101
|
Wu A, Cremaschi P, Wetterskog D, Conteduca V, Franceschini GM, Kleftogiannis D, Jayaram A, Sandhu S, Wong SQ, Benelli M, Salvi S, Gurioli G, Feber A, Pereira MB, Wingate AM, Gonzalez-Billalebeitia E, De Giorgi U, Demichelis F, Lise S, Attard G. Genome-wide plasma DNA methylation features of metastatic prostate cancer. J Clin Invest 2020; 130:1991-2000. [PMID: 32149736 PMCID: PMC7108919 DOI: 10.1172/jci130887] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 01/08/2020] [Indexed: 12/25/2022] Open
Abstract
Tumor DNA circulates in the plasma of cancer patients admixed with DNA from noncancerous cells. The genomic landscape of plasma DNA has been characterized in metastatic castration-resistant prostate cancer (mCRPC) but the plasma methylome has not been extensively explored. Here, we performed next-generation sequencing (NGS) on plasma DNA with and without bisulfite treatment from mCRPC patients receiving either abiraterone or enzalutamide in the pre- or post-chemotherapy setting. Principal component analysis on the mCRPC plasma methylome indicated that the main contributor to methylation variance (principal component one, or PC1) was strongly correlated with genomically determined tumor fraction (r = -0.96; P < 10-8) and characterized by hypermethylation of targets of the polycomb repressor complex 2 components. Further deconvolution of the PC1 top-correlated segments revealed that these segments are comprised of methylation patterns specific to either prostate cancer or prostate normal epithelium. To extract information specific to an individual's cancer, we then focused on an orthogonal methylation signature, which revealed enrichment for androgen receptor binding sequences and hypomethylation of these segments associated with AR copy number gain. Individuals harboring this methylation pattern had a more aggressive clinical course. Plasma methylome analysis can accurately quantitate tumor fraction and identify distinct biologically relevant mCRPC phenotypes.
Collapse
Affiliation(s)
- Anjui Wu
- University College London Cancer Institute, London, United Kingdom
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Paolo Cremaschi
- University College London Cancer Institute, London, United Kingdom
| | | | - Vincenza Conteduca
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | | | | | - Anuradha Jayaram
- University College London Cancer Institute, London, United Kingdom
| | - Shahneen Sandhu
- Peter MacCallum Cancer Centre and the University of Melbourne, Melbourne, Victoria, Australia
| | - Stephen Q. Wong
- Peter MacCallum Cancer Centre and the University of Melbourne, Melbourne, Victoria, Australia
| | - Matteo Benelli
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Samanta Salvi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giorgia Gurioli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Andrew Feber
- University College London Cancer Institute, London, United Kingdom
| | | | | | - Enrique Gonzalez-Billalebeitia
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, IMIB-Universidad de Murcia, Murcia, Spain
| | - Ugo De Giorgi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Francesca Demichelis
- Centre for Integrative Biology, University of Trento, Trento, Italy
- Institute of Computational Biomedicine, Weill Cornell Medicine, New York, New York, USA
| | - Stefano Lise
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Gerhardt Attard
- University College London Cancer Institute, London, United Kingdom
| |
Collapse
|
102
|
Mateo J, Seed G, Bertan C, Rescigno P, Dolling D, Figueiredo I, Miranda S, Nava Rodrigues D, Gurel B, Clarke M, Atkin M, Chandler R, Messina C, Sumanasuriya S, Bianchini D, Barrero M, Petermolo A, Zafeiriou Z, Fontes M, Perez-Lopez R, Tunariu N, Fulton B, Jones R, McGovern U, Ralph C, Varughese M, Parikh O, Jain S, Elliott T, Sandhu S, Porta N, Hall E, Yuan W, Carreira S, de Bono JS. Genomics of lethal prostate cancer at diagnosis and castration resistance. J Clin Invest 2020; 130:1743-1751. [PMID: 31874108 PMCID: PMC7108902 DOI: 10.1172/jci132031] [Citation(s) in RCA: 192] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022] Open
Abstract
The genomics of primary prostate cancer differ from those of metastatic castration-resistant prostate cancer (mCRPC). We studied genomic aberrations in primary prostate cancer biopsies from patients who developed mCRPC, also studying matching, same-patient, diagnostic, and mCRPC biopsies following treatment. We profiled 470 treatment-naive prostate cancer diagnostic biopsies and, for 61 cases, mCRPC biopsies, using targeted and low-pass whole-genome sequencing (n = 52). Descriptive statistics were used to summarize mutation and copy number profile. Prevalence was compared using Fisher's exact test. Survival correlations were studied using log-rank test. TP53 (27%) and PTEN (12%) and DDR gene defects (BRCA2 7%; CDK12 5%; ATM 4%) were commonly detected. TP53, BRCA2, and CDK12 mutations were markedly more common than described in the TCGA cohort. Patients with RB1 loss in the primary tumor had a worse prognosis. Among 61 men with matched hormone-naive and mCRPC biopsies, differences were identified in AR, TP53, RB1, and PI3K/AKT mutational status between same-patient samples. In conclusion, the genomics of diagnostic prostatic biopsies acquired from men who develop mCRPC differ from those of the nonlethal primary prostatic cancers. RB1/TP53/AR aberrations are enriched in later stages, but the prevalence of DDR defects in diagnostic samples is similar to mCRPC.
Collapse
Affiliation(s)
- Joaquin Mateo
- Vall d’Hebron Institute of Oncology (VHIO) and Vall d’Hebron University Hospital, Barcelona, Spain
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - George Seed
- The Institute of Cancer Research, London, United Kingdom
| | - Claudia Bertan
- The Institute of Cancer Research, London, United Kingdom
| | - Pasquale Rescigno
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - David Dolling
- The Institute of Cancer Research, London, United Kingdom
| | | | - Susana Miranda
- The Institute of Cancer Research, London, United Kingdom
| | | | - Bora Gurel
- The Institute of Cancer Research, London, United Kingdom
| | - Matthew Clarke
- The Institute of Cancer Research, London, United Kingdom
| | - Mark Atkin
- The Institute of Cancer Research, London, United Kingdom
| | - Rob Chandler
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Carlo Messina
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Semini Sumanasuriya
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Diletta Bianchini
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Maialen Barrero
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Antonella Petermolo
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Zafeiris Zafeiriou
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Mariane Fontes
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Instituto Oncoclinicas-Grupo Oncoclinicas, Rio de Janeiro, Brazil
| | - Raquel Perez-Lopez
- Vall d’Hebron Institute of Oncology (VHIO) and Vall d’Hebron University Hospital, Barcelona, Spain
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Nina Tunariu
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Ben Fulton
- The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Robert Jones
- The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | | | - Christy Ralph
- St James’s University Hospital, Leeds, United Kingdom
| | | | - Omi Parikh
- Royal Blackburn Hospital, Blackburn, United Kingdom
| | - Suneil Jain
- Belfast City Hospital, Belfast, United Kingdom
| | - Tony Elliott
- The Christie Hospital, Manchester, United Kingdom
| | | | - Nuria Porta
- The Institute of Cancer Research, London, United Kingdom
| | - Emma Hall
- The Institute of Cancer Research, London, United Kingdom
| | - Wei Yuan
- The Institute of Cancer Research, London, United Kingdom
| | | | - Johann S. de Bono
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
103
|
Du M, Tian Y, Tan W, Wang L, Wang L, Kilari D, Huang CC, Wang L, Kohli M. Plasma cell-free DNA-based predictors of response to abiraterone acetate/prednisone and prognostic factors in metastatic castration-resistant prostate cancer. Prostate Cancer Prostatic Dis 2020; 23:705-713. [PMID: 32203070 PMCID: PMC7501185 DOI: 10.1038/s41391-020-0224-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/02/2020] [Accepted: 03/06/2020] [Indexed: 11/24/2022]
Abstract
Background: The combination of abiraterone acetate and prednisone (AA/P) is used to treat metastatic prostate cancer, but molecular predictors of treatment response are not well elucidated. We evaluated plasma circulating tumor DNA– (ctDNA-) based copy number alterations (CNAs) to determine treatment-related predictive and prognostic biomarkers for metastatic castration-resistant prostate cancer (mCRPC). Methods: Serial plasma specimens were prospectively collected from 88 chemotherapy-naive mCRPC patients before and after 12 weeks of AA/P treatment. Sequencing-based CNA analyses were performed on 174 specimens. We evaluated CNA-associated 12-week responses for primary resistance, time to treatment change (TTTC) for secondary resistance, and overall survival for prognosis (P < .05). Associations with primary resistance were analyzed using the Fisher exact test. Kaplan–Meier survival curves and Cox regression analyses were used to determine the associations of CNAs with acquired resistance and overall survival. Results: ctDNA reduced by 3.89% in responders and increased by 0.94% in nonresponders (P = .0043). Thirty-one prostate cancer–related genes from whole genome CNAs were tested. AR and AR enhancer amplification were associated with primary resistance (P = .0039) and shorter TTTC (P = .0003). ZFHX3 deletion and PIK3CA amplification were associated with primary resistance (P = .026 and P = .017, respectively), shorter TTTC (P = .0008 and P= .0016, respectively), and poor survival (P = .0025 and P = .0022, respectively). CNA-based risk scores combining selected significant associations (AR, NKX3.1, and PIK3CA) at the univariate level with TTTC were predictive of secondary resistance (P = .0002). and established prognoses for survival based on CNAs in ZFHX3, RB1, PIK3CA, and OPHN1 (P = .002). Multigene risk scores were more predictive than individual genes or clinical risk factors (P < .05). Conclusion: Plasma ctDNA CNAs and risk scores can predict mCRPC-state treatment and survival outcomes.
Collapse
Affiliation(s)
- Meijun Du
- Department of the Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yijun Tian
- Department of the Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, WI, USA
| | - Winston Tan
- Department of Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Liguo Wang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Deepak Kilari
- Department of Medicine, Medical College of Wisconsin and Milwaukee VA Medical Center, Milwaukee, WI, USA
| | - Chiang-Ching Huang
- Department of Biostatistics, University of Wisconsin, Milwaukee, WI, USA
| | - Liang Wang
- Department of the Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, WI, USA. .,Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Manish Kohli
- Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
104
|
Maia MC, Salgia M, Pal SK. Harnessing cell-free DNA: plasma circulating tumour DNA for liquid biopsy in genitourinary cancers. Nat Rev Urol 2020; 17:271-291. [PMID: 32203306 DOI: 10.1038/s41585-020-0297-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2020] [Indexed: 12/11/2022]
Abstract
In the era of precision oncology, liquid biopsy techniques, especially the use of plasma circulating tumour DNA (ctDNA) analysis, represent a paradigm shift in the use of genomic biomarkers with considerable implications for clinical practice. Compared with tissue-based tumour DNA analysis, plasma ctDNA is more convenient to test, more readily accessible, faster to obtain and less invasive, minimizing procedure-related risks and offering the opportunity to perform serial monitoring. Additionally, genomic profiles of ctDNA have been shown to reflect tumour heterogeneity, which has important implications for the identification of resistant clones and selection of targeted therapy well before clinical and radiographic changes occur. Moreover, plasma ctDNA testing can also be applied to cancer screening, risk stratification and quantification of minimal residual disease. These features provide an unprecedented opportunity for early treatment of patients, improving the chances of treatment success.
Collapse
Affiliation(s)
- Manuel Caitano Maia
- Department of Medical Oncology, Centro de Oncologia do Paraná, Curitiba, PR, Brazil. .,Latin American Cooperative Oncology Group, Genitourinary Group, Porto Alegre, Brazil.
| | - Meghan Salgia
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Sumanta K Pal
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
105
|
Brown LC, Lu C, Antonarakis ES, Luo J, Armstrong AJ. Androgen receptor variant-driven prostate cancer II: advances in clinical investigation. Prostate Cancer Prostatic Dis 2020; 23:367-380. [PMID: 32094489 DOI: 10.1038/s41391-020-0215-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/01/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Approximately 10-30% of men with mCRPC will test positive for AR-V7 using one of two analytically and clinically validated circulating tumor cell (CTC)-based assays. These men have poor outcomes with approved AR-targeting therapies but may retain sensitivity to chemotherapy. Here, we discuss the clinical implications of testing and strategies that may benefit AR splice variant (AR-V)-positive men and discuss whether such variants are passengers or drivers of aggressive clinical behavior. METHODS We conducted a systemic review of the literature, covering updates since our 2016 review on androgen receptor variants in mCRPC, outcomes, and existing and novel approaches to therapy. We provide an expert opinion about management strategies for AR-V7-positive men and key unanswered research questions. RESULTS AR-V7-positive men, defined by Epic nuclear protein detection or the modified AdnaTest mRNA detection in CTCs, identify a subset of men with mCRPC that have a low probability of response to AR-targeting therapy with short progression-free and overall survival in multivariable analyses. AR-variants do not exist in isolation, but rather in the context of a complex, heterogeneous, and evolving mCRPC genome and phenotype as well as patient-specific clinical heterogeneity, and multiple mechanisms of resistance likely exist in patients regardless of AR-V7 detection. Efforts to develop broader resistance assays are needed, and effective treatment strategies beyond taxanes are needed to address the causal driver role of AR-variants and to benefit patients with AR-V-expressing prostate cancer. CONCLUSIONS CTC AR-V7 detection using the AdnaTest mRNA or Epic nuclear protein assays represents the first analytically and prospective clinically validated liquid biopsy assays that may inform treatment decisions in men with mCRPC, particularly after failure of first-line AR-therapy. The importance of AR-variants is likely to increase with the earlier use of AR-targeting strategies in other settings, and novel interventions for these men are needed.
Collapse
Affiliation(s)
- Landon C Brown
- Departments of Medicine, Surgery, and Pharmacology and Cancer Biology, Divisions of Medical Oncology and Urology, Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC, USA
| | - Changxue Lu
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emmanuel S Antonarakis
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jun Luo
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew J Armstrong
- Departments of Medicine, Surgery, and Pharmacology and Cancer Biology, Divisions of Medical Oncology and Urology, Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC, USA.
| |
Collapse
|
106
|
Circulating cell-free DNA: Translating prostate cancer genomics into clinical care. Mol Aspects Med 2020; 72:100837. [PMID: 31954523 DOI: 10.1016/j.mam.2019.100837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/12/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
Abstract
Only in the past decade tremendous advances have been made in understanding prostate cancer genomics and consequently in applying new treatment strategies. As options regarding treatments are increasing so are the challenges in selecting the right treatment option for each patient and not the least, understanding the optimal time-point and sequence of applying available treatments. Critically, without reliable methods that enable sequential monitoring of evolving genotypes in individual patients, we will never reach effective personalised driven treatment approaches. This review focuses on the clinical implications of prostate cancer genomics and the potential of cfDNA in facilitating treatment management.
Collapse
|
107
|
Bacon JVW, Annala M, Soleimani M, Lavoie JM, So A, Gleave ME, Fazli L, Wang G, Chi KN, Kollmannsberger CK, Wyatt AW, Nappi L. Plasma Circulating Tumor DNA and Clonal Hematopoiesis in Metastatic Renal Cell Carcinoma. Clin Genitourin Cancer 2020; 18:322-331.e2. [PMID: 32046920 DOI: 10.1016/j.clgc.2019.12.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 12/26/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND There is a lack of molecularly-informed biomarkers for patients with metastatic renal cell carcinoma (RCC). Plasma cell-free DNA (cfDNA) sequencing is a minimally-invasive alternative to tissue for profiling the genome in other cancers but relevance in metastatic RCC remains unclear. MATERIALS AND METHODS Whole blood was collected from 55 patients with metastatic RCC. Plasma cfDNA and leukocyte DNA were subjected to targeted sequencing across 981 cancer genes. Matched tumor tissue from 14 patients was analyzed. RESULTS Thirty-three percent of patients had evidence for RCC-derived circulating tumor DNA (ctDNA), significantly lower than patients with metastatic prostate or bladder cancer analyzed using the same approach. Among ctDNA-positive patients, ctDNA fraction averaged only 3.9% and showed no strong association with clinical variables. In these patients, the most commonly mutated genes were VHL, BAP1, and PBRM1, and matched tissue concordance was 77%. Evidence of somatic expansions unrelated to RCC, such as clonal hematopoiesis of indeterminate potential, were detected in 43% of patients. Pathogenic germline mutations in DNA repair genes were detected in 11% of patients. CtDNA-positive patients had shorter overall survival and progression-free survival on first-line therapy. Patients with evidence of clonal hematopoiesis of indeterminate potential had an intermediate prognosis compared with ctDNA-positive and -negative patients. CONCLUSIONS CfDNA sequencing enables straightforward characterization of the somatic RCC genome in a minority of patients with metastatic RCC. Owing to low ctDNA abundance, and the presence of non-RCC derived somatic clones in circulation, cfDNA sequencing may not be a simple pan-patient alternative to tissue biopsy in metastatic RCC.
Collapse
Affiliation(s)
- Jack V W Bacon
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Matti Annala
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Maryam Soleimani
- Department of Medical Oncology, BC Cancer, British Columbia, Canada
| | | | - Alan So
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Martin E Gleave
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Gang Wang
- Department of Medical Oncology, BC Cancer, British Columbia, Canada
| | - Kim N Chi
- Department of Medical Oncology, BC Cancer, British Columbia, Canada
| | | | - Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada.
| | - Lucia Nappi
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada; Department of Medical Oncology, BC Cancer, British Columbia, Canada
| |
Collapse
|
108
|
Gupta S, Hovelson DH, Kemeny G, Halabi S, Foo WC, Anand M, Somarelli JA, Tomlins SA, Antonarakis ES, Luo J, Dittamore RV, George DJ, Rothwell C, Nanus DM, Armstrong AJ, Gregory SG. Discordant and heterogeneous clinically relevant genomic alterations in circulating tumor cells vs plasma DNA from men with metastatic castration resistant prostate cancer. Genes Chromosomes Cancer 2019; 59:225-239. [PMID: 31705765 DOI: 10.1002/gcc.22824] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Circulating tumor cell (CTC) and cell-free (cf) DNA-based genomic alterations are increasingly being used for clinical decision-making in oncology. However, the concordance and discordance between paired CTC and cfDNA genomic profiles remain largely unknown. We performed comparative genomic hybridization (CGH) on CTCs and cfDNA, and low-pass whole genome sequencing (lpWGS) on cfDNA to characterize genomic alterations (CNA) and tumor content in two independent prospective studies of 93 men with mCRPC treated with enzalutamide/abiraterone, or radium-223. Comprehensive analysis of 69 patient CTCs and 72 cfDNA samples from 93 men with mCRPC, including 64 paired samples, identified common concordant gains in FOXA1, AR, and MYC, and losses in BRCA1, PTEN, and RB1 between CTCs and cfDNA. Concordant PTEN loss and discordant BRCA2 gain were associated with significantly worse outcomes in Epic AR-V7 negative men with mCRPC treated with abiraterone/enzalutamide. We identified and externally validated CTC-specific genomic alternations that were discordant in paired cfDNA, even in samples with high tumor content. These CTC/cfDNA-discordant regions included key genomic regulators of lineage plasticity, osteomimicry, and cellular differentiation, including MYCN gain in CTCs (31%) that was rarely detected in cfDNA. CTC MYCN gain was associated with poor clinical outcomes in AR-V7 negative men and small cell transformation. In conclusion, we demonstrated concordance of multiple genomic alterations across CTC and cfDNA platforms; however, some genomic alterations displayed substantial discordance between CTC DNA and cfDNA despite the use of identical copy number analysis methods, suggesting tumor heterogeneity and divergent evolution associated with poor clinical outcomes.
Collapse
Affiliation(s)
- Santosh Gupta
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina.,Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Daniel H Hovelson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Gabor Kemeny
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina
| | - Wen-Chi Foo
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Monika Anand
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Jason A Somarelli
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina.,Department of Medicine, Surgery, Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Scott A Tomlins
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Emmanuel S Antonarakis
- Prostate Cancer Research Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jun Luo
- James Buchanan Brady Urological Institute and the Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Daniel J George
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Colin Rothwell
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - David M Nanus
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina.,Department of Medicine, Surgery, Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Simon G Gregory
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina.,Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| |
Collapse
|
109
|
Chapman L, Ledet EM, Barata PC, Cotogno P, Manogue C, Moses M, Christensen BR, Steinwald P, Ranasinghe L, Layton JL, Lewis BE, Sartor O. TP53 Gain-of-Function Mutations in Circulating Tumor DNA in Men With Metastatic Castration-Resistant Prostate Cancer. Clin Genitourin Cancer 2019; 18:148-154. [PMID: 31822380 DOI: 10.1016/j.clgc.2019.10.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/10/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Circulating tumor DNA (ctDNA), which can be assessed by liquid biopsy, can provide valuable genomic information that may affect treatment response in prostate cancer. The aim of this study was to characterize TP53 mutations and treatment history in prostate cancer. PATIENTS AND METHODS This study included 143 patients with metastatic castration-resistant prostate cancer who had undergone ctDNA sequencing via Guardant360 testing. The presence or absence of TP53 mutations was analyzed along with treatment history for this group. TP53 mutations were further classified as gain of function (GOF) or not GOF, and analyzed with prior therapies. RESULTS Chi-square analysis was performed for treatment history and TP53 status (further specified as all TP53 mutations or only TP53 GOF mutations). There were no associations between prior receipt of abiraterone/enzalutamide therapy and all TP53 mutations, or between docetaxel therapy and all TP53 mutations. However, TP53 GOF mutations had a positive association with prior abiraterone/enzalutamide therapy (P = .047). There was no association of TP53 GOF mutations with prior docetaxel therapy. The most frequent alterations co-occurring with all TP53 mutations were in AR, BRAF, EGFR, MYC, and PIK3CA. Common coalterations with TP53 GOF mutations included AR, BRAF, EGFR, RB1, NF1, and PIK3CA. There was an association of RB1 mutations with TP53 GOF mutations, versus RB1 mutations and no TP53 GOF mutations (P = .0036). CONCLUSION TP53 GOF mutations may provide a valuable pathway to delineate metastatic castration-resistant prostate cancer TP53 mutations into therapeutic categories. Association with disease progression while receiving abiraterone/enzalutamide therapy was apparent in this study; however, further studies are needed to elaborate the therapeutic and prognostic implications.
Collapse
Affiliation(s)
- Lynne Chapman
- Tulane University School of Medicine, New Orleans, LA
| | - Elisa M Ledet
- Tulane University School of Medicine, New Orleans, LA; Tulane Cancer Center, New Orleans, LA
| | - Pedro C Barata
- Tulane University School of Medicine, New Orleans, LA; Tulane Cancer Center, New Orleans, LA
| | - Patrick Cotogno
- Tulane University School of Medicine, New Orleans, LA; Tulane Cancer Center, New Orleans, LA
| | - Charlotte Manogue
- Tulane University School of Medicine, New Orleans, LA; Tulane Cancer Center, New Orleans, LA
| | - Marcus Moses
- Tulane University School of Medicine, New Orleans, LA; Tulane Cancer Center, New Orleans, LA
| | | | | | | | | | | | - Oliver Sartor
- Tulane University School of Medicine, New Orleans, LA; Tulane Cancer Center, New Orleans, LA.
| |
Collapse
|
110
|
Josefsson A, Damber JE, Welén K. AR-V7 expression in circulating tumor cells as a potential prognostic marker in metastatic hormone-sensitive prostate cancer. Acta Oncol 2019; 58:1660-1664. [PMID: 31286815 DOI: 10.1080/0284186x.2019.1637540] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Andreas Josefsson
- Department of Urology, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgical and Perioperative Sciences, Urology & Andrology, Umeå University, Umeå, Sweden
- Wallenberg Center for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Jan-Erik Damber
- Department of Urology, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Welén
- Department of Urology, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
111
|
Mollica V, Di Nunno V, Santoni M, Cimadamore A, Scarpelli M, Lopez-Beltran A, Cheng L, Mariani C, Battelli N, Montironi R, Massari F. An evaluation of current prostate cancer diagnostic approaches with emphasis on liquid biopsies and prostate cancer. Expert Rev Mol Diagn 2019; 20:207-217. [PMID: 31640441 DOI: 10.1080/14737159.2019.1684265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: Knowledge of the complex biology of prostate cancer is constantly growing, opening the field up to new therapeutic advances. The selection of patients on the basis of prognostic and predictive biomarkers is a challenging and emerging clinical need, not yet completely fulfilled. In this scenario, liquid biopsy offers a noninvasive and attractive approach to give important information about tumor biology and eventual resistance to treatments.Areas covered: The aim of this review of the literature is to evaluate the current knowledge and the promising value of liquid biopsy in prostate cancer. Circulating tumor cells and circulating tumor DNA identified by liquid biopsies are currently under evaluation to guide therapeutic decisions in prostate cancer management, even though practical applications of these approaches are still very limited. We examined the current areas of interest in which circulating tumor cells and circulating tumor DNA are being investigated, such as their prognostic and predictive role in response to chemotherapy or androgen receptor signaling inhibition, especially in the castration-resistant setting.Expert opinion: As the body of knowledge on liquid biopsy rapidly grows, we need to identify which can be the real applications of this technique in clinical practice and to overcome the problems that are limiting its routinely use.
Collapse
Affiliation(s)
- Veronica Mollica
- Division of Oncology, S.Orsola-Malpighi Hospital, Bologna, Italy
| | | | | | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | | |
Collapse
|
112
|
Boerrigter E, Groen LN, Van Erp NP, Verhaegh GW, Schalken JA. Clinical utility of emerging biomarkers in prostate cancer liquid biopsies. Expert Rev Mol Diagn 2019; 20:219-230. [DOI: 10.1080/14737159.2019.1675515] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Emmy Boerrigter
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Levi N. Groen
- Department of Experimental Urology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Nielka P. Van Erp
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Gerald W. Verhaegh
- Department of Experimental Urology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Jack A. Schalken
- Department of Experimental Urology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
113
|
Jayaram A, Wingate A, Wetterskog D, Conteduca V, Khalaf D, Sharabiani MTA, Calabrò F, Barwell L, Feyerabend S, Grande E, Martinez-Carrasco A, Font A, Berruti A, Sternberg CN, Jones R, Lefresne F, Lahaye M, Thomas S, Joshi S, Shen D, Ricci D, Gormley M, Merseburger AS, Tombal B, Annala M, Chi KN, De Giorgi U, Gonzalez-Billalabeitia E, Wyatt AW, Attard G. Plasma Androgen Receptor Copy Number Status at Emergence of Metastatic Castration-Resistant Prostate Cancer: A Pooled Multicohort Analysis. JCO Precis Oncol 2019; 3:1900123. [PMID: 32923850 DOI: 10.1200/po.19.00123] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2019] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Increases in androgen receptor (AR) copy number (CN) can be detected in plasma DNA when patients develop metastatic castration-resistant prostate cancer. We aim to evaluate the association between AR CN as a continuous variable and clinical outcome. PATIENTS AND METHODS PCR2023 was an international, multi-institution, open-label, phase II study of abiraterone acetate plus prednisolone (AAP) or abiraterone acetate plus dexamethasone that included plasma AR assessment as a predefined exploratory secondary end point. Plasma AR CN data (ClinicalTrials.gov identifier: NCT01867710) from this study (n = 133) were pooled with data from the following three other cohorts: cohort A, which was treated with either AAP or enzalutamide (n = 73); the PREMIERE trial (ClinicalTrials.gov identifier: NCT02288936) of biomarkers for enzalutamide (n = 94); and a phase II trial from British Columbia (ClinicalTrials.gov identifier: NCT02125357) that randomly assigned men to either AAP or enzalutamide (n = 201). The primary outcome measures for the biomarker analysis were overall survival and progression-free survival. RESULTS Using multivariable fractional polynomials analysis using Cox regression models, a nonlinear relationship between plasma AR CN and outcome was identified for overall survival, where initially for small incremental gains in CN there was a large added hazard ratio that plateaued at higher CN. The CN cut point associated with the highest local hazard ratio was 1.92. A similar nonlinear association was observed with progression-free survival. In an exploratory analysis of PCR2023, the time from start of long-term androgen-deprivation therapy to start of AAP or abiraterone acetate plus dexamethasone was significantly shorter in patients with plasma AR CN of 1.92 or greater than patients with plasma AR CN of less than 1.92 (43 v 130 weeks, respectively; P = .005). This was confirmed in cohort A (P = .003), the PREMIERE cohort (P = .03), and the British Colombia cohort (P = .003). CONCLUSION Patients with metastatic castration-resistant prostate cancer can be dichotomized by a plasma AR CN cut point of 1.92. Plasma AR CN value of 1.92 or greater identifies aggressive disease that is poorly responsive to AR targeting and is associated with a prior short response to primary androgen-deprivation therapy.
Collapse
Affiliation(s)
- Anuradha Jayaram
- University College London Cancer Institute, London, United Kingdom
| | - Anna Wingate
- University College London Cancer Institute, London, United Kingdom
| | | | - Vincenza Conteduca
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Daniel Khalaf
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - Lorraine Barwell
- University of Glasgow, The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | | | | | - Alberto Martinez-Carrasco
- Hospital Universitario Morales Meseguer, Biobanco Nodo 3, Instituto Murciano de Investigación Biosanitaria-Universidad de Murcia, Murcia, Spain
| | - Albert Font
- Institut Catala d'Oncologia-Hospital Germans Trias i Pujol, Badalona, Spain
| | - Alfredo Berruti
- University of Brescia, Spedali Civili Hospital, Brescia, Italy
| | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY
| | - Rob Jones
- University of Glasgow, The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | | | | | - Shibu Thomas
- Janssen Research and Development, Spring House, PA
| | | | - Dong Shen
- Janssen Research and Development, Spring House, PA
| | | | | | | | - Bertrand Tombal
- Institut de Recherche Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Matti Annala
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada.,Prostate Cancer Research Center, University of Tampere, Tampere, Finland
| | - Kim N Chi
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada.,BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Ugo De Giorgi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Meldola, Italy
| | - Enrique Gonzalez-Billalabeitia
- Hospital Universitario Morales Meseguer, Biobanco Nodo 3, Instituto Murciano de Investigación Biosanitaria-Universidad de Murcia, Murcia, Spain
| | - Alexander W Wyatt
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gerhardt Attard
- University College London Cancer Institute, London, United Kingdom
| |
Collapse
|
114
|
Sieuwerts AM, Onstenk W, Kraan J, Beaufort CM, Van M, De Laere B, Dirix LY, Hamberg P, Beeker A, Meulenbeld HJ, Creemers G, van Weerden WM, Jenster GW, Nieuweboer AJM, Mathijssen RHJ, de Wit R, Martens JWM, Sleijfer S. AR splice variants in circulating tumor cells of patients with castration-resistant prostate cancer: relation with outcome to cabazitaxel. Mol Oncol 2019; 13:1795-1807. [PMID: 31180178 PMCID: PMC6670012 DOI: 10.1002/1878-0261.12529] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 06/01/2019] [Accepted: 06/07/2019] [Indexed: 12/11/2022] Open
Abstract
The androgen receptor splice variant (AR-V) 7 in circulating tumor cells (CTCs) is a predictor for resistance to anti-AR-targeted treatment, but not to taxane-based chemotherapy in metastatic castration-resistant prostate cancer (mCRPC). In this study, we investigated whether the presence of two constitutively active variants (AR-V3, AR-V7) and two other conditionally activated variants (AR-V1, AR-V9) vs full-length androgen receptor (AR-FL) measured in CTCs from patients with mCRPC were associated with outcome to therapy with the taxane cabazitaxel. Blood was collected at baseline and after two cycles of cabazitaxel from 118 mCRPC patients starting cabazitaxel in a prospective phase II trial. CellSearch-enriched CTCs were enumerated and in parallel characterized for the presence of the AR-Vs by reverse transcription quantitative polymerase chain reaction. Correlations with CTC and prostate-specific antigen response to cabazitaxel as well as associations with overall survival (OS) were investigated. All AR-Vs were frequently present and co-expressed at frequencies of 31-48% at baseline and at 19-40% after two cycles of cabazitaxel. No specific directions of change in the measured variants were detected between the start of treatment and after two cycles of cabazitaxel. No associations between the presence of AR-V3 and AR-V7 and outcome to cabazitaxel were observed. While a reduction in CTCs to < 5 CTCs during treatment (CTC5-response) was less often observed in patients with AR-V9-positive CTCs at baseline (P = 0.004), the CTC5-adjusted detection of AR-V1 after two cycles of cabazitaxel was an independent prognostic factor for OS [HR 2.4 (95% CI 1.1-5.1, P = 0.03)]. These novel findings are expected to contribute to more personalized treatment approaches in mCRPC patients.
Collapse
Affiliation(s)
- Anieta M. Sieuwerts
- Department of Medical OncologyErasmus MC Cancer InstituteRotterdamThe Netherlands
- Department of Medical OncologyCancer Genomics NetherlandsRotterdamThe Netherlands
| | - Wendy Onstenk
- Department of Medical OncologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Jaco Kraan
- Department of Medical OncologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Corine M. Beaufort
- Department of Medical OncologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Mai Van
- Department of Medical OncologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Bram De Laere
- GZA Hospitals Sint‐AugustinusWilrijkBelgium
- Center for Oncological ResearchUniversity of AntwerpAntwerpBelgium
| | - Luc Y. Dirix
- GZA Hospitals Sint‐AugustinusWilrijkBelgium
- Center for Oncological ResearchUniversity of AntwerpAntwerpBelgium
| | - Paul Hamberg
- Department of Internal MedicineFranciscus Gasthuis and VlietlandRotterdamThe Netherlands
| | - Aart Beeker
- Department of Internal MedicineSpaarne GasthuisHoofddorpThe Netherlands
| | | | - Geert‐Jan Creemers
- Department of Internal MedicineCatharina ZiekenhuisEindhovenThe Netherlands
| | | | | | | | - Ron H. J. Mathijssen
- Department of Medical OncologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Ronald de Wit
- Department of Medical OncologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - John W. M. Martens
- Department of Medical OncologyErasmus MC Cancer InstituteRotterdamThe Netherlands
- Department of Medical OncologyCancer Genomics NetherlandsRotterdamThe Netherlands
| | - Stefan Sleijfer
- Department of Medical OncologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| |
Collapse
|
115
|
Testa U, Castelli G, Pelosi E. Cellular and Molecular Mechanisms Underlying Prostate Cancer Development: Therapeutic Implications. MEDICINES (BASEL, SWITZERLAND) 2019; 6:E82. [PMID: 31366128 PMCID: PMC6789661 DOI: 10.3390/medicines6030082] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/19/2019] [Accepted: 07/25/2019] [Indexed: 12/15/2022]
Abstract
Prostate cancer is the most frequent nonskin cancer and second most common cause of cancer-related deaths in man. Prostate cancer is a clinically heterogeneous disease with many patients exhibiting an aggressive disease with progression, metastasis, and other patients showing an indolent disease with low tendency to progression. Three stages of development of human prostate tumors have been identified: intraepithelial neoplasia, adenocarcinoma androgen-dependent, and adenocarcinoma androgen-independent or castration-resistant. Advances in molecular technologies have provided a very rapid progress in our understanding of the genomic events responsible for the initial development and progression of prostate cancer. These studies have shown that prostate cancer genome displays a relatively low mutation rate compared with other cancers and few chromosomal loss or gains. The ensemble of these molecular studies has led to suggest the existence of two main molecular groups of prostate cancers: one characterized by the presence of ERG rearrangements (~50% of prostate cancers harbor recurrent gene fusions involving ETS transcription factors, fusing the 5' untranslated region of the androgen-regulated gene TMPRSS2 to nearly the coding sequence of the ETS family transcription factor ERG) and features of chemoplexy (complex gene rearrangements developing from a coordinated and simultaneous molecular event), and a second one characterized by the absence of ERG rearrangements and by the frequent mutations in the E3 ubiquitin ligase adapter SPOP and/or deletion of CDH1, a chromatin remodeling factor, and interchromosomal rearrangements and SPOP mutations are early events during prostate cancer development. During disease progression, genomic and epigenomic abnormalities accrued and converged on prostate cancer pathways, leading to a highly heterogeneous transcriptomic landscape, characterized by a hyperactive androgen receptor signaling axis.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
116
|
De Laere B, Ost P, Grönberg H, Lindberg J. Has the PROPHECY of AR-V7 Been Fulfilled? J Clin Oncol 2019; 37:2181-2182. [PMID: 31265360 DOI: 10.1200/jco.19.01015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Bram De Laere
- Bram De Laere, PhD, Karolinska Institutet, Stockholm, Sweden; Piet Ost, MD, PhD, Ghent University Hospital, Ghent, Belgium; and Henrik Grönberg, MD, PhD and Johan Lindberg, PhD, Karolinska Institutet, Stockholm, Sweden
| | - Piet Ost
- Bram De Laere, PhD, Karolinska Institutet, Stockholm, Sweden; Piet Ost, MD, PhD, Ghent University Hospital, Ghent, Belgium; and Henrik Grönberg, MD, PhD and Johan Lindberg, PhD, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Grönberg
- Bram De Laere, PhD, Karolinska Institutet, Stockholm, Sweden; Piet Ost, MD, PhD, Ghent University Hospital, Ghent, Belgium; and Henrik Grönberg, MD, PhD and Johan Lindberg, PhD, Karolinska Institutet, Stockholm, Sweden
| | - Johan Lindberg
- Bram De Laere, PhD, Karolinska Institutet, Stockholm, Sweden; Piet Ost, MD, PhD, Ghent University Hospital, Ghent, Belgium; and Henrik Grönberg, MD, PhD and Johan Lindberg, PhD, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
117
|
Armstrong AJ, Halabi S, Luo J, Nanus DM, Scher HI, Antonarakis ES, George DJ. Reply to L. Dirix, B. De Laere et al, and A. Sharp et al. J Clin Oncol 2019; 37:2184-2186. [PMID: 31265357 DOI: 10.1200/jco.19.01230] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Andrew J Armstrong
- Andrew J. Armstrong, MD, ScM and Susan Halabi, PhD, Duke University, Durham, NC; Jun Luo, PhD, Johns Hopkins University, Baltimore, MD; David M. Nanus, MD, Weill Cornell Medical College, New York, NY; Howard I. Scher, MD, Memorial Sloan Kettering Cancer Center, New York, NY; Emmanuel S. Antonarakis, MD, Johns Hopkins University, Baltimore, MD; and Daniel J. George, MD, Duke University, Durham, NC
| | - Susan Halabi
- Andrew J. Armstrong, MD, ScM and Susan Halabi, PhD, Duke University, Durham, NC; Jun Luo, PhD, Johns Hopkins University, Baltimore, MD; David M. Nanus, MD, Weill Cornell Medical College, New York, NY; Howard I. Scher, MD, Memorial Sloan Kettering Cancer Center, New York, NY; Emmanuel S. Antonarakis, MD, Johns Hopkins University, Baltimore, MD; and Daniel J. George, MD, Duke University, Durham, NC
| | - Jun Luo
- Andrew J. Armstrong, MD, ScM and Susan Halabi, PhD, Duke University, Durham, NC; Jun Luo, PhD, Johns Hopkins University, Baltimore, MD; David M. Nanus, MD, Weill Cornell Medical College, New York, NY; Howard I. Scher, MD, Memorial Sloan Kettering Cancer Center, New York, NY; Emmanuel S. Antonarakis, MD, Johns Hopkins University, Baltimore, MD; and Daniel J. George, MD, Duke University, Durham, NC
| | - David M Nanus
- Andrew J. Armstrong, MD, ScM and Susan Halabi, PhD, Duke University, Durham, NC; Jun Luo, PhD, Johns Hopkins University, Baltimore, MD; David M. Nanus, MD, Weill Cornell Medical College, New York, NY; Howard I. Scher, MD, Memorial Sloan Kettering Cancer Center, New York, NY; Emmanuel S. Antonarakis, MD, Johns Hopkins University, Baltimore, MD; and Daniel J. George, MD, Duke University, Durham, NC
| | - Howard I Scher
- Andrew J. Armstrong, MD, ScM and Susan Halabi, PhD, Duke University, Durham, NC; Jun Luo, PhD, Johns Hopkins University, Baltimore, MD; David M. Nanus, MD, Weill Cornell Medical College, New York, NY; Howard I. Scher, MD, Memorial Sloan Kettering Cancer Center, New York, NY; Emmanuel S. Antonarakis, MD, Johns Hopkins University, Baltimore, MD; and Daniel J. George, MD, Duke University, Durham, NC
| | - Emmanuel S Antonarakis
- Andrew J. Armstrong, MD, ScM and Susan Halabi, PhD, Duke University, Durham, NC; Jun Luo, PhD, Johns Hopkins University, Baltimore, MD; David M. Nanus, MD, Weill Cornell Medical College, New York, NY; Howard I. Scher, MD, Memorial Sloan Kettering Cancer Center, New York, NY; Emmanuel S. Antonarakis, MD, Johns Hopkins University, Baltimore, MD; and Daniel J. George, MD, Duke University, Durham, NC
| | - Daniel J George
- Andrew J. Armstrong, MD, ScM and Susan Halabi, PhD, Duke University, Durham, NC; Jun Luo, PhD, Johns Hopkins University, Baltimore, MD; David M. Nanus, MD, Weill Cornell Medical College, New York, NY; Howard I. Scher, MD, Memorial Sloan Kettering Cancer Center, New York, NY; Emmanuel S. Antonarakis, MD, Johns Hopkins University, Baltimore, MD; and Daniel J. George, MD, Duke University, Durham, NC
| |
Collapse
|
118
|
De Laere B, Rajan P, Grönberg H, Dirix L, Lindberg J. Androgen Receptor Burden and Poor Response to Abiraterone or Enzalutamide in TP53 Wild-Type Metastatic Castration-Resistant Prostate Cancer. JAMA Oncol 2019; 5:1060-1062. [PMID: 31046065 PMCID: PMC6499126 DOI: 10.1001/jamaoncol.2019.0869] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/01/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Bram De Laere
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Center for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
| | - Prabhakar Rajan
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Luc Dirix
- Center for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
- Department of Oncology, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
119
|
Agnoletto C, Corrà F, Minotti L, Baldassari F, Crudele F, Cook WJJ, Di Leva G, d'Adamo AP, Gasparini P, Volinia S. Heterogeneity in Circulating Tumor Cells: The Relevance of the Stem-Cell Subset. Cancers (Basel) 2019; 11:cancers11040483. [PMID: 30959764 PMCID: PMC6521045 DOI: 10.3390/cancers11040483] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/16/2019] [Accepted: 03/30/2019] [Indexed: 12/20/2022] Open
Abstract
The release of circulating tumor cells (CTCs) into vasculature is an early event in the metastatic process. The analysis of CTCs in patients has recently received widespread attention because of its clinical implications, particularly for precision medicine. Accumulated evidence documents a large heterogeneity in CTCs across patients. Currently, the most accepted view is that tumor cells with an intermediate phenotype between epithelial and mesenchymal have the highest plasticity. Indeed, the existence of a meta-stable or partial epithelial–mesenchymal transition (EMT) cell state, with both epithelial and mesenchymal features, can be easily reconciled with the concept of a highly plastic stem-like state. A close connection between EMT and cancer stem cells (CSC) traits, with enhanced metastatic competence and drug resistance, has also been described. Accordingly, a subset of CTCs consisting of CSC, present a stemness profile, are able to survive chemotherapy, and generate metastases after xenotransplantation in immunodeficient mice. In the present review, we discuss the current evidence connecting CTCs, EMT, and stemness. An improved understanding of the CTC/EMT/CSC connections may uncover novel therapeutic targets, irrespective of the tumor type, since most cancers seem to harbor a pool of CSCs, and disclose important mechanisms underlying tumorigenicity.
Collapse
Affiliation(s)
- Chiara Agnoletto
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Fabio Corrà
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Linda Minotti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Federica Baldassari
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Francesca Crudele
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | | | - Gianpiero Di Leva
- School of Environment and Life Sciences, University of Salford, Salford M5 4WT, UK.
| | - Adamo Pio d'Adamo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Paolo Gasparini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Stefano Volinia
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
120
|
Vandekerkhove G, Struss WJ, Annala M, Kallio HML, Khalaf D, Warner EW, Herberts C, Ritch E, Beja K, Loktionova Y, Hurtado-Coll A, Fazli L, So A, Black PC, Nykter M, Tammela T, Chi KN, Gleave ME, Wyatt AW. Circulating Tumor DNA Abundance and Potential Utility in De Novo Metastatic Prostate Cancer. Eur Urol 2019; 75:667-675. [PMID: 30638634 DOI: 10.1016/j.eururo.2018.12.042] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Several systemic therapeutic options exist for metastatic castrate-sensitive prostate cancer (mCSPC). Circulating tumor DNA (ctDNA) can molecularly profile metastatic castration-resistant prostate cancer and can influence decision-making, but remains untested in mCSPC. OBJECTIVE To determine ctDNA abundance at de novo mCSPC diagnosis and whether ctDNA provides complementary clinically relevant information to a prostate biopsy. DESIGN, SETTING, AND PARTICIPANTS We collected plasma cell-free DNA (cfDNA) from 53 patients newly diagnosed with mCSPC and, where possible, during treatment. Targeted sequencing was performed on cfDNA and DNA from diagnostic prostate tissue. RESULTS AND LIMITATIONS The median ctDNA fraction was 11% (range 0-84%) among untreated patients but was lower (1.0%, range 0-51%) among patients after short-term (median 22d) androgen deprivation therapy (ADT). TP53 mutations and DNA repair defects were identified in 47% and 21% of the cohort, respectively. The concordance for mutation detection in matched samples was 80%. Combined ctDNA and tissue analysis identified potential driver alterations in 94% of patients, whereas ctDNA or prostate biopsy alone was insufficient in 19 cases (36%). Limitations include the use of a narrow gene panel and undersampling of primary disease by prostate biopsy. CONCLUSIONS ctDNA provides additional information to a prostate biopsy in men with de novo mCSPC, but ADT rapidly reduces ctDNA availability. Primary tissue and ctDNA share relevant somatic alterations, suggesting that either is suitable for molecular subtyping in de novo mCSPC. The optimal approach for biomarker development should utilize both a tissue and liquid biopsy at diagnosis, as neither captures clinically relevant somatic alterations in all patients. PATIENT SUMMARY In men with advanced prostate cancer, tumor DNA shed into the bloodstream can be measured via a blood test. The information from this test provides complementary information to a prostate needle biopsy and could be used to guide management strategies. Sequencing data were deposited in the European Genome-phenome Archive (EGA) under study identifier EGAS00001003351.
Collapse
Affiliation(s)
- Gillian Vandekerkhove
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Werner J Struss
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Matti Annala
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Heini M L Kallio
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Daniel Khalaf
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - Evan W Warner
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Cameron Herberts
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Elie Ritch
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Kevin Beja
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Yulia Loktionova
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Antonio Hurtado-Coll
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Ladan Fazli
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Alan So
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Peter C Black
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Teuvo Tammela
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Kim N Chi
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada; Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - Martin E Gleave
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Alexander W Wyatt
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
121
|
Rebello RJ, Oing C, Gillessen S, Bristow RG. TP53 and Prognosis in mCRPC Survival: Biology or Coincidence? Clin Cancer Res 2019; 25:1699-1701. [DOI: 10.1158/1078-0432.ccr-18-3401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/27/2018] [Accepted: 01/02/2019] [Indexed: 11/16/2022]
|
122
|
Abstract
Over the last decade, advancements in massively-parallel DNA sequencing and computational biology have allowed for unprecedented insights into the fundamental mutational processes that underlie virtually every major cancer type. Two major cancer genomics consortia-The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC)-have produced rich databases of mutational, pathological, and clinical data that can be mined through web-based portals, allowing for correlative studies and testing of novel hypotheses on well-powered patient cohorts.In this chapter, we will review the impact of these technological developments on the understanding of molecular subtypes that promote prostate cancer initiation, progression, metastasis, and clinical aggression. In particular, we will focus on molecular subtypes that define clinically-relevant patient cohorts and assess how a better understanding of how these subtypes-in both somatic and germline genomes-may influence the clinical course for individual men diagnosed with prostate cancer.
Collapse
|
123
|
Wu A, Attard G. Plasma DNA Analysis in Prostate Cancer: Opportunities for Improving Clinical Management. Clin Chem 2019; 65:100-107. [PMID: 30538124 DOI: 10.1373/clinchem.2018.287250] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/05/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND Molecular characterization of tumors could be important for clinical management. Plasma DNA obtained noninvasively as a liquid biopsy could be widely applicable for clinical implementation in biomarker-based treatment strategies. CONTENT Prostate cancer is a disease with variable clinical outcomes and molecular features and therefore presents multiple opportunities for biomarker-based treatment optimization. Tissue analysis may not be representative of the lethal clone in localized disease or of intrapatient, intermetastases heterogeneity; fresh tissue is often challenging to obtain by biopsy of metastasis, whereas archival samples may not represent current disease and may be of insufficient quality. Plasma DNA is of variable tumor-to-normal fraction that requires accurate estimation using sensitively measured genomic events. In plasma with sufficient tumor content, the spectrum of genomic aberrations closely resembles tissue and could be used to molecularly characterize patients in real time. In this review we discuss the opportunities for improving clinical management by using plasma DNA analysis in different clinical scenarios across the disease spectrum, from detection of prostate cancer and disease relapse to treatment response prediction, response assessment, and interrogation of treatment resistance in metastatic prostate cancer. Combinational strategies may incorporate other modalities, including circulating tumor cells, circulating microRNA, and extracellular vesicles analysis, which could help to achieve more accurate characterization. SUMMARY There are many opportunities for plasma DNA analyses to change clinical management. However, there are challenges that need to be addressed to clinically implement a test, including the development of accurate, fit for purpose, and technically reproducible assay, followed by prospective validation in a large cohort of patients.
Collapse
Affiliation(s)
- Anjui Wu
- University College London Cancer Institute, London, UK
- The Institute of Cancer Research, London, UK
| | | |
Collapse
|
124
|
Storbeck KH, Mostaghel EA. Canonical and Noncanonical Androgen Metabolism and Activity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:239-277. [PMID: 31900912 DOI: 10.1007/978-3-030-32656-2_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Androgens are critical drivers of prostate cancer. In this chapter we first discuss the canonical pathways of androgen metabolism and their alterations in prostate cancer progression, including the classical, backdoor and 5α-dione pathways, the role of pre-receptor DHT metabolism, and recent findings on oncogenic splicing of steroidogenic enzymes. Next, we discuss the activity and metabolism of non-canonical 11-oxygenated androgens that can activate wild-type AR and are less susceptible to glucuronidation and inactivation than the canonical androgens, thereby serving as an under-recognized reservoir of active ligands. We then discuss an emerging literature on the potential non-canonical role of androgen metabolizing enzymes in driving prostate cancer. We conclude by discussing the potential implications of these findings for prostate cancer progression, particularly in context of new agents such as abiraterone and enzalutamide, which target the AR-axis for prostate cancer therapy, including mechanisms of response and resistance and implications of these findings for future therapy.
Collapse
Affiliation(s)
- Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Elahe A Mostaghel
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA. .,Department of Medicine, University of Washington, Seattle, WA, USA. .,Geriatric Research, Education and Clinical Center S-182, VA Puget Sound Health Care System, Seattle, WA, USA.
| |
Collapse
|
125
|
Mayrhofer M, De Laere B, Whitington T, Van Oyen P, Ghysel C, Ampe J, Ost P, Demey W, Hoekx L, Schrijvers D, Brouwers B, Lybaert W, Everaert E, De Maeseneer D, Strijbos M, Bols A, Fransis K, Oeyen S, van Dam PJ, Van den Eynden G, Rutten A, Aly M, Nordström T, Van Laere S, Rantalainen M, Rajan P, Egevad L, Ullén A, Yachnin J, Dirix L, Grönberg H, Lindberg J. Cell-free DNA profiling of metastatic prostate cancer reveals microsatellite instability, structural rearrangements and clonal hematopoiesis. Genome Med 2018; 10:85. [PMID: 30458854 PMCID: PMC6247769 DOI: 10.1186/s13073-018-0595-5] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 11/05/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND There are multiple existing and emerging therapeutic avenues for metastatic prostate cancer, with a common denominator, which is the need for predictive biomarkers. Circulating tumor DNA (ctDNA) has the potential to cost-efficiently accelerate precision medicine trials to improve clinical efficacy and diminish costs and toxicity. However, comprehensive ctDNA profiling in metastatic prostate cancer to date has been limited. METHODS A combination of targeted and low-pass whole genome sequencing was performed on plasma cell-free DNA and matched white blood cell germline DNA in 364 blood samples from 217 metastatic prostate cancer patients. RESULTS ctDNA was detected in 85.9% of baseline samples, correlated to line of therapy and was mirrored by circulating tumor cell enumeration of synchronous blood samples. Comprehensive profiling of the androgen receptor (AR) revealed a continuous increase in the fraction of patients with intra-AR structural variation, from 15.4% during first-line metastatic castration-resistant prostate cancer therapy to 45.2% in fourth line, indicating a continuous evolution of AR during the course of the disease. Patients displayed frequent alterations in DNA repair deficiency genes (18.0%). Additionally, the microsatellite instability phenotype was identified in 3.81% of eligible samples (≥ 0.1 ctDNA fraction). Sequencing of non-repetitive intronic and exonic regions of PTEN, RB1, and TP53 detected biallelic inactivation in 47.5%, 20.3%, and 44.1% of samples with ≥ 0.2 ctDNA fraction, respectively. Only one patient carried a clonal high-impact variant without a detectable second hit. Intronic high-impact structural variation was twice as common as exonic mutations in PTEN and RB1. Finally, 14.6% of patients presented false positive variants due to clonal hematopoiesis, commonly ignored in commercially available assays. CONCLUSIONS ctDNA profiles appear to mirror the genomic landscape of metastatic prostate cancer tissue and may cost-efficiently provide somatic information in clinical trials designed to identify predictive biomarkers. However, intronic sequencing of the interrogated tumor suppressors challenges the ubiquitous focus on coding regions and is vital, together with profiling of synchronous white blood cells, to minimize erroneous assignments which in turn may confound results and impede true associations in clinical trials.
Collapse
Affiliation(s)
- Markus Mayrhofer
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Bram De Laere
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Centre for Oncological Research, University of Antwerp, Antwerp, Belgium
| | - Tom Whitington
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Jozef Ampe
- Department of Urology, AZ Sint-Jan, Brugge, Belgium
| | - Piet Ost
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Wim Demey
- Department of Oncology, AZ KLINA, Brasschaat, Belgium
| | - Lucien Hoekx
- Department of Urology, Antwerp University Hospital, Antwerp, Belgium
| | | | | | - Willem Lybaert
- Department of Oncology, AZ Nikolaas, Sint-Niklaas, Belgium
| | - Els Everaert
- Department of Oncology, AZ Nikolaas, Sint-Niklaas, Belgium
| | | | | | - Alain Bols
- Department of Oncology, AZ Sint-Jan, Brugge, Belgium
| | - Karen Fransis
- Department of Urology, Antwerp University Hospital, Antwerp, Belgium
| | - Steffi Oeyen
- Centre for Oncological Research, University of Antwerp, Antwerp, Belgium
| | - Pieter-Jan van Dam
- Centre for Oncological Research, University of Antwerp, Antwerp, Belgium
| | | | - Annemie Rutten
- Department of Oncology, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Markus Aly
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Tobias Nordström
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Steven Van Laere
- Centre for Oncological Research, University of Antwerp, Antwerp, Belgium
| | - Mattias Rantalainen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Prabhakar Rajan
- Centre for Molecular Oncology, Barts Cancer Institute, Cancer Research UK Barts Centre, Queen Mary University of London, London, UK
| | - Lars Egevad
- Department of Oncology-Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Anders Ullén
- Department of Oncology-Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Jeffrey Yachnin
- Department of Oncology-Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Luc Dirix
- Centre for Oncological Research, University of Antwerp, Antwerp, Belgium
- Department of Oncology, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|