1
|
Kwan EM, Ng SWS, Tolmeijer SH, Emmett L, Sandhu S, Buteau JP, Iravani A, Joshua AM, Francis RJ, Subhash V, Lee ST, Scott AM, Martin AJ, Stockler MR, Donnellan G, Annala M, Herberts C, Davis ID, Hofman MS, Azad AA, Wyatt AW, TheraP Investigators and the ANZUP Cancer Trials Group. Lutetium-177-PSMA-617 or cabazitaxel in metastatic prostate cancer: circulating tumor DNA analysis of the randomized phase 2 TheraP trial. Nat Med 2025:10.1038/s41591-025-03704-9. [PMID: 40425844 DOI: 10.1038/s41591-025-03704-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 04/07/2025] [Indexed: 05/29/2025]
Abstract
The prostate-specific membrane antigen (PSMA)-targeted radioligand [¹⁷⁷Lu]Lu-PSMA-617 is a new standard treatment for metastatic castration-resistant prostate cancer (mCRPC), but predictive genomic biomarkers informing its rational use are unknown. We performed detailed dissection of prostate cancer driver genes across 290 serial plasma cell-free DNA samples from 180 molecular imaging-selected patients with mCRPC from the randomized TheraP trial of [¹⁷⁷Lu]Lu-PSMA-617 (n = 97) versus cabazitaxel chemotherapy (n = 83). The primary endpoint was PSA50 biochemical response, with secondary endpoints of progression-free survival (PFS) and overall survival (OS). In this post-hoc biomarker analysis, a low pretreatment circulating tumor DNA (ctDNA) fraction predicted a superior biochemical response (100% versus 58%, P = 0.0067) and PFS (median 14.7 versus 6.0 months; hazard ratio 0.12, P = 2.5 × 10-4) on [¹⁷⁷Lu]Lu-PSMA-617 independent of predictive PSMA-positron emission tomography imaging parameters, although this benefit did not extend to OS. Deleterious PTEN alterations were associated with worse PFS and OS on cabazitaxel, whereas ATM defects were observed in select patients with favorable [¹⁷⁷Lu]Lu-PSMA-617 outcomes. Comparing pretreatment and progression ctDNA revealed population flux but no evidence that alterations in individual mCRPC genes (or FOLH1) are dominant causes of acquired [¹⁷⁷Lu]Lu-PSMA-617 or cabazitaxel resistance. Our results nominate new candidate biomarkers for [¹⁷⁷Lu]Lu-PSMA-617 selection and ultimately expand the mCRPC predictive biomarker repertoire. We anticipate our ctDNA fraction-aware analytical framework will aid future precision management strategies for [¹⁷⁷Lu]Lu-PSMA-617 and other PSMA-targeted therapeutics. ClinicalTrials.gov identifier: NCT03392428 .
Collapse
Affiliation(s)
- Edmond M Kwan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia
- Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Sarah W S Ng
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sofie H Tolmeijer
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Shahneen Sandhu
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - James P Buteau
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Amir Iravani
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Division of Nuclear Medicine, Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Anthony M Joshua
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Medical Oncology, Kinghorn Cancer Centre, Sydney, New South Wales, Australia
| | - Roslyn J Francis
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Vinod Subhash
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Sydney, New South Wales, Australia
| | - Sze-Ting Lee
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Andrew M Scott
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Andrew J Martin
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
- Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Martin R Stockler
- Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Gráinne Donnellan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Matti Annala
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Cameron Herberts
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ian D Davis
- Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia.
- Cancer Services, Eastern Health, Melbourne, Victoria, Australia.
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Arun A Azad
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada.
| | | |
Collapse
Collaborators
Tim Akhurst, Ramin Alipour, Dale L Bailey, Patricia Banks, Alexis Beaulieu, Louise Campbell, Wei Chua, Megan Crumbaker, Nattakorn Dhiantravan, Kate Ford, Craig Gedye, Jeffrey C Goh, Alex D Guminski, Anis Hamid, Mohammad B Haskali, Rodney J Hicks, Edward Hsiao, Terry Hung, Ian D Kirkwood, Grace Kong, Ailsa Langford, Nicola Lawrence, Jeremy Lewin, Peter Lin, Michael McCarthy, Margaret M McJannett, William McDonald, Kate Moodie, Declan G Murphy, Siobhan Ng, Andrew Nguyen, David A Pattison, David Pook, Izabella Pokorski, Shakher Ramdave, Nisha Rana, Aravind S Ravi Kumar, Andrew D Redfern, Paul Roach, Peter Roselt, Natalie K Rutherford, Javad Saghebi, Geoffrey Schembri, Lavinia Spain, Shalini Subramaniam, Thean Hsiang Tan, Sue Ping Thang, Paul Thomas, Ben Tran, John A Violet, Roslyn Wallace, Andrew Weickhardt, Scott G Williams, Sonia Yip, Alison Y Zhang,
Collapse
|
2
|
Rajpar S, Ibrahim T, Carmel A, Merabet Z, Vielh P, Foulon S, Lesaunier F, Delva R, Rolland F, Priou F, Ferrero JM, Houédé N, Mourey L, Théodore C, Krakowski I, Faivre L, Habibian M, Culine S, Gravis G, Chauchereau A, Fizazi K. The Benefit of Combining Docetaxel with Androgen Deprivation Therapy in Localized and Metastatic Hormone-sensitive Prostate Cancer is Predicted by ERG Expression: An Analysis of Two GETUG Phase 3 Trials. Eur Urol Oncol 2025; 8:296-305. [PMID: 39034169 DOI: 10.1016/j.euo.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/02/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND AND OBJECTIVE Docetaxel has become a standard component of care for advanced prostate cancer (PC); however, its benefits are not universal among patients. A subset of PC cases exhibit TMPRSS2-ERG gene fusion, resulting in ERG overexpression in tumors. Our aim was to assess biomarkers for docetaxel efficacy in men with hormone-sensitive PC (HSPC). METHODS Pretreatment prostate biopsies were obtained from participants in two randomized phase 3 clinical trials investigating docetaxel in high-risk localized PC (GETUG 12) and metastatic HSPC (GETUG 15). Immunohistochemistry staining for Ki67, PTEN, RB, and phosphorylated RB was conducted for GETUG 12 samples, and ERG staining for GETUG 12 and GETUG 15 samples. We examined biomarker association with outcomes using univariate and multivariable analyses adjusted for other validated prognostic factors. KEY FINDINGS AND LIMITATIONS Among GETUG 12 patients, Ki67 was associated with a worse relapse-free survival (RFS; hazard ratio [HR] 1.72; p = 0.0092). A pooled analysis for the two trials (pinteraction = 0.056) revealed that docetaxel-based chemotherapy improved failure-free survival for patients with ERG-positive cancer (HR 0.58; p = 0.03), but not patients with ERG-negative cancer (HR 1.08; p = 0.72). In the ERG-positive subgroup in GETUG 12 (high-risk localized PC), median RFS was 7.79 yr with androgen deprivation therapy (ADT) alone, and was not reached with ADT + docetaxel. In the ERG-negative subgroup, median progression-free survival (mPFS) was 7.79 yr with ADT alone versus 7.08 yr with ADT + docetaxel. In the ERG-positive subgroup in GETUG 15 (metastatic HSPC), mPFS was 10.7 mo with ADT alone versus 18.8 mo with ADT + docetaxel. In the ERG-negative subgroup, mPFS was 10.6 mo with ADT alone versus 13.2 mo with ADT + docetaxel. CONCLUSIONS AND CLINICAL IMPLICATIONS Ki67 may serve as a prognostic factor in HSPC, while ERG expression appears to predict a response to docetaxel in both high-risk localized and metastatic HSPC. PATIENT SUMMARY We assessed factors that could predict outcomes after docetaxel chemotherapy in patients with advanced prostate cancer. We found that expression of a protein called ERG can predict a good response to docetaxel in these patients.
Collapse
Affiliation(s)
| | - Tony Ibrahim
- INSERM U981, Prostate Cancer Group, Université Paris-Saclay, Gustave Roussy, Villejuif, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Alexandra Carmel
- Biostatistics Department, Gustave Roussy, Paris-Saclay University, Paris, France
| | - Zahira Merabet
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - Philippe Vielh
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France; Medipath and American Hospital of Paris, Paris, France
| | - Stephanie Foulon
- Biostatistics Department, Gustave Roussy, Paris-Saclay University, Paris, France
| | | | - Rémy Delva
- Institut de Cancerologie de l'Ouest, Angers, France
| | - Frederic Rolland
- Department of Medical Oncology, Centre René Gauducheau, Saint-Herblin, France
| | - Frank Priou
- Department of Medical Oncology, Centre Hospitalier La Roche-sur-Yon, La Roche-sur-Yon, France
| | - Jean-Marc Ferrero
- Medical Oncology Department, Centre Antoine Lacassagne, University Côte d'Azur, Nice, France
| | - Nadine Houédé
- Medical Oncology, Institut de Cancérologie du Gard, Montpellier University, Nimes, France
| | | | | | - Ivan Krakowski
- Department of Medical Oncology, Centre Alexis Vautrin, Vandoeuvre Les Nancy, France
| | - Laura Faivre
- Biostatistics Department, Gustave Roussy, Paris-Saclay University, Paris, France
| | | | - Stéphane Culine
- Department of Medical Oncology, Hôpital Saint-Louis, AP-HP, Paris, France
| | | | - Anne Chauchereau
- INSERM U981, Prostate Cancer Group, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Karim Fizazi
- INSERM U981, Prostate Cancer Group, Université Paris-Saclay, Gustave Roussy, Villejuif, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France.
| |
Collapse
|
3
|
De Laere B, Crippa A, Discacciati A, Larsson B, Persson M, Johansson S, D'hondt S, Bergström R, Chellappa V, Mayrhofer M, Banijamali M, Kotsalaynen A, Schelstraete C, Vanwelkenhuyzen JP, Hjälm-Eriksson M, Pettersson L, Ullén A, Lumen N, Enblad G, Thellenberg Karlsson C, Jänes E, Sandzén J, Schatteman P, Nyre Vigmostad M, Olsson M, Ghysel C, Sautois B, De Roock W, Van Bruwaene S, Anden M, Verbiene I, De Maeseneer D, Everaert E, Darras J, Aksnessether BY, Luyten D, Strijbos M, Mortezavi A, Oldenburg J, Ost P, Eklund M, Grönberg H, Lindberg J. Androgen receptor pathway inhibitors and taxanes in metastatic prostate cancer: an outcome-adaptive randomized platform trial. Nat Med 2024; 30:3291-3302. [PMID: 39164518 PMCID: PMC11564108 DOI: 10.1038/s41591-024-03204-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024]
Abstract
ProBio is the first outcome-adaptive platform trial in prostate cancer utilizing a Bayesian framework to evaluate efficacy within predefined biomarker signatures across systemic treatments. Prospective circulating tumor DNA and germline DNA analysis was performed in patients with metastatic castration-resistant prostate cancer before randomization to androgen receptor pathway inhibitors (ARPIs), taxanes or a physician's choice control arm. The primary endpoint was the time to no longer clinically benefitting (NLCB). Secondary endpoints included overall survival and (serious) adverse events. Upon reaching the time to NLCB, patients could be re-randomized. The primary endpoint was met after 218 randomizations. ARPIs demonstrated ~50% longer time to NLCB compared to taxanes (median, 11.1 versus 6.9 months) and the physician's choice arm (median, 11.1 versus 7.4 months) in the biomarker-unselected or 'all' patient population. ARPIs demonstrated longer overall survival (median, 38.7 versus 21.7 and 21.8 months for taxanes and physician's choice, respectively). Biomarker signature findings suggest that the largest increase in time to NLCB was observed in AR (single-nucleotide variant/genomic structural rearrangement)-negative and TP53 wild-type patients and TMPRSS2-ERG fusion-positive patients, whereas no difference between ARPIs and taxanes was observed in TP53-altered patients. In summary, ARPIs outperform taxanes and physician's choice treatment in patients with metastatic castration-resistant prostate cancer with detectable circulating tumor DNA. ClinicalTrials.gov registration: NCT03903835 .
Collapse
Affiliation(s)
- Bram De Laere
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Alessio Crippa
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Discacciati
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Berit Larsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Maria Persson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Johansson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Sanne D'hondt
- Health, Innovation and Research Institute (Clinical Trial Unit), University Hospital Ghent, Ghent, Belgium
| | - Rebecka Bergström
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Venkatesh Chellappa
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Markus Mayrhofer
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Mahsan Banijamali
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Anastasijia Kotsalaynen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Jan Pieter Vanwelkenhuyzen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | | | - Linn Pettersson
- Department of Oncology, Länssjukhuset Ryhov, Jönköping, Sweden
| | - Anders Ullén
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Nicolaas Lumen
- Department of Urology, University Hospital Ghent, Ghent, Belgium
| | - Gunilla Enblad
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | | | - Elin Jänes
- Department of Oncology, Sundsvalls sjukhus, Sundsvall, Sweden
| | - Johan Sandzén
- Department of Oncology, Centralsjukhuset Karlstad, Karlstad, Sweden
| | - Peter Schatteman
- Department of Urology, Onze Lieve Vrouwziekenhuis, Aalst, Belgium
| | | | - Martha Olsson
- Department of Oncology, Centrallasarettet Växjö, Växjö, Sweden
| | | | - Brieuc Sautois
- Department of Oncology, CHU de Liège - site Sart Tilman, Liège, Belgium
| | - Wendy De Roock
- Department of Oncology, Ziekenhuis Oost- Limburg, Genk, Belgium
| | | | - Mats Anden
- Department of Oncology, Länssjukhuset i Kalmar, Kalmar, Sweden
| | | | | | - Els Everaert
- Department of Oncology, Vitaz campus Sint-Niklaas Lodewijk, Sint-Niklaas, Belgium
| | - Jochen Darras
- Department of Urology, AZ Damiaan, Oostende, Belgium
| | | | - Daisy Luyten
- Department of Oncology, Virga Jessa, Hasselt, Belgium
| | | | - Ashkan Mortezavi
- Department of Urology, Universitätsspital Basel, Basel, Switzerland
- Department of Urology, Universitätsspital Zürich, Zürich, Switzerland
| | - Jan Oldenburg
- Department of Oncology, Akershus University Hospital, Nordbyhagen, Norway
| | - Piet Ost
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Department of Radiation Oncology, GZA Sint-Augustinus, Antwerpen, Belgium
| | - Martin Eklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
- Prostatacancer Centrum, Capio S:t Görans Sjukhus, Stockholm, Sweden.
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Zhang D, Weng H, Zhu Z, Gong W, Ma Y. Evaluating first-line therapeutic strategies for metastatic castration-resistant prostate cancer: a comprehensive network meta-analysis and systematic review. Front Oncol 2024; 14:1378993. [PMID: 38686197 PMCID: PMC11056588 DOI: 10.3389/fonc.2024.1378993] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Objective This study aimed to evaluate the relative efficacy and safety of first-line treatment options for metastatic castration-resistant prostate cancer (mCRPC). Methods We systematically searched electronic databases, including PubMed and Web of Science, for studies published from their inception to April 3rd, 2023. Inclusion criteria were: 1) Completed Phase III or IV randomized controlled trials (RCTs) registered on ClinicalTrials.gov; 2) Patients with a confirmed diagnosis of mCRPC who had not previously received chemotherapy or novel endocrine therapies. We conducted a network meta-analysis using R software (version 3.4.0). Network graphs and risk of bias graphs were generated using Stata 14.0 and RevMan 5.4, respectively. The primary outcome was overall survival (OS), and the secondary outcome was the incidence of severe adverse events (SAEs). Results Seven RCTs encompassing 6,641 patients were included. The network meta-analysis revealed that both docetaxel+prednisone (DP) and cabazitaxel+prednisone (CP) significantly improved OS compared to abiraterone. Compared to placebo, DP showed comparable results to both cabazitaxel 20 mg/m^2+prednisone (C20P) and cabazitaxel 25 mg/m^2+prednisone (C25P) in terms of OS. For SAEs, both DP and C20P were superior to C25P, with no statistical difference between C20P and DP. The probability ranking plots indicated that C25P ranked highest for OS, while DP ranked highest for SAEs. Conclusions Based on our network meta-analysis, we recommend cabazitaxel 20 mg/m^2+prednisone (C20P) as the primary choice for first-line management of mCRPC, followed by DP. Enzalutamide and abiraterone are suggested as subsequent options. Radium-223 may be considered for patients presenting with bone metastases. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023443943.
Collapse
Affiliation(s)
- Duojie Zhang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Haimin Weng
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhangji Zhu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Weilun Gong
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yinfeng Ma
- Department of Urology, The Second Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Gupta S, To TM, Graf R, Kadel EE, Reilly N, Albarmawi H. Real-World Overall Survival and Treatment Patterns by PTEN Status in Metastatic Castration-Resistant Prostate Cancer. JCO Precis Oncol 2024; 8:e2300562. [PMID: 38547419 PMCID: PMC10994466 DOI: 10.1200/po.23.00562] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/16/2024] [Accepted: 02/07/2024] [Indexed: 04/02/2024] Open
Abstract
PURPOSE It is estimated that the PTEN tumor suppressor gene is functionally lost in 40%-50% of patients with metastatic castration-resistant prostate cancer (mCRPC). There is limited information on the prognostic significance of PTEN status identified with genomic testing. This real-world cohort study assessed PTEN as a genetic biomarker using data from US-based oncology practices. METHODS This retrospective real-world cohort study used a deidentified US-based metastatic prostate cancer clinicogenomic database linked to longitudinal clinical data derived from electronic health records. Patients were aged 18 years and older and diagnosed with mCRPC between January 1, 2018, and June 30, 2021. Comprehensive genomic profiling (CGP) of tumor specimens was performed using next-generation sequencing. First-line (1L) and second-line (2L) treatment patterns were assessed and stratified by PTEN status. Kaplan-Meier methods and a multivariable Cox model were used to compare the real-world overall survival by PTEN status among patients who received 1L novel hormone therapy or taxanes. RESULTS In patients with mCRPC who underwent CGP, PTEN loss of function (LOF) was associated with decreased survival compared with intact PTEN (hazard ratio, 1.61 [95% CI, 1.07 to 2.42]; P = .024). The results were not influenced by 1L treatment type. 1L treatment patterns were similar between intact PTEN and PTEN LOF subgroups, with abiraterone and enzalutamide being the two most common treatments in both groups. Patients with PTEN LOF were less likely to receive 2L treatments than patients with intact PTEN. CONCLUSION PTEN LOF, identified with genomic testing, was associated with decreased survival and negative prognoses in patients with mCRPC.
Collapse
Affiliation(s)
- Shilpa Gupta
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Tu My To
- Genentech, Inc, South San Francisco, CA
| | - Ryon Graf
- Foundation Medicine, Inc, Cambridge, MA
| | | | | | | |
Collapse
|
6
|
Baston C, Preda A, Iordache A, Olaru V, Surcel C, Sinescu I, Gingu C. How to Integrate Prostate Cancer Biomarkers in Urology Clinical Practice: An Update. Cancers (Basel) 2024; 16:316. [PMID: 38254807 PMCID: PMC10813985 DOI: 10.3390/cancers16020316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/04/2024] [Accepted: 01/07/2024] [Indexed: 01/24/2024] Open
Abstract
Nowadays, the management of prostate cancer has become more and more challenging due to the increasing number of available treatment options, therapeutic agents, and our understanding of its carcinogenesis and disease progression. Moreover, currently available risk stratification systems used to facilitate clinical decision-making have limitations, particularly in providing a personalized and patient-centered management strategy. Although prognosis and prostate cancer-specific survival have improved in recent years, the heterogenous behavior of the disease among patients included in the same risk prognostic group negatively impacts not only our clinical decision-making but also oncological outcomes, irrespective of the treatment strategy. Several biomarkers, along with available tests, have been developed to help clinicians in difficult decision-making scenarios and guide management strategies. In this review article, we focus on the scientific evidence that supports the clinical use of several biomarkers considered by professional urological societies (and included in uro-oncological guidelines) in the diagnosis process and specific difficult management strategies for clinically localized or advanced prostate cancer.
Collapse
Affiliation(s)
- Catalin Baston
- Department of Nephrology, Urology, Immunology and Immunology of Transplant, Dermatology, Allergology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.B.); (V.O.); (C.S.); (I.S.); (C.G.)
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| | - Adrian Preda
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| | - Alexandru Iordache
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| | - Vlad Olaru
- Department of Nephrology, Urology, Immunology and Immunology of Transplant, Dermatology, Allergology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.B.); (V.O.); (C.S.); (I.S.); (C.G.)
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| | - Cristian Surcel
- Department of Nephrology, Urology, Immunology and Immunology of Transplant, Dermatology, Allergology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.B.); (V.O.); (C.S.); (I.S.); (C.G.)
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| | - Ioanel Sinescu
- Department of Nephrology, Urology, Immunology and Immunology of Transplant, Dermatology, Allergology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.B.); (V.O.); (C.S.); (I.S.); (C.G.)
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| | - Constantin Gingu
- Department of Nephrology, Urology, Immunology and Immunology of Transplant, Dermatology, Allergology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.B.); (V.O.); (C.S.); (I.S.); (C.G.)
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| |
Collapse
|
7
|
Zhao JL, Antonarakis ES, Cheng HH, George DJ, Aggarwal R, Riedel E, Sumiyoshi T, Schonhoft JD, Anderson A, Mao N, Haywood S, Decker B, Curley T, Abida W, Feng FY, Knudsen K, Carver B, Lacouture ME, Wyatt AW, Rathkopf D. Phase 1b study of enzalutamide plus CC-115, a dual mTORC1/2 and DNA-PK inhibitor, in men with metastatic castration-resistant prostate cancer (mCRPC). Br J Cancer 2024; 130:53-62. [PMID: 37980367 PMCID: PMC10781677 DOI: 10.1038/s41416-023-02487-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 10/13/2023] [Accepted: 10/30/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND CC-115, a dual mTORC1/2 and DNA-PK inhibitor, has promising antitumour activity when combined with androgen receptor (AR) inhibition in pre-clinical models. METHODS Phase 1b multicentre trial evaluating enzalutamide with escalating doses of CC-115 in AR inhibitor-naive mCRPC patients (n = 41). Primary endpoints were safety and RP2D. Secondary endpoints included PSA response, time-to-PSA progression, and radiographic progression. RESULTS Common adverse effects included rash (31.7% Grades 1-2 (Gr); 31.7% Gr 3), pruritis (43.9% Gr 1-2), diarrhoea (37% Gr 1-2), and hypertension (17% Gr 1-2; 9.8% Gr 3). CC-115 RP2D was 5 mg twice a day. In 40 evaluable patients, 80% achieved ≥50% reduction in PSA (PSA50), and 58% achieved ≥90% reduction in PSA (PSA90) by 12 weeks. Median time-to-PSA progression was 14.7 months and median rPFS was 22.1 months. Stratification by PI3K alterations demonstrated a non-statistically significant trend towards improved PSA50 response (PSA50 of 94% vs. 67%, p = 0.08). Exploratory pre-clinical analysis suggested CC-115 inhibited mTOR pathway strongly, but may be insufficient to inhibit DNA-PK at RP2D. CONCLUSIONS The combination of enzalutamide and CC-115 was well tolerated. A non-statistically significant trend towards improved PSA response was observed in patients harbouring PI3K pathway alterations, suggesting potential predictive biomarkers of response to a PI3K/AKT/mTOR pathway inhibitor. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT02833883.
Collapse
Affiliation(s)
- Jimmy L Zhao
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- University of Minnesota Masonic Cancer Center, Minneapolis, MN, 55455, USA
| | - Emmanuel S Antonarakis
- The Sidney Kimmel Cancer Comprehensive Cancer Center at Johns Hopkins, 401 N. Broadway, Baltimore, MD, 21231, USA
- R&D in Oncology, AstraZeneca, New York, NY, 10016, USA
| | - Heather H Cheng
- University of Washington and Fred Hutch Cancer Research Center, 1144 Eastlake Avenue, Seattle, WA, 98109, USA
| | - Daniel J George
- Duke Cancer Institute, 20 Duke Medicine Circle, Durham, NC, 27710, USA
| | - Rahul Aggarwal
- University of California, San Francisco Helen Diller Family Comprehensive Cancer Center, 1825 4th Street, San Francisco, CA, 94158, USA
| | - Elyn Riedel
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Takayuki Sumiyoshi
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | | | - Amanda Anderson
- Epic Sciences, 9381 Judicial Drive Suite 200, San Diego, CA, 92121, USA
| | - Ninghui Mao
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Samuel Haywood
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Brooke Decker
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Tracy Curley
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Wassim Abida
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Felix Y Feng
- University of California, San Francisco Helen Diller Family Comprehensive Cancer Center, 1825 4th Street, San Francisco, CA, 94158, USA
| | - Karen Knudsen
- Sidney Kimmel Cancer Center, Thomas Jefferson University, 914 Chestnut Street, Philadelphia, PA, 19107, USA
| | - Brett Carver
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Mario E Lacouture
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Dana Rathkopf
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
8
|
Assayag J, Kim C, Chu H, Webster J. The prognostic value of Eastern Cooperative Oncology Group performance status on overall survival among patients with metastatic prostate cancer: a systematic review and meta-analysis. Front Oncol 2023; 13:1194718. [PMID: 38162494 PMCID: PMC10757350 DOI: 10.3389/fonc.2023.1194718] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 11/15/2023] [Indexed: 01/03/2024] Open
Abstract
Background There is heterogeneity in the literature regarding the strength of association between Eastern Cooperative Oncology Group performance status (ECOG PS) and mortality. We conducted a systematic review and meta-analysis of studies reporting the prognostic value of ECOG PS on overall survival (OS) in metastatic prostate cancer (mPC). Methods PubMed was searched from inception to March 21, 2022. A meta-analysis pooling the effect of ECOG PS categories (≥2 vs. <2, 2 vs. <2, and ≥1 vs. <1) on OS was performed separately for studies including patients with metastatic castration-resistant prostate cancer (mCRPC) and metastatic castration-sensitive prostate cancer (mCSPC) using a random-effects model. Analyses were stratified by prior chemotherapy and study type. Results Overall, 75 studies, comprising 32,298 patients, were included. Most studies (72/75) included patients with mCRPC. Higher ECOG PS was associated with a significant increase in mortality risk, with the highest estimate observed among patients with mCRPC with an ECOG PS of ≥2 versus <2 (hazard ratio [HR]: 2.10, 95% confidence interval [CI]: 1.87-2.37). When stratifying by study type, there was a higher risk estimate of mortality among patients with mCRPC with an ECOG PS of ≥1 versus <1 in real-world data studies (HR: 1.98, 95% CI: 1.72-2.26) compared with clinical trials (HR: 1.32, 95% CI: 1.13-1.54; p < 0.001). There were no significant differences in the HR of OS stratified by previous chemotherapy. Conclusion ECOG PS was a significant predictor of OS regardless of category, previous chemotherapy, and mPC population. Additional studies are needed to better characterize the effect of ECOG PS on OS in mCSPC.
Collapse
Affiliation(s)
- Jonathan Assayag
- Evidence Generation Platform, Pfizer Inc., New York, NY, United States
| | - Chai Kim
- Evidence Generation Platform, Pfizer Inc., New York, NY, United States
| | - Haitao Chu
- Statistical Research and Data Science Center, Global Biometrics and Data Management, Pfizer Inc., New York, NY, United States
| | - Jennifer Webster
- Evidence Generation Platform, Pfizer Inc., New York, NY, United States
| |
Collapse
|
9
|
Al-Toubat M, Serrano S, Elshafei A, Koul K, Feibus AH, Balaji KC. Metastatic prostate cancer is associated with distinct higher frequency of genetic mutations at diagnosis. Urol Oncol 2023; 41:455.e7-455.e15. [PMID: 37838503 DOI: 10.1016/j.urolonc.2023.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/16/2023]
Abstract
INTRODUCTION AND OBJECTIVES We explored characteristic genetic mutations associated with metastatic prostate cancer (PCa) by comparing next generation sequencing (NGS) data between men with or without metastatic disease at diagnosis. METHODS We queried the American Association for Cancer Research Project Genomics Evidence Neoplasia Information Exchange (GENIE) registry for men diagnosed with PCa. Patients were categorized into with (M1) or without metastatic disease (M0) groups. The difference in the frequency of genetic mutations between the two groups and the prognostic significance of the mutations were analyzed using SPSS V28. We included frequency rate of > 5% and P values < 0.05 were considered statistically significant to maintain over 95% true positive detection rate. RESULTS Of a total of 10,580 patients with diagnosis of PCa in the dataset, we selected a study cohort of 1268 patients without missing data; 700 (55.2%) had nonmetastatic PCa, 421 (33.2%) and 147 (11.6%) patients had metastatic castration sensitive and resistant PCa respectively. The median age at diagnosis and serum prostate specific antigen (PSA) level for the entire cohort was 62.8 years (IQR 56.3-68.4) and 8.0 ng/ml (IQR 4.9-20.9) respectively. A vast majority of the cohort were of Caucasian ancestry (89.1%). Of a total of 561 genes sequenced, there were mutations in 79 genes (14.1%). The mutation frequency was significantly higher in M1PCa compared to M0PCa, 35.7% and 23.3%, respectively (P = <0.001). The median tumor mutational burden was also significantly higher in the samples from M1PCa (2.59 mut/MB) compared to M0PCa (1.96 mut/MB) (P < 0.001). Compared to M0PCa patients, M1PCa patients demonstrated significantly higher rate of genetic mutations; TP53 (38.73% vs. 17.71% P < 0.001), PTEN (25.70% vs. 11.71% P < 0.001), AR (17.25% vs. 1.43% P < 0.001), APC (11.8% vs. 4.43% P < 0.001), TMPRSS2 (31.5% vs. 11.14% P < 0.001), ERG (23.59% vs. 13.13% P < 0.001), FOXA1 (17.43% vs. 6.33% P < 0.001), MYC (8.45% vs. 2.29% P < 0.001), RB1 (10.39% vs. 2.43% P < 0.001) and CDK12 (8.45% vs. 1.31% P < 0.001). Of the various cellular signaling pathways, the androgen receptor signaling pathway was most often impacted. In the cohort with M1 disease, compared to men without genetic mutations the men with genetic mutations demonstrated worse survival (P = <0.001, log rank test). Compared to castration sensitive M1 patients, AR (57% vs. 4% P < 0.001), TP53 (50.7% vs. 34% P < 0.001), PTEN (35.2% vs. 22.1% P < 0.001), RB1(23.9% vs. 4.75% P < 0.001) were significantly more frequently mutated in castration resistant M1 patients. In contrast, mutations of SPOP (13.3% vs. 7.9% P < 0.001), FOXA1 (17.6% vs. 5.3% P < 0.001) and CDK12 (12% vs. 6.45% P < 0.001) were significantly more frequently found in castration sensitive M1 patients compared to castration resistant patients. CONCLUSION Patients with M1PCa demonstrated characteristic genetic mutations compared to M0PCa, which most often influenced androgen receptor signaling and is associated with worse survival. In addition, we identified distinct genetic mutations between castration sensitive and resistant M1PCa. These findings may be used to further our understanding and management of men with PCa.
Collapse
Affiliation(s)
- Mohammed Al-Toubat
- Department of Urology, University of Florida College of Medicine, Jacksonville, FL
| | - Samuel Serrano
- Department of Urology, University of Florida College of Medicine, Jacksonville, FL
| | - Ahmed Elshafei
- Department of Urology, University of Florida College of Medicine, Jacksonville, FL
| | - Kashyap Koul
- Department of Urology, University of Florida College of Medicine, Jacksonville, FL
| | - Allison H Feibus
- Department of Urology, University of Florida College of Medicine, Jacksonville, FL
| | - K C Balaji
- Department of Urology, University of Florida College of Medicine, Jacksonville, FL.
| |
Collapse
|
10
|
Davoudi F, Moradi A, Becker TM, Lock JG, Abbey B, Fontanarosa D, Haworth A, Clements J, Ecker RC, Batra J. Genomic and Phenotypic Biomarkers for Precision Medicine Guidance in Advanced Prostate Cancer. Curr Treat Options Oncol 2023; 24:1451-1471. [PMID: 37561382 PMCID: PMC10547634 DOI: 10.1007/s11864-023-01121-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 08/11/2023]
Abstract
OPINION STATEMENT Prostate cancer (PCa) is the second most diagnosed malignant neoplasm and is one of the leading causes of cancer-related death in men worldwide. Despite significant advances in screening and treatment of PCa, given the heterogeneity of this disease, optimal personalized therapeutic strategies remain limited. However, emerging predictive and prognostic biomarkers based on individual patient profiles in combination with computer-assisted diagnostics have the potential to guide precision medicine, where patients may benefit from therapeutic approaches optimally suited to their disease. Also, the integration of genotypic and phenotypic diagnostic methods is supporting better informed treatment decisions. Focusing on advanced PCa, this review discusses polygenic risk scores for screening of PCa and common genomic aberrations in androgen receptor (AR), PTEN-PI3K-AKT, and DNA damage response (DDR) pathways, considering clinical implications for diagnosis, prognosis, and treatment prediction. Furthermore, we evaluate liquid biopsy, protein biomarkers such as serum testosterone levels, SLFN11 expression, total alkaline phosphatase (tALP), neutrophil-to-lymphocyte ratio (NLR), tissue biopsy, and advanced imaging tools, summarizing current phenotypic biomarkers and envisaging more effective utilization of diagnostic and prognostic biomarkers in advanced PCa. We conclude that prognostic and treatment predictive biomarker discovery can improve the management of patients, especially in metastatic stages of advanced PCa. This will result in decreased mortality and enhanced quality of life and help design a personalized treatment regimen.
Collapse
Affiliation(s)
- Fatemeh Davoudi
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, 4059 Australia
- Department of Medical Genetics, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Afshin Moradi
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, 4059 Australia
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, 4059 Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, 4102 Australia
| | - Therese M. Becker
- Ingham Institute for Applied Medical Research, University of Western Sydney and University of New South Wales, Liverpool, 2170 Australia
| | - John G. Lock
- Ingham Institute for Applied Medical Research, University of Western Sydney and University of New South Wales, Liverpool, 2170 Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, 2052 Australia
| | - Brian Abbey
- Department of Mathematical and Physical Sciences, School of Computing Engineering and Mathematical Sciences, La Trobe Institute for Molecular Sciences, La Trobe University, Bundoora, VIC Australia
| | - Davide Fontanarosa
- School of Clinical Sciences, Queensland University of Technology, Gardens Point Campus, 2 George St, Brisbane, QLD 4000 Australia
- Centre for Biomedical Technologies (CBT), Queensland University of Technology, Brisbane, QLD 4000 Australia
| | - Annette Haworth
- Institute of Medical Physics, School of Physics, University of Sydney, Camperdown, NSW 2006 Australia
| | - Judith Clements
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, 4059 Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, 4102 Australia
| | - Rupert C. Ecker
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, 4059 Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, 4102 Australia
- TissueGnostics GmbH, EU 1020 Vienna, Austria
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, 4059 Australia
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, 4059 Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, 4102 Australia
| |
Collapse
|
11
|
Nigon E, Lefeuvre-Plesse C, Martinez A, Chauleur C, Lortholary A, Favier L, Bats AS, Guille A, AdélaÏde J, Finetti P, de Casteljac V, Provansal M, Mamessier E, Bertucci F, Ray-Coquard I, Sabatier R. Clinical, pathological, and comprehensive molecular analysis of the uterine clear cell carcinoma: a retrospective national study from TMRG and GINECO network. J Transl Med 2023; 21:408. [PMID: 37353806 PMCID: PMC10288685 DOI: 10.1186/s12967-023-04264-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/09/2023] [Indexed: 06/25/2023] Open
Abstract
BACKGROUND Uterine clear cell carcinomas (CCC) represent less than 5% of uterine cancers. Their biological characteristics and clinical management remain uncertain. A multicenter study to explore both clinical and molecular features of these rare tumors was conducted. METHODS This multicenter retrospective national study was performed within the French TMRG (Rare Gynecologic Malignant Tumors) network. Clinical data and, when available, FFPE blocks were collected. Clinical features, treatments, and outcome (progression-free survival (PFS) and overall survival (OS)) were analyzed and correlated to the protein (tissue micro-array), RNA (Nanostring nCounter® technology), and DNA (array-Comparative Genomic hybridization and target-next generation sequencing) levels using the tumor samples available. RESULTS Sixty-eight patients with uterine CCC were enrolled, 61 from endometrial localization and 5 with cervix localization. Median age at diagnosis was 68.9 years old (range 19-89.7). Most tumors were diagnosed at an early stage (78% FIGO stage I-II). Hysterectomy (performed in 90%) and lymph node dissection (80%) were the most frequent surgical treatment. More than 70% of patients received external beam radiotherapy and 57% received brachytherapy. Nearly half (46%) of the patients received chemotherapy. After a median follow-up of 24.7 months, median PFS was 64.8 months (95 CI [5.3-124.4]) and median OS was 79.7 (IC95 [31.0-128.4]). Low hormone receptor expression (13% estrogen-receptor positive), frequent PI3K pathway alterations (58% PTEN loss, 50% PIK3CA mutations), and P53 abnormalities (41%) were observed. Mismatch repair deficiency was identified in 20%. P16 expression was associated with shorter PFS (HR = 5.88, 95 CI [1.56-25], p = 0.009). Transcriptomic analyzes revealed a specific transcriptomic profile notably with a high expression of immune response-associated genes in uterine CCC displaying a very good overall prognosis. CONCLUSIONS Uterine CCC reported to be potentially MSI high, hormone receptors negative, and sometimes TP53 mutated. However, some patients with immune response-associated features and better prognosis may be candidate to treatment de-escalation and immunotherapy.
Collapse
Affiliation(s)
- Elsa Nigon
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, 232 Boulevard Sainte Marguerite, Marseille, France
| | | | - Alejandra Martinez
- Department of Surgical Oncology, Institut Universitaire du Cancer Toulouse Oncopole, Toulouse, France
| | - Céline Chauleur
- Department of Medical Oncology, Institut de Cancérologie de la Loire, Saint Etienne, France
| | - Alain Lortholary
- Hôpital privé du Confluent, Institut de Cancérologie Catherine de Sienne, Nantes, France
| | - Laure Favier
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Anne-Sophie Bats
- Department of Surgical Oncology, Hôpital Européen Georges Pompidou, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Arnaud Guille
- CRCM, Predictive Oncology laboratory, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France
| | - José AdélaÏde
- CRCM, Predictive Oncology laboratory, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France
| | - Pascal Finetti
- CRCM, Predictive Oncology laboratory, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France
| | - Victoire de Casteljac
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, 232 Boulevard Sainte Marguerite, Marseille, France
| | - Magali Provansal
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, 232 Boulevard Sainte Marguerite, Marseille, France
| | - Emilie Mamessier
- CRCM, Predictive Oncology laboratory, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France
| | - François Bertucci
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, 232 Boulevard Sainte Marguerite, Marseille, France
- CRCM, Predictive Oncology laboratory, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France
| | - Isabelle Ray-Coquard
- Department of Medical Oncology, Centre Léon Bérard, University Claude Bernard Lyon 1, Lyon, France
| | - Renaud Sabatier
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, 232 Boulevard Sainte Marguerite, Marseille, France.
- CRCM, Predictive Oncology laboratory, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France.
| |
Collapse
|
12
|
Kamisawa K, Kosaka T, Nakamura K, Yasumizu Y, Hongo H, Takeda T, Matsumoto K, Nishihara H, Oya M. Influence of response to prior docetaxel on sensitivity to cabazitaxel in prostate cancer patients with PTEN alterations. Cancer Sci 2022; 113:3161-3168. [PMID: 35754315 PMCID: PMC9459256 DOI: 10.1111/cas.15473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/31/2022] [Accepted: 06/15/2022] [Indexed: 12/02/2022] Open
Abstract
The purpose of this study was to investigate factors predicting the sensitivity to cabazitaxel therapy in metastatic castration‐resistant prostate cancer (mCRPC) patients with phosphatase and tensin homolog deleted from chromosome 10 (PTEN) alterations. This single‐institution, retrospective study included 12 mCRPC patients with PTEN alterations who had received cabazitaxel therapy. Five patients (41%) responded to cabazitaxel therapy with a prostate‐specific antigen (PSA) level decline of ≥30% from baseline, and all of them had responded to prior docetaxel therapy with a PSA decline of ≥30%. None of the patients with a poor response to prior docetaxel therapy responded well to cabazitaxel therapy. Of the seven patients who did not respond to cabazitaxel and whose PSA declined from baseline was <30%, five (71%) were also refractory to prior docetaxel therapy. The PSA responses to docetaxel and cabazitaxel were significantly correlated (p = 0.027). Kaplan–Meier analysis revealed that progression‐free survival (PFS) for cabazitaxel was significantly shorter for prior docetaxel nonresponders (3.3 versus 9.1 months, p = 0.028). Multivariate analysis revealed that a poor response to prior docetaxel (PSA decline < 30%) (hazard ratio [HR] = 6.382, 95% confidence interval [CI] 1.172–34.750, p = 0.032) and baseline PSA of ≥20 ng/ml (HR = 33.584, 95% CI 2.332–483.671, p = 0.010) were independent prognostic factors for PFS with cabazitaxel therapy. These results demonstrate cross‐resistance between docetaxel and cabazitaxel. The response to prior docetaxel therapy can influence the sensitivity to cabazitaxel therapy in mCRPC patients with PTEN alterations.
Collapse
Affiliation(s)
- Ken Kamisawa
- Department of Urology, Keio University School of Medicine
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine
| | - Kohei Nakamura
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine
| | - Yota Yasumizu
- Department of Urology, Keio University School of Medicine
| | - Hiroshi Hongo
- Department of Urology, Keio University School of Medicine
| | | | | | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine
| |
Collapse
|
13
|
Velez MG, Kosiorek HE, Egan JB, McNatty AL, Riaz IB, Hwang SR, Stewart GA, Ho TH, Moore CN, Singh P, Sharpsten RK, Costello BA, Bryce AH. Differential impact of tumor suppressor gene (TP53, PTEN, RB1) alterations and treatment outcomes in metastatic, hormone-sensitive prostate cancer. Prostate Cancer Prostatic Dis 2022; 25:479-483. [PMID: 34294873 PMCID: PMC9385473 DOI: 10.1038/s41391-021-00430-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/25/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Altered tumor suppressor genes (TSG-alt) in prostate cancer are associated with worse outcomes. The prognostic value of TSG-alt in metastatic, hormone-sensitive prostate cancer (M1-HSPC) is unknown. We evaluated the effects of TSG-alt on outcomes in M1-HSPC and their prognostic impact by first-line treatment. METHODS We retrospectively identified patients with M1-HSPC at our institution treated with first-line androgen deprivation therapy plus docetaxel (ADT + D) or abiraterone acetate (ADT + A). TSG-alt was defined as any alteration in one or more TSG. The main outcomes were Kaplan-Meier-estimated progression-free survival (PFS) and overall survival, analyzed with the log-rank test. Clinical characteristics were compared with the χ2 test and Kruskal-Wallis rank sum test. Cox regression was used for univariate and multivariable analyses. RESULTS We identified 97 patients with M1-HSPC: 48 (49%) with ADT + A and 49 (51%) with ADT + D. Of 96 patients with data available, 33 (34%) had 1 TSG-alt, 16 (17%) had 2 TSG-alt, and 2 (2%) had 3 TSG-alt. The most common alterations were in TP53 (36%) and PTEN (31%); 6% had RB1 alterations. Median PFS was 13.1 (95% CI, 10.3-26.0) months for patients with normal TSGs (TSG-normal) vs. 7.8 (95% CI, 5.8-10.5) months for TSG-alt (P = 0.005). Median PFS was lower for patients with TSG-alt vs TSG-normal for those with ADT + A (TSG-alt: 8.0 [95% CI, 5.8-13.8] months vs. TSG-normal: 23.2 [95% CI, 13.1-not estimated] months), but not with ADT + D (TSG-alt: 7.8 [95% CI, 5.7-12.9] months vs. TSG-normal: 9.5 [95% CI, 4.8-24.7] months). On multivariable analysis, only TSG-alt predicted worse PFS (hazard ratio, 2.37; 95% CI, 1.42-3.96; P < 0.001). CONCLUSIONS The presence of TSG-alt outperforms clinical criteria for predicting early progression during first-line treatment of M1-HSPC. ADT + A was less effective in patients with than without TSG-alt. Confirmation of these findings may establish the need for inclusion of molecular stratification in treatment algorithms.
Collapse
Affiliation(s)
| | | | - Jan B. Egan
- grid.470142.40000 0004 0443 9766Center for Individualized Medicine, Mayo Clinic, Phoenix, AZ USA
| | | | - Irbaz B. Riaz
- Division of Hematology and Medical Oncology, Phoenix, AZ USA
| | | | | | - Thai H. Ho
- Division of Hematology and Medical Oncology, Phoenix, AZ USA
| | | | - Parminder Singh
- Division of Hematology and Medical Oncology, Phoenix, AZ USA
| | | | - Brian A. Costello
- grid.66875.3a0000 0004 0459 167XDivision of Medical Oncology, Mayo Clinic, Rochester, MN USA
| | - Alan H. Bryce
- Division of Hematology and Medical Oncology, Phoenix, AZ USA
| |
Collapse
|
14
|
Giunta EF, Annaratone L, Bollito E, Porpiglia F, Cereda M, Banna GL, Mosca A, Marchiò C, Rescigno P. Molecular Characterization of Prostate Cancers in the Precision Medicine Era. Cancers (Basel) 2021; 13:4771. [PMID: 34638258 PMCID: PMC8507555 DOI: 10.3390/cancers13194771] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer (PCa) therapy has been recently revolutionized by the approval of new therapeutic agents in the metastatic setting. However, the optimal therapeutic strategy in such patients should be individualized in the light of prognostic and predictive molecular factors, which have been recently studied: androgen receptor (AR) alterations, PTEN-PI3K-AKT pathway deregulation, homologous recombination deficiency (HRD), mismatch repair deficiency (MMRd), and tumor microenvironment (TME) modifications. In this review, we highlighted the clinical impact of prognostic and predictive molecular factors in PCa patients' outcomes, identifying biologically distinct subtypes. We further analyzed the relevant methods to detect these factors, both on tissue, i.e., immunohistochemistry (IHC) and molecular tests, and blood, i.e., analysis of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). Moreover, we discussed the main pros and cons of such techniques, depicting their present and future roles in PCa management, throughout the precision medicine era.
Collapse
Affiliation(s)
- Emilio Francesco Giunta
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80131 Naples, Italy;
| | - Laura Annaratone
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.A.); (C.M.)
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy
| | - Enrico Bollito
- Department of Pathology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, 10043 Turin, Italy;
| | - Francesco Porpiglia
- Department of Urology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, 10043 Turin, Italy;
| | - Matteo Cereda
- Cancer Genomics and Bioinformatics Unit, IIGM-Italian Institute for Genomic Medicine, c/o IRCCS Candiolo, 10060 Turin, Italy;
- Candiolo Cancer Institute, FPO—IRCCS, Str. Prov.le 142, km 3.95, 10060 Candiolo, Italy
| | - Giuseppe Luigi Banna
- Department of Oncology, Portsmouth Hospitals University NHS Trust, Portsmouth PO2 8QD, UK;
| | - Alessandra Mosca
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy;
| | - Caterina Marchiò
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.A.); (C.M.)
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy
| | - Pasquale Rescigno
- Interdisciplinary Group for Translational Research and Clinical Trials, Urological Cancers (GIRT-Uro), Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy
| |
Collapse
|
15
|
Vlajnic T, Brunner P, Eppenberger-Castori S, Rentsch CA, Zellweger T, Bubendorf L. High Inter- and Intratumoral Variability of Ki67 Labeling Index in Newly Diagnosed Prostate Cancer with High Gleason Scores. Pathobiology 2021; 89:74-80. [PMID: 34555829 DOI: 10.1159/000519007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 07/23/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The majority of studies investigating the role of Ki67 labeling index (LI) in prostate carcinoma (PC) focused on localized PC treated radically, where Ki67 LI is regarded as a prognostic marker. The relevance of Ki67 in advanced PC remains largely unexplored. While Gleason score is still one of the best indicators of clinical outcomes in PC, differences in progression-free survival and overall survival in patients with high Gleason scores suggest that additional factors are involved in tumor progression. Understanding the underlying mechanisms could help to optimize treatment strategies for an individual patient. Here, we aimed to determine the inter- and intratumoral distribution of Ki67 LI in patients with PC with high Gleason scores and to correlate Ki67 LI with the status of ERG, PTEN, and Bcl-2. METHODS Immunohistochemistry for Ki67, ERG, PTEN, and Bcl-2 was performed on core needle biopsies from 112 patients with newly diagnosed PC Gleason score 8, 9, and 10. RESULTS Using a cutoff of ≥10%, 17/112 cases (15%) had a homogeneously low and 95/112 cases (85%) a high Ki67 LI. 41% of cases showed intratumoral heterogeneity containing areas with low and high proliferation. There was no association between Ki67 LI and ERG, PTEN, or Bcl-2 status. CONCLUSIONS Our data demonstrate major inter- and intratumoral variability of Ki67 LI in high-grade PC with a surprisingly low Ki67 LI in a subset of cases. Further studies are necessary to explore the molecular basis and potential clinical implications of a paradoxically low proliferation rate in high-grade PC.
Collapse
Affiliation(s)
- Tatjana Vlajnic
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Patrik Brunner
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Serenella Eppenberger-Castori
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Cyrill A Rentsch
- Department of Urology, University Hospital Basel, Basel, Switzerland
| | | | - Lukas Bubendorf
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
16
|
Sweeney C, Bracarda S, Sternberg CN, Chi KN, Olmos D, Sandhu S, Massard C, Matsubara N, Alekseev B, Parnis F, Atduev V, Buchschacher GL, Gafanov R, Corrales L, Borre M, Stroyakovskiy D, Alves GV, Bournakis E, Puente J, Harle-Yge ML, Gallo J, Chen G, Hanover J, Wongchenko MJ, Garcia J, de Bono JS. Ipatasertib plus abiraterone and prednisolone in metastatic castration-resistant prostate cancer (IPATential150): a multicentre, randomised, double-blind, phase 3 trial. Lancet 2021; 398:131-142. [PMID: 34246347 DOI: 10.1016/s0140-6736(21)00580-8] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/25/2021] [Accepted: 03/02/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND The PI3K/AKT and androgen-receptor pathways are dysregulated in metastatic castration-resistant prostate cancers (mCRPCs); tumours with functional PTEN-loss status have hyperactivated AKT signalling. Dual pathway inhibition with AKT inhibitor ipatasertib plus abiraterone might have greater benefit than abiraterone alone. We aimed to compare ipatasertib plus abiraterone with placebo plus abiraterone in patients with previously untreated mCRPC with or without tumour PTEN loss. METHODS We did a randomised, double-blind, phase 3 trial at 200 sites across 26 countries or regions. Patients aged 18 years or older with previously untreated asymptomatic or mildly symptomatic mCRPC who had progressive disease and Eastern Collaborative Oncology Group performance status of 0 or 1 were randomly assigned (1:1; permuted block method) to receive ipatasertib (400 mg once daily orally) plus abiraterone (1000 mg once daily orally) and prednisolone (5 mg twice a day orally) or placebo plus abiraterone and prednisolone (with the same dosing schedule). Patients received study treatment until disease progression, intolerable toxicity, withdrawal from the study, or study completion. Stratification factors were previous taxane-based therapy for hormone-sensitive prostate cancer, type of progression, presence of visceral metastasis, and tumour PTEN-loss status by immunohistochemistry. Patients, investigators, and the study sponsor were masked to the treatment allocation. The coprimary endpoints were investigator-assessed radiographical progression-free survival in the PTEN-loss-by-immunohistochemistry population and in the intention-to-treat population. This study is ongoing and is registered with ClinicalTrials.gov, NCT03072238. FINDINGS Between June 30, 2017, and Jan 17, 2019, 1611 patients were screened for eligibility and 1101 (68%) were enrolled; 554 (50%) were assigned to the placebo-abiraterone group and 547 (50%) to the ipatasertib-abiraterone group. At data cutoff (March 16, 2020), median follow-up duration was 19 months (range 0-33). In the 521 (47%) patients who had tumours with PTEN loss by immunohistochemistry (261 in the placebo-abiraterone group and 260 in the ipatasertib-abiraterone group), median radiographical progression-free survival was 16·5 months (95% CI 13·9-17·0) in the placebo-abiraterone group and 18·5 months (16·3-22·1) in the ipatasertib-abiraterone group (hazard ratio [HR] 0·77 [95% CI 0·61-0·98]; p=0·034; significant at α=0·04). In the intention-to-treat population, median progression-free survival was 16·6 months (95% CI 15·6-19·1) in the placebo-abiraterone group and 19·2 months (16·5-22·3) in the ipatasertib-abiraterone group (HR 0·84 [95% CI 0·71-0·99]; p=0·043; not significant at α=0·01). Grade 3 or higher adverse events occurred in 213 (39%) of 546 patients in the placebo-abiraterone group and in 386 (70%) of 551 patients in the ipatasertib-abiraterone group; adverse events leading to discontinuation of placebo or ipatasertib occurred in 28 (5%) in the placebo-abiraterone group and 116 (21%) in the ipatasertib-abiraterone group. Deaths due to adverse events deemed related to treatment occurred in two patients (<1%; acute myocardial infarction [n=1] and lower respiratory tract infection [n=1]) in the placebo-abiraterone group and in two patients (<1%; hyperglycaemia [n=1] and chemical pneumonitis [n=1]) in the ipastasertb-abiraterone group. INTERPRETATION Ipatasertib plus abiraterone significantly improved radiographical progression-free survival compared with placebo plus abiraterone among patients with mCRPC with PTEN-loss tumours, but there was no significant difference between the groups in the intention-to-treat population. Adverse events were consistent with the known safety profiles of each agent. These data suggest that combined AKT and androgen-receptor signalling pathway inhibition with ipatasertib and abiraterone is a potential treatment for men with PTEN-loss mCRPC, a population with a poor prognosis. FUNDING F Hoffmann-La Roche and Genentech.
Collapse
Affiliation(s)
| | | | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York-Presbyterian, New York City, NY, USA
| | | | - David Olmos
- Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Shahneen Sandhu
- Peter MacCallum Cancer Centre and University of Melbourne, Melbourne, VIC, Australia
| | | | | | - Boris Alekseev
- P A Herzen Moscow Oncology Research Institute, Moscow, Russia
| | - Francis Parnis
- Ashford Cancer Centre Research, Kurralta Park, SA, Australia
| | - Vagif Atduev
- Volga District Medical Centre under Federal Medical and Biological Agency of Russia, Nizhny Novgorod, Russia
| | - Gary L Buchschacher
- Kaiser Permanente Southern California, Los Angeles Medical Center, Los Angeles, CA, USA
| | - Rustem Gafanov
- Russian Scientific Center of Roentgenology and Radiology, Moscow, Russia
| | - Luis Corrales
- Medical Oncology, Centro de Investigación y Manejo del Cáncer CIMCA, San José, Costa Rica
| | | | - Daniil Stroyakovskiy
- Moscow City Oncology Hospital Number 62 of Moscow Healthcare Department, Moscow Oblast, Russia
| | - Gustavo Vasconcelos Alves
- Centro Integrado de Pesquisa em Oncologia, Hospital Nossa Senhora da Conceição, Porto Alegre, Brazil
| | - Evangelos Bournakis
- Oncologic Clinical Trials and Research Clinic, Metropolitan General Hospital, Athens, Greece
| | - Javier Puente
- Medical Oncology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), CIBERONC, Madrid, Spain
| | | | | | - Geng Chen
- Genentech, South San Francisco, CA, USA
| | | | | | | | - Johann S de Bono
- Division of Clinical Studies, The Institute of Cancer Research, London, UK; Drug Development Unit, Royal Marsden Hospital, Sutton, UK.
| |
Collapse
|
17
|
Suzuki H, Castellano D, de Bono J, Sternberg CN, Fizazi K, Tombal B, Wülfing C, Foster MC, Ozatilgan A, Geffriaud-Ricouard C, de Wit R. Cabazitaxel versus abiraterone or enzalutamide in metastatic castration-resistant prostate cancer: post hoc analysis of the CARD study excluding chemohormonal therapy for castrate-naive disease. Jpn J Clin Oncol 2021; 51:1287-1297. [PMID: 33738495 PMCID: PMC8521736 DOI: 10.1093/jjco/hyab028] [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: 01/14/2021] [Accepted: 02/16/2021] [Indexed: 11/12/2022] Open
Abstract
Background In the CARD study (NCT02485691), cabazitaxel significantly improved clinical outcomes versus abiraterone or enzalutamide in patients with metastatic castration-resistant prostate cancer previously treated with docetaxel and the alternative androgen-signalling-targeted inhibitor. However, some patients received docetaxel or the prior alternative androgen-signalling-targeted inhibitor in the metastatic hormone-sensitive (mHSPC) setting. Therefore, the CARD results cannot be directly translated to a Japanese population. Methods Patients (N = 255) received cabazitaxel (25 mg/m2 IV Q3W, prednisone, G-CSF) versus abiraterone (1000 mg PO, prednisone) or enzalutamide (160 mg PO) after prior docetaxel and progression ≤12 months on the alternative androgen-signalling-targeted inhibitor. Patients who received combination therapy for mHSPC were excluded (n = 33) as docetaxel is not approved in this setting in Japan. Results A total of 222 patients (median age 70 years) were included in this subanalysis. Median number of cycles was higher for cabazitaxel versus androgen-signalling-targeted inhibitors (7 versus 4). Clinical outcomes favoured cabazitaxel over abiraterone or enzalutamide including, radiographic progression-free survival (rPFS; median 8.2 versus 3.4 months; P < 0.0001), overall survival (OS; 13.9 versus 11.8 months; P = 0.0102), PFS (4.4 versus 2.7 months; P < 0.0001), confirmed prostate-specific antigen response (37.0 versus 14.4%; P = 0.0006) and objective tumour response (38.9 versus 11.4%; P = 0.0036). For cabazitaxel versus androgen-signalling-targeted inhibitor, grade ≥ 3 adverse events occurred in 55% versus 44% of patients, with adverse events leading to death on study in 2.7% versus 5.7%. Conclusions Cabazitaxel significantly improved outcomes including rPFS and OS versus abiraterone or enzalutamide and are reflective of the Japanese patient population. Cabazitaxel should be considered the preferred treatment option over abiraterone or enzalutamide in this setting.
Collapse
Affiliation(s)
- Hiroyoshi Suzuki
- Department of Urology, Toho University Sakura Medical Center, Chiba, Japan
| | - Daniel Castellano
- Medical Oncology Department, 12 de Octubre University Hospital, Madrid, Spain
| | - Johann de Bono
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden Hospital, London, UK
| | - Cora N Sternberg
- Division of Hematology and Medical Oncology, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Karim Fizazi
- Department of Cancer Medicine, Gustave Roussy Institute and Paris Sud University, Villejuif, France
| | - Bertrand Tombal
- Division of Urology, Université Catholique de Louvain, Louvain, Belgium
| | | | | | - Ayse Ozatilgan
- Global Medical Affairs Oncology, Sanofi, Cambridge, MA, USA
| | | | - Ronald de Wit
- Department Medical Oncology, Erasmus University Hospital, Rotterdam, the Netherlands
| |
Collapse
|
18
|
Crabb SJ, Griffiths G, Marwood E, Dunkley D, Downs N, Martin K, Light M, Northey J, Wilding S, Whitehead A, Shaw E, Birtle AJ, Bahl A, Elliott T, Westbury C, Sundar S, Robinson A, Jagdev S, Kumar S, Rooney C, Salinas-Souza C, Stephens C, Khoo V, Jones RJ. Pan-AKT Inhibitor Capivasertib With Docetaxel and Prednisolone in Metastatic Castration-Resistant Prostate Cancer: A Randomized, Placebo-Controlled Phase II Trial (ProCAID). J Clin Oncol 2021; 39:190-201. [PMID: 33326257 PMCID: PMC8078455 DOI: 10.1200/jco.20.01576] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/05/2020] [Accepted: 10/20/2020] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Capivasertib is a pan-AKT inhibitor. Preclinical data indicate activity in metastatic castration-resistant prostate cancer (mCRPC) and synergism with docetaxel. PATIENTS AND METHODS ProCAID was a placebo controlled randomized phase II trial in mCRPC. Patients received up to ten 21-day cycles of docetaxel (75 mg/m2 intravenous, day 1) and prednisolone (5 mg twice daily, oral, day 1-21) and were randomly assigned (1:1) to oral capivasertib (320 mg twice daily, 4 days on/3 days off, from day 2 each cycle), or placebo, until disease progression. Treatment allocation used minimization factors: bone metastases; visceral metastases; investigational site; and prior abiraterone or enzalutamide. The primary objective, by intention to treat, determined if the addition of capivasertib prolonged a composite progression-free survival (cPFS) end point that included prostate-specific antigen progression events. cPFS and overall survival (OS) were also assessed by composite biomarker subgroup for PI3K/AKT/PTEN pathway activation status. RESULTS One hundred and fifty patients were enrolled. Median cPFS was 7.03 (95% CI, 6.28 to 8.25) and 6.70 months (95% CI, 5.52 to 7.36) with capivasertib and placebo respectively (hazard ratio [HR], 0.92; 80% CI, 0.73 to 1.16; one-sided P = .32). Median OS was 31.15 (95% CI, 20.07 to not reached) and 20.27 months (95% CI, 17.51 to 24.18), respectively (HR, 0.54; 95% CI, 0.34 to 0.88; two-sided P = .01). cPFS and OS results were consistent irrespective of PI3K/AKT/PTEN pathway activation status. Grade III-IV adverse events were equivalent between arms (62.2%). The most common adverse events of any grade deemed related to capivasertib were diarrhea, fatigue, nausea, and rash. CONCLUSION The addition of capivasertib to chemotherapy did not extend cPFS in mCRPC irrespective of PI3K/AKT/PTEN pathway activation status. The observed OS result (a secondary end point) will require prospective validation in future studies to address potential for bias.
Collapse
Affiliation(s)
- Simon J. Crabb
- Southampton Clinical Trials Unit, University of Southampton, Southampton, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- Southampton Experimental Cancer Medicine Centre, University of Southampton, Southampton, United Kingdom
| | - Gareth Griffiths
- Southampton Clinical Trials Unit, University of Southampton, Southampton, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Ellice Marwood
- Southampton Clinical Trials Unit, University of Southampton, Southampton, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Denise Dunkley
- Southampton Clinical Trials Unit, University of Southampton, Southampton, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- Southampton Experimental Cancer Medicine Centre, University of Southampton, Southampton, United Kingdom
| | - Nichola Downs
- Southampton Clinical Trials Unit, University of Southampton, Southampton, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Karen Martin
- Southampton Clinical Trials Unit, University of Southampton, Southampton, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Michelle Light
- Southampton Clinical Trials Unit, University of Southampton, Southampton, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Josh Northey
- Southampton Clinical Trials Unit, University of Southampton, Southampton, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Sam Wilding
- Southampton Clinical Trials Unit, University of Southampton, Southampton, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Amy Whitehead
- Southampton Clinical Trials Unit, University of Southampton, Southampton, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Emily Shaw
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Alison J. Birtle
- Lancashire Teaching Hospitals NHS Foundation Trust, Preston, United Kingdom
| | - Amit Bahl
- Bristol Oncology and Haematology Centre, Bristol, United Kingdom
| | - Tony Elliott
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | | | - Santhanam Sundar
- Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | | | | | | | - Claire Rooney
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | | | - Christine Stephens
- Early Oncology Clinical, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Vincent Khoo
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Robert J. Jones
- University of Glasgow, Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| |
Collapse
|
19
|
Conteduca V, Mosca A, Brighi N, de Giorgi U, Rescigno P. New Prognostic Biomarkers in Metastatic Castration-Resistant Prostate Cancer. Cells 2021; 10:193. [PMID: 33478015 PMCID: PMC7835961 DOI: 10.3390/cells10010193] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer is one of the most frequent cancers in men and is a common cause of cancer-related death. Despite significant progress in the diagnosis and treatment of this tumor, patients who relapse after radical treatments inevitably develop metastatic disease. Patient stratification is therefore key in this type of cancer, and there is an urgent need for prognostic biomarkers that can define patients' risk of cancer-related death. In the last 10 years, multiple prognostic factors have been identified and studied. Here, we review the literature available and discuss the most common aberrant genomic pathways in metastatic castration-resistant prostate cancer shown to have a prognostic relevance in this setting.
Collapse
Affiliation(s)
- Vincenza Conteduca
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.C.); (N.B.); (U.d.G.)
| | - Alessandra Mosca
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy;
| | - Nicole Brighi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.C.); (N.B.); (U.d.G.)
| | - Ugo de Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.C.); (N.B.); (U.d.G.)
| | - Pasquale Rescigno
- Interdisciplinary Group for Translational Research and Clinical Trials, Urological Cancers (GIRT-Uro), Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy
| |
Collapse
|
20
|
Rescigno P, Gurel B, Pereira R, Crespo M, Rekowski J, Rediti M, Barrero M, Mateo J, Bianchini D, Messina C, Fenor de la Maza MD, Chandran K, Carmichael J, Guo C, Paschalis A, Sharp A, Seed G, Figueiredo I, Lambros M, Miranda S, Ferreira A, Bertan C, Riisnaes R, Porta N, Yuan W, Carreira S, de Bono JS. Characterizing CDK12-Mutated Prostate Cancers. Clin Cancer Res 2021; 27:566-574. [PMID: 32988971 PMCID: PMC7855716 DOI: 10.1158/1078-0432.ccr-20-2371] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/17/2020] [Accepted: 09/23/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Cyclin-dependent kinase 12 (CDK12) aberrations have been reported as a biomarker of response to immunotherapy for metastatic castration-resistant prostate cancer (mCRPC). Herein, we characterize CDK12-mutated mCRPC, presenting clinical, genomic, and tumor-infiltrating lymphocyte (TIL) data. EXPERIMENTAL DESIGN Patients with mCRPC consented to the molecular analyses of diagnostic and mCRPC biopsies. Genomic analyses involved targeted next-generation (MiSeq; Illumina) and exome sequencing (NovaSeq; Illumina). TILs were assessed by validated immunocytochemistry coupled with deep learning-based artificial intelligence analyses including multiplex immunofluorescence assays for CD4, CD8, and FOXP3 evaluating TIL subsets. The control group comprised a randomly selected mCRPC cohort with sequencing and clinical data available. RESULTS Biopsies from 913 patients underwent targeted sequencing between February 2015 and October 2019. Forty-three patients (4.7%) had tumors with CDK12 alterations. CDK12-altered cancers had distinctive features, with some revealing high chromosomal break numbers in exome sequencing. Biallelic CDK12-aberrant mCRPCs had shorter overall survival from diagnosis than controls [5.1 years (95% confidence interval (CI), 4.0-7.9) vs. 6.4 years (95% CI, 5.7-7.8); hazard ratio (HR), 1.65 (95% CI, 1.07-2.53); P = 0.02]. Median intratumoral CD3+ cell density was higher in CDK12 cancers, although this was not statistically significant (203.7 vs. 86.7 cells/mm2; P = 0.07). This infiltrate primarily comprised of CD4+FOXP3- cells (50.5 vs. 6.2 cells/mm2; P < 0.0001), where high counts tended to be associated with worse survival from diagnosis (HR, 1.64; 95% CI, 0.95-2.84; P = 0.077) in the overall population. CONCLUSIONS CDK12-altered mCRPCs have worse prognosis, with these tumors surprisingly being primarily enriched for CD4+FOXP3- cells that seem to associate with worse outcome and may be immunosuppressive.See related commentary by Lotan and Antonarakis, p. 380.
Collapse
Affiliation(s)
- Pasquale Rescigno
- The Institute of Cancer Research, Sutton, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, Sutton, London, United Kingdom
| | - Bora Gurel
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Rita Pereira
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Mateus Crespo
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Jan Rekowski
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Mattia Rediti
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Maialen Barrero
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Joaquin Mateo
- Vall d'Hebron Institute of Oncology (VHIO) and Vall d'Hebron University Hospital, Barcelona, Spain
| | - Diletta Bianchini
- The Royal Marsden NHS Foundation Trust, Sutton, London, United Kingdom
| | - Carlo Messina
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Maria D Fenor de la Maza
- The Institute of Cancer Research, Sutton, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, Sutton, London, United Kingdom
| | - Khobe Chandran
- The Royal Marsden NHS Foundation Trust, Sutton, London, United Kingdom
| | - Juliet Carmichael
- The Institute of Cancer Research, Sutton, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, Sutton, London, United Kingdom
| | - Christina Guo
- The Institute of Cancer Research, Sutton, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, Sutton, London, United Kingdom
| | - Alec Paschalis
- The Institute of Cancer Research, Sutton, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, Sutton, London, United Kingdom
| | - Adam Sharp
- The Institute of Cancer Research, Sutton, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, Sutton, London, United Kingdom
| | - George Seed
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Ines Figueiredo
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Maryou Lambros
- The Institute of Cancer Research, Sutton, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, Sutton, London, United Kingdom
| | - Susana Miranda
- The Institute of Cancer Research, Sutton, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, Sutton, London, United Kingdom
| | - Ana Ferreira
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Claudia Bertan
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Ruth Riisnaes
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Nuria Porta
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Wei Yuan
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Suzanne Carreira
- The Institute of Cancer Research, Sutton, London, United Kingdom
| | - Johann S de Bono
- The Institute of Cancer Research, Sutton, London, United Kingdom.
- The Royal Marsden NHS Foundation Trust, Sutton, London, United Kingdom
| |
Collapse
|
21
|
Caffo O, Maines F, Kinspergher S, Veccia A, Messina C. To treat or not to treat: is it acceptable to avoid active therapies in advanced prostate cancer today? Expert Rev Anticancer Ther 2020; 21:389-400. [PMID: 33245666 DOI: 10.1080/14737140.2021.1856661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Recently, there has been a paradigm shift in the treatment of advanced prostate cancer (PCa) because the approval of a number of new agents has significantly improved overall survival. However, as PCa is a heterogeneous disease that may be more or less aggressive and patients may be more or less responsive to treatment, it is often debated whether or not it is acceptable to avoid active therapies.Areas covered: This review discusses different settings of advanced PCa.Expert opinion: In metastatic castration-resistant PCa, it is unethical not to use active treatments but the use of both androgen receptor targeting agents (ARTA) in sequence should be avoided in most patients and the use of the available agents for fourth-line treatment or beyond should only be considered for highly selected patients. In metastatic hormone-sensitive PCa, patients with de novo disease should receive one additional agent in combination with androgen deprivation therapy (ADT), whereas patients in relapse should be managed with ADT alone. In non-metastatic castration-resistant prostate cancer (PCa), all patients with a PSA doubling time of ≤6 months should receive one ARTA, whereas the others might wait until there is an acceleration in the kinetics of their PSA levels.
Collapse
Affiliation(s)
- Orazio Caffo
- Department of Medical Oncology, Santa Chiara Hospital, Largo Medaglie d'Oro, Trento, Italy
| | - Francesca Maines
- Department of Medical Oncology, Santa Chiara Hospital, Largo Medaglie d'Oro, Trento, Italy
| | - Stefania Kinspergher
- Department of Medical Oncology, Santa Chiara Hospital, Largo Medaglie d'Oro, Trento, Italy
| | - Antonello Veccia
- Department of Medical Oncology, Santa Chiara Hospital, Largo Medaglie d'Oro, Trento, Italy
| | - Carlo Messina
- Department of Medical Oncology, Santa Chiara Hospital, Largo Medaglie d'Oro, Trento, Italy
| |
Collapse
|
22
|
Norz V, Rausch S. Treatment and resistance mechanisms in castration-resistant prostate cancer: new implications for clinical decision making? Expert Rev Anticancer Ther 2020; 21:149-163. [PMID: 33106066 DOI: 10.1080/14737140.2021.1843430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Introduction: The armamentarium of treatment options in metastatic and non-metastatic CRPC is rapidly evolving. However, the question of how individual treatment decisions should be balanced by available predictive clinical parameters, pharmacogenetic and drug interaction profiles, or compound-associated molecular biomarkers is a major challenge for clinical practice.Areas covered: We discuss treatment and resistance mechanisms in PC with regard to their association to drug efficacy and tolerability. Current efforts of combination treatment and putative predictive biomarkers of available and upcoming compounds are highlighted with regard to their implication on clinical decision-making.Expert opinion: Several treatment approaches are delineated, where identification of resistance mechanisms in CRPC may guide treatment selection. To date, most of these candidate biomarkers will however be found only in a small subset of patients. While current approaches of combination treatment in CRPC are proving synergistic effects on cancer biology, higher complexity with regard to biomarker analysis and interaction profiles of the respective compounds may be expected. Among other aspects of personalized treatment, consideration of drug-drug interaction and pharmacogenetics is an underrepresented issue. However, the non-metastatic castration resistant prostate cancer situation may be an example for treatment selection based on drug interaction profiles in the future.
Collapse
Affiliation(s)
- Valentina Norz
- Department of Urology, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| | - Steffen Rausch
- Department of Urology, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| |
Collapse
|
23
|
Turnham DJ, Bullock N, Dass MS, Staffurth JN, Pearson HB. The PTEN Conundrum: How to Target PTEN-Deficient Prostate Cancer. Cells 2020; 9:E2342. [PMID: 33105713 PMCID: PMC7690430 DOI: 10.3390/cells9112342] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
Loss of the tumor suppressor phosphatase and tensin homologue deleted on chromosome 10 (PTEN), which negatively regulates the PI3K-AKT-mTOR pathway, is strongly linked to advanced prostate cancer progression and poor clinical outcome. Accordingly, several therapeutic approaches are currently being explored to combat PTEN-deficient tumors. These include classical inhibition of the PI3K-AKT-mTOR signaling network, as well as new approaches that restore PTEN function, or target PTEN regulation of chromosome stability, DNA damage repair and the tumor microenvironment. While targeting PTEN-deficient prostate cancer remains a clinical challenge, new advances in the field of precision medicine indicate that PTEN loss provides a valuable biomarker to stratify prostate cancer patients for treatments, which may improve overall outcome. Here, we discuss the clinical implications of PTEN loss in the management of prostate cancer and review recent therapeutic advances in targeting PTEN-deficient prostate cancer. Deepening our understanding of how PTEN loss contributes to prostate cancer growth and therapeutic resistance will inform the design of future clinical studies and precision-medicine strategies that will ultimately improve patient care.
Collapse
Affiliation(s)
- Daniel J. Turnham
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| | - Nicholas Bullock
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Manisha S. Dass
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| | - John N. Staffurth
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| |
Collapse
|
24
|
de Wit R, de Bono J, Sternberg CN, Fizazi K, Tombal B, Wülfing C, Kramer G, Eymard JC, Bamias A, Carles J, Iacovelli R, Melichar B, Sverrisdóttir Á, Theodore C, Feyerabend S, Helissey C, Ozatilgan A, Geffriaud-Ricouard C, Castellano D. Cabazitaxel versus Abiraterone or Enzalutamide in Metastatic Prostate Cancer. N Engl J Med 2019; 381:2506-2518. [PMID: 31566937 DOI: 10.1056/nejmoa1911206] [Citation(s) in RCA: 430] [Impact Index Per Article: 71.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The efficacy and safety of cabazitaxel, as compared with an androgen-signaling-targeted inhibitor (abiraterone or enzalutamide), in patients with metastatic castration-resistant prostate cancer who were previously treated with docetaxel and had progression within 12 months while receiving the alternative inhibitor (abiraterone or enzalutamide) are unclear. METHODS We randomly assigned, in a 1:1 ratio, patients who had previously received docetaxel and an androgen-signaling-targeted inhibitor (abiraterone or enzalutamide) to receive cabazitaxel (at a dose of 25 mg per square meter of body-surface area intravenously every 3 weeks, plus prednisone daily and granulocyte colony-stimulating factor) or the other androgen-signaling-targeted inhibitor (either 1000 mg of abiraterone plus prednisone daily or 160 mg of enzalutamide daily). The primary end point was imaging-based progression-free survival. Secondary end points of survival, response, and safety were assessed. RESULTS A total of 255 patients underwent randomization. After a median follow-up of 9.2 months, imaging-based progression or death was reported in 95 of 129 patients (73.6%) in the cabazitaxel group, as compared with 101 of 126 patients (80.2%) in the group that received an androgen-signaling-targeted inhibitor (hazard ratio, 0.54; 95% confidence interval [CI], 0.40 to 0.73; P<0.001). The median imaging-based progression-free survival was 8.0 months with cabazitaxel and 3.7 months with the androgen-signaling-targeted inhibitor. The median overall survival was 13.6 months with cabazitaxel and 11.0 months with the androgen-signaling-targeted inhibitor (hazard ratio for death, 0.64; 95% CI, 0.46 to 0.89; P = 0.008). The median progression-free survival was 4.4 months with cabazitaxel and 2.7 months with an androgen-signaling-targeted inhibitor (hazard ratio for progression or death, 0.52; 95% CI, 0.40 to 0.68; P<0.001), a prostate-specific antigen response occurred in 35.7% and 13.5% of the patients, respectively (P<0.001), and tumor response was noted in 36.5% and 11.5% (P = 0.004). Adverse events of grade 3 or higher occurred in 56.3% of patients receiving cabazitaxel and in 52.4% of those receiving an androgen-signaling-targeted inhibitor. No new safety signals were observed. CONCLUSIONS Cabazitaxel significantly improved a number of clinical outcomes, as compared with the androgen-signaling-targeted inhibitor (abiraterone or enzalutamide), in patients with metastatic castration-resistant prostate cancer who had been previously treated with docetaxel and the alternative androgen-signaling-targeted agent (abiraterone or enzalutamide). (Funded by Sanofi; CARD ClinicalTrials.gov number, NCT02485691.).
Collapse
Affiliation(s)
- Ronald de Wit
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Johann de Bono
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Cora N Sternberg
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Karim Fizazi
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Bertrand Tombal
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Christian Wülfing
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Gero Kramer
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Jean-Christophe Eymard
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Aristotelis Bamias
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Joan Carles
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Roberto Iacovelli
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Bohuslav Melichar
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Ásgerður Sverrisdóttir
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Christine Theodore
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Susan Feyerabend
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Carole Helissey
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Ayse Ozatilgan
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Christine Geffriaud-Ricouard
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| | - Daniel Castellano
- From the Erasmus Medical Center, Rotterdam, the Netherlands (R.W.); the Institute of Cancer Research and the Royal Marsden Hospital, London (J.B.); Englander Institute for Precision Medicine, Weill Cornell Medicine, New York (C.N.S.); Institut Gustave Roussy and University of Paris Sud, Villejuif (K.F.), Jean Godinot Institute and Reims Champagne-Ardenne University, Reims (J.-C.E.), Foch Hospital, Suresnes (C.T.), Hôpital d'Instruction des Armées Bégin, Saint Mandé (C.H.), and Sanofi, Europe Medical Oncology, Paris (C.G.-R.) - all in France; Institut de Recherche Clinique, Université Catholique de Louvain, Louvain, Belgium (B.T.); the Department of Urology, Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg (C.W.), and Studienpraxis Urologie, Nürtingen (S.F.) - both in Germany; the Medical University of Vienna, Vienna (G.K.); Alexandra Hospital, National and Kapodistrian University of Athens, Athens (A.B.); Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona (J.C.); Azienda Ospedaliera Universitaria Integrata, Verona, and Fondazione Policlinico Agostino Gemelli IRCCS, Rome - both in Italy (R.I.); Palacky University Medical School and Teaching Hospital, Olomouc, Czech Republic (B.M.); Landspitali University Hospital, Reykjavik, Iceland (Á.S.); Sanofi, Global Medical Oncology, Cambridge, MA (A.O.); and 12 de Octubre University Hospital, Madrid (D.C.)
| |
Collapse
|
25
|
Sekino Y, Han X, Kawaguchi T, Babasaki T, Goto K, Inoue S, Hayashi T, Teishima J, Shiota M, Yasui W, Matsubara A. TUBB3 Reverses Resistance to Docetaxel and Cabazitaxel in Prostate Cancer. Int J Mol Sci 2019; 20:ijms20163936. [PMID: 31412591 PMCID: PMC6719236 DOI: 10.3390/ijms20163936] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/03/2019] [Accepted: 08/05/2019] [Indexed: 12/18/2022] Open
Abstract
Recent studies have reported that TUBB3 overexpression is involved in docetaxel (DTX) resistance in prostate cancer (PCa). The aim of this study was to clarify the role of TUBB3 in DTX and cabazitaxel (CBZ) resistance, and cross-resistance between DTX and CBZ in PCa. We analyzed the effect of TUBB3 knockdown on DTX and CBZ resistance and examined the interaction between TUBB3 and PTEN. We also investigated the role of phosphoinositide 3-kinases (PI3K) inhibitor (LY294002) in DTX and CBZ resistance. TUBB3 expression was upregulated in DTX-resistant and CBZ-resistant cells. TUBB3 knockdown re-sensitized DTX-resistant cells to DTX and CBZ-resistant cells to CBZ. Additionally, TUBB3 knockdown re-sensitized DTX-resistant cell lines to CBZ, indicating that TUBB3 mediates cross-resistance between DTX and CBZ. Knockdown of TUBB3 enhanced PTEN expression, and PTEN knockout enhanced TUBB3 expression. LY294002 suppressed TUBB3 expression in DTX-resistant and CBZ-resistant cell lines. LY294002 re-sensitized DTX-resistant cell lines to DTX and CBZ-resistant cell lines to CBZ. These results suggest that TUBB3 is involved in DTX resistance and CBZ resistance. A combination of LY294002/DTX and that of LY294002/CBZ could be potential strategies for PCa treatment.
Collapse
Affiliation(s)
- Yohei Sekino
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan.
| | - Xiangrui Han
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Takafumi Kawaguchi
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Takashi Babasaki
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Keisuke Goto
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Shogo Inoue
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Tetsutaro Hayashi
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Jun Teishima
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Wataru Yasui
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Akio Matsubara
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| |
Collapse
|
26
|
Marín‐Aguilera M, Reig Ò, Milà‐Guasch M, Font A, Domènech M, Rodríguez‐Vida A, Carles J, Suárez C, Alba AG, Jiménez N, Victoria I, Sala‐González N, Ribal MJ, López S, Etxaniz O, Anguera G, Maroto P, Fernández PL, Prat A, Mellado B. The influence of treatment sequence in the prognostic value of
TMPRSS2‐ERG
as biomarker of taxane resistance in castration‐resistant prostate cancer. Int J Cancer 2019; 145:1970-1981. [DOI: 10.1002/ijc.32238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/22/2019] [Accepted: 02/07/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Mercedes Marín‐Aguilera
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryFundació Clínic per a la Recerca Biomèdica Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
| | - Òscar Reig
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryFundació Clínic per a la Recerca Biomèdica Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
| | - Maria Milà‐Guasch
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
| | - Albert Font
- Department of Medical OncologyInstitut Català d'Oncologia Badalona Spain
| | | | | | - Joan Carles
- Department of Medical OncologyVall d'Hebron Institute of Oncology. Vall d'Hebron University Hospital Barcelona Spain
| | - Cristina Suárez
- Department of Medical OncologyVall d'Hebron Institute of Oncology. Vall d'Hebron University Hospital Barcelona Spain
| | - Aránzazu González Alba
- Department of Medical OncologyHospital Universitario Puerta de Hierro Majadahonda Madrid Spain
| | - Natalia Jiménez
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
| | - Iván Victoria
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryFundació Clínic per a la Recerca Biomèdica Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
| | | | - Maria José Ribal
- Department of UrologyHospital Clinic Barcelona Spain
- Faculty of MedicineUniversity of Barcelona Barcelona Spain
| | - Sandra López
- Department of Medical OncologyHospital Clínic Barcelona Spain
| | - Olatz Etxaniz
- Department of Medical OncologyInstitut Català d'Oncologia Badalona Spain
| | - Geòrgia Anguera
- Department of Medical OncologyHospital de la Santa Cruz y San Pablo Barcelona Spain
| | - Pablo Maroto
- Department of Medical OncologyHospital de la Santa Cruz y San Pablo Barcelona Spain
| | - Pedro Luis Fernández
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Faculty of MedicineUniversity of Barcelona Barcelona Spain
- Department of PathologyHospital Clínic Barcelona Spain
| | - Aleix Prat
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryFundació Clínic per a la Recerca Biomèdica Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
- Faculty of MedicineUniversity of Barcelona Barcelona Spain
| | - Begoña Mellado
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryFundació Clínic per a la Recerca Biomèdica Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
- Faculty of MedicineUniversity of Barcelona Barcelona Spain
| |
Collapse
|
27
|
Küronya Z, Sükösd F, Varga L, Bíró K, Gyergyay F, Géczi L, Nagyiványi K, Jorgo K, Szarvas T, Kovács Á, Laczó I, Varga Z, Pósfai B, Pepó J, Maráz A. ERG expression can predict the outcome of docetaxel combinedwith androgen deprivation therapy in metastatic hormone-sensitiveprostate cancer. Urol Oncol 2019; 37:289.e1-289.e9. [PMID: 30679082 DOI: 10.1016/j.urolonc.2018.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 12/02/2018] [Accepted: 12/05/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Our study aimed to analyze the potential association between clinical parameters and ERG expression and the outcome of docetaxel chemotherapy among patients with metastatic hormone-sensitive prostate cancer. PATIENTS AND METHODS Fifty-five patients with metastatic hormone-sensitive prostate cancer were treated with docetaxel in addition to androgen deprivation therapy. Patient characteristics, clinical factors, and tumor expression of ERG by immunohistochemistry were analyzed with respect to therapeutic response and survival data. RESULTS Relapse free survival (RFS) and overal survival (OS) were 10.5 and 40.4 months, respectively, and both correlated with PSA response (RFS: 16.8 with a ≥50% decrease in PSA vs. 5.9 months in the case of <50% decrease, P < 0.001; OS: 40.4 vs. 11.6 months, respectively, P < 0.001). There was an association between OS and early progression (OS: 40.4 months with progression after 12 months vs. 17.9 months with progression within 12 months, P = 0.009). ERG expression was detected in 21 (42%) samples. ERG positivity was associated with favorable RFS (ERG pos. vs. neg.: 26.0 vs. 11.4 months, P = 0.003). CONCLUSION ERG expression may have a potential predictive value with respect to the effectiveness and outcome of docetaxel chemotherapy combined with androgen deprivation therapy.
Collapse
Affiliation(s)
- Zsófia Küronya
- National Institute of Oncology, Department of Medical Oncology and Clinical Pharmacology "C", Budapest, Hungary
| | - Farkas Sükösd
- Department of Pathology, University of Szeged, Szeged, Hungary
| | - Linda Varga
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - Krisztina Bíró
- National Institute of Oncology, Department of Medical Oncology and Clinical Pharmacology "C", Budapest, Hungary
| | - Fruzsina Gyergyay
- National Institute of Oncology, Department of Medical Oncology and Clinical Pharmacology "C", Budapest, Hungary
| | - Lajos Géczi
- National Institute of Oncology, Department of Medical Oncology and Clinical Pharmacology "C", Budapest, Hungary
| | - Krisztián Nagyiványi
- National Institute of Oncology, Department of Medical Oncology and Clinical Pharmacology "C", Budapest, Hungary
| | - Kliton Jorgo
- National Institute of Oncology, Department of Radiotherapy, Budapest, Hungary; Semmelweis University, Department of Oncology, Budapest
| | - Tibor Szarvas
- Department of Urology, Semmelweis University, Budapest, Hungary
| | - Ágnes Kovács
- Boehringer Ingelheim RCV GmbH & Co. KG Branch Office Hungary, Budapest, Hungary
| | | | - Zoltán Varga
- University of Szeged, Department of Oncotherapy, Szeged, Hungary
| | - Boglárka Pósfai
- University of Szeged, Department of Oncotherapy, Szeged, Hungary
| | - Judit Pepó
- University of Szeged, Department of Oncotherapy, Szeged, Hungary
| | - Anikó Maráz
- University of Szeged, Department of Oncotherapy, Szeged, Hungary.
| |
Collapse
|