomedical Research in Málaga (IBIMA), Málaga, Spain
| | - David Olmos
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
- Genitourinary Cancer Translational Research Group, The Institute of Biomedical Research in Málaga (IBIMA), Málaga, Spain.
| |
Collapse
|
448
|
Phycocyanin from Arthrospira platensis as Potential Anti-Cancer Drug: Review of In Vitro and In Vivo Studies. Life (Basel) 2021; 11:life11020091. [PMID: 33513794 PMCID: PMC7911896 DOI: 10.3390/life11020091] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/18/2022] Open
Abstract
The application of cytostatic drugs or natural substances to inhibit cancer growth and progression is an important and evolving subject of cancer research. There has been a surge of interest in marine bioresources, particularly algae, as well as cyanobacteria and their bioactive ingredients. Dried biomass products of Arthrospira and Chlorella have been categorized as “generally recognized as safe” (GRAS) by the US Food and Drug Administration (FDA). Of particular importance is an ingredient of Arthrospira: phycocyanin, a blue-red fluorescent, water-soluble and non-toxic biliprotein pigment. It is reported to be the main active ingredient of Arthrospira and was shown to have therapeutic properties, including anti-oxidant, anti-inflammatory, immune-modulatory and anti-cancer activities. In the present review, in vitro and in vivo data on the effects of phycocyanin on various tumor cells and on cells from healthy tissues are summarized. The existing knowledge of underlying molecular mechanisms, and strategies to improve the efficiency of potential phycocyanin-based anti-cancer therapies are discussed.
Collapse
|
449
|
Merseburger AS, Waldron N, Ribal MJ, Heidenreich A, Perner S, Fizazi K, Sternberg CN, Mateo J, Wirth MP, Castro E, Olmos D, Petrylak DP, Chowdhury S. Genomic Testing in Patients with Metastatic Castration-resistant Prostate Cancer: A Pragmatic Guide for Clinicians. Eur Urol 2021; 79:519-529. [PMID: 33494937 DOI: 10.1016/j.eururo.2020.12.039] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/26/2020] [Indexed: 12/19/2022]
Abstract
CONTEXT Genomic testing is becoming increasingly important in patients with advanced prostate cancer (PC) and is being incorporated in clinical practice to guide treatment. OBJECTIVE To review the current understanding of genomic alterations and the status of genomic testing in patients with metastatic castration-resistant PC (mCRPC), and the potential use of genomic tests in clinical practice. EVIDENCE ACQUISITION We reviewed recent publications (past 15 yr) from PubMed, proceedings of scientific conferences, and published guidelines. Reports on mCRPC in the following areas were selected: development, testing, and validation of techniques for identifying genomic alterations; molecular characterization; and trials of genetically targeted therapies. EVIDENCE SYNTHESIS mCRPC tumors harbor molecular alterations that are possible targets for treatment, and a number of therapies are in development to exploit these alterations (eg, PD-1 inhibitors, PARP inhibitors, tyrosine kinase inhibitors). Next-generation sequencing of DNA from tumor tissue can identify somatic alterations that would not be identified by germline testing. Work is ongoing to evaluate the use of less invasive somatic testing methods (eg, sequencing of cell-free circulating tumor DNA). Current international guidelines recommend germline and/or somatic testing for men with advanced and/or high-risk PC regardless of family history to identify those with homologous recombination repair gene mutations or mismatch repair defects/microsatellite instability who may be eligible for treatment with a PARP inhibitor or pembrolizumab, respectively. CONCLUSIONS Genomic testing for mCRPC may provide information on prognostic, predictive, and resistance biomarkers. Although the incorporation of testing into clinical practice remains challenging, routine genomic testing of men with advanced PC is recommended to guide management and treatment decisions. PATIENT SUMMARY Similar to many cancers, prostate cancer is caused by defects in the cancer's DNA, which are called genetic or genomic defects. New treatments targeting these defects are approved for metastatic castration-resistant prostate cancer. Specific new tests are under development to detect these potentially treatable genetic defects.
Collapse
Affiliation(s)
| | | | - Maria J Ribal
- Hospital Clínic, University of Barcelona, Barcelona, Spain
| | | | - Sven Perner
- Institute of Pathology, University Hospital Schleswig Holstein, Campus Lübeck, Lübeck, Germany; Pathology Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Karim Fizazi
- University of Paris Institut Gustave Roussy, Villejuif Cedex, France
| | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York-Presbyterian, New York, NY, USA
| | - Joaquin Mateo
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | - Elena Castro
- Spanish National Cancer Research Centre, Madrid, Spain; University Hospitals Regional and Virgen de la Victoria, Málaga, Spain
| | - David Olmos
- Spanish National Cancer Research Centre, Madrid, Spain; University Hospitals Regional and Virgen de la Victoria, Málaga, Spain
| | | | - Simon Chowdhury
- Guy's Hospital, London, UK; Sarah Cannon Research Institute, London, UK
| |
Collapse
|
450
|
Streamlining Germline Genetic Testing in Prostate Cancer. Eur Urol Oncol 2021; 4:10-11. [PMID: 33483264 DOI: 10.1016/j.euo.2020.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 12/22/2020] [Indexed: 11/23/2022]
|