20
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Genomic correlates of clinical outcome in advanced prostate cancer

      research-article
      a , b , c , d , e , f , g , h , e , h , i , j , b , e , b , b , g , h , h , h , k , l , b , m , n , n , g , k , l , k , l , k , l , k , l , k , l , o , o , g , i , p , q , r , a , a , i , d , k , l , e , g , 7 , b , c , 7 , h , s , 7 , f , t , 7
      Proceedings of the National Academy of Sciences of the United States of America
      National Academy of Sciences
      castration-resistant prostate cancer, integrative genomics, clinical outcomes, biomarkers

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Significance

          The genomic landscape of metastatic castration-resistant prostate cancer (mCRPC) has been well-defined, but the association of genomic findings with patient clinical outcomes and with other characteristics including histology and transcriptional pathway activity remains poorly understood. Here, we describe comprehensive integrative analysis of genomic and transcriptomic profiles, histology, and clinical outcomes for 429 patients with mCRPC. Of all the molecular factors we examined, alterations in RB1 had the strongest association with poor outcome. Our study identifies molecularly defined groups of patients who may benefit from a more aggressive treatment approach, with the genomic and outcome data made available to the research community for further interrogation.

          Abstract

          Heterogeneity in the genomic landscape of metastatic prostate cancer has become apparent through several comprehensive profiling efforts, but little is known about the impact of this heterogeneity on clinical outcome. Here, we report comprehensive genomic and transcriptomic analysis of 429 patients with metastatic castration-resistant prostate cancer (mCRPC) linked with longitudinal clinical outcomes, integrating findings from whole-exome, transcriptome, and histologic analysis. For 128 patients treated with a first-line next-generation androgen receptor signaling inhibitor (ARSI; abiraterone or enzalutamide), we examined the association of 18 recurrent DNA- and RNA-based genomic alterations, including androgen receptor ( AR) variant expression, AR transcriptional output, and neuroendocrine expression signatures, with clinical outcomes. Of these, only RB1 alteration was significantly associated with poor survival, whereas alterations in RB1, AR, and TP53 were associated with shorter time on treatment with an ARSI. This large analysis integrating mCRPC genomics with histology and clinical outcomes identifies RB1 genomic alteration as a potent predictor of poor outcome, and is a community resource for further interrogation of clinical and molecular associations.

          Related collections

          Most cited references18

          • Record: found
          • Abstract: found
          • Article: not found

          Clinical and Genomic Characterization of Treatment-Emergent Small-Cell Neuroendocrine Prostate Cancer: A Multi-institutional Prospective Study

          Purpose The prevalence and features of treatment-emergent small-cell neuroendocrine prostate cancer (t-SCNC) are not well characterized in the era of modern androgen receptor (AR)–targeting therapy. We sought to characterize the clinical and genomic features of t-SCNC in a multi-institutional prospective study. Methods Patients with progressive, metastatic castration-resistant prostate cancer (mCRPC) underwent metastatic tumor biopsy and were followed for survival. Metastatic biopsy specimens underwent independent, blinded pathology review along with RNA/DNA sequencing. Results A total of 202 consecutive patients were enrolled. One hundred forty-eight (73%) had prior disease progression on abiraterone and/or enzalutamide. The biopsy evaluable rate was 79%. The overall incidence of t-SCNC detection was 17%. AR amplification and protein expression were present in 67% and 75%, respectively, of t-SCNC biopsy specimens. t-SCNC was detected at similar proportions in bone, node, and visceral organ biopsy specimens. Genomic alterations in the DNA repair pathway were nearly mutually exclusive with t-SCNC differentiation ( P = .035). Detection of t-SCNC was associated with shortened overall survival among patients with prior AR-targeting therapy for mCRPC (hazard ratio, 2.02; 95% CI, 1.07 to 3.82). Unsupervised hierarchical clustering of the transcriptome identified a small-cell–like cluster that further enriched for adverse survival outcomes (hazard ratio, 3.00; 95% CI, 1.25 to 7.19). A t-SCNC transcriptional signature was developed and validated in multiple external data sets with > 90% accuracy. Multiple transcriptional regulators of t-SCNC were identified, including the pancreatic neuroendocrine marker PDX1 . Conclusion t-SCNC is present in nearly one fifth of patients with mCRPC and is associated with shortened survival. The near-mutual exclusivity with DNA repair alterations suggests t-SCNC may be a distinct subset of mCRPC. Transcriptional profiling facilitates the identification of t-SCNC and novel therapeutic targets.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Androgen Receptor Pathway-Independent Prostate Cancer Is Sustained through FGF Signaling.

            Androgen receptor (AR) signaling is a distinctive feature of prostate carcinoma (PC) and represents the major therapeutic target for treating metastatic prostate cancer (mPC). Though highly effective, AR antagonism can produce tumors that bypass a functional requirement for AR, often through neuroendocrine (NE) transdifferentiation. Through the molecular assessment of mPCs over two decades, we find a phenotypic shift has occurred in mPC with the emergence of an AR-null NE-null phenotype. These "double-negative" PCs are notable for elevated FGF and MAPK pathway activity, which can bypass AR dependence. Pharmacological inhibitors of MAPK or FGFR repressed the growth of double-negative PCs in vitro and in vivo. Our results indicate that FGF/MAPK blockade may be particularly efficacious against mPCs with an AR-null phenotype.
              Bookmark
              • Record: found
              • Abstract: not found
              • Article: not found

              Circulating Tumor DNA Genomics Correlate with Resistance to Abiraterone and Enzalutamide in Prostate Cancer

                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                4 June 2019
                6 May 2019
                6 May 2019
                : 116
                : 23
                : 11428-11436
                Affiliations
                [1] aDepartment of Medicine, Memorial Sloan Kettering Cancer Center , New York, NY 10065;
                [2] bDepartment of Pathology, Weill Medical College of Cornell University , New York, NY 10021;
                [3] cDepartment for Biomedical Research, University of Bern , 3008 Bern, Switzerland;
                [4] dDepartment of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center , New York, NY 10065;
                [5] eDepartment of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy;
                [6] fHuman Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center , New York, NY 10065;
                [7] gFred Hutchinson Cancer Center, University of Washington , Seattle, WA 98109;
                [8] hDepartment of Pathology, University of Michigan , Ann Arbor, MI 48109;
                [9] iDepartment of Medical Oncology, Dana-Farber Cancer Institute, Boston , MA 02215;
                [10] jBroad Institute , Cambridge, MA 02142;
                [11] kInstitute of Cancer Research , London SW7 3RP, United Kingdom;
                [12] lThe Royal Marsden National Health Service Foundation Trust , London SM2 5NG, United Kingdom ;
                [13] mDepartment of Oncologic Pathology, Dana-Farber Cancer Institute , Boston, MA 02215;
                [14] nDepartment of Pathology, Memorial Sloan Kettering Cancer Center , New York, NY 10065;
                [15] oProstate Cancer Clinical Trials Consortium , New York, NY 10065;
                [16] pDepartment of Medicine, Weill Medical College of Cornell University , New York, NY 10021;
                [17] qDepartment of Pathology, Wayne State University School of Medicine , Detroit, MI 48201;
                [18] rDepartment of Oncology, Barbara Ann Karmanos Cancer Institute , Detroit, MI 48201;
                [19] sHoward Hughes Medical Institute, University of Michigan , Ann Arbor, MI 48109;
                [20] tHoward Hughes Medical Institute, Memorial Sloan Kettering Cancer Center , New York, NY 10065
                Author notes

                Contributed by Charles L. Sawyers, March 27, 2019 (sent for review February 19, 2019; reviewed by Samuel Aparicio, John T. Isaacs, and Nandita Mitra)

                Author contributions: W.A., G.H., F.D., P.S.N., M.A.R., A.M.C., and C.L.S. designed research; J.C., N.S., F.D., and M.A.R. performed research; J.M., D.B., S.M., S.C., P.R., R.B.M., H.B., E.I.H., H.I.S., P.W.K., M.-E.T., and J.S.d. contributed new reagents/analytic tools; W.A., G.H., D.P., J.A., I.C., M.C., M.B., D.R., E.M.V.A., A.S., T.F., J.M.M., B.D.R., N.D.S., L.P.K., S.T., Y.M.W., D.N.R., M.L., A.G., V.E.R., C.C.P., J.F., J.V., N.S., F.D., P.S.N., M.A.R., and A.M.C. analyzed data; and W.A., F.D., P.S.N., M.A.R., A.M.C., and C.L.S. wrote the paper.

                Reviewers: S.A., BC Cancer Agency and University of British Columbia; J.T.I., Johns Hopkins Oncology Center; and N.M., University of Pennsylvania.

                1W.A., J.C., and G.H. contributed equally to this work.

                2Present address: Pathology Department, Institut Curie, 75005 Paris, France.

                3Present address: Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, CB4 0WG Cambridge, United Kingdom.

                4Present address: Bioinformatics Unit, Hospital of Prato, 59100 Prato, Italy.

                5Present address: Prostate Cancer Translational Research Group, Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain.

                6M.-E.T., N.S., J.S.d., F.D., P.S.N., M.A.R., A.M.C., and C.L.S. contributed equally to this work.

                Author information
                http://orcid.org/0000-0003-4955-6475
                Article
                201902651
                10.1073/pnas.1902651116
                6561293
                31061129
                6f776f90-f399-4751-a503-68ac7fb62b5b
                Copyright © 2019 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

                History
                Page count
                Pages: 9
                Funding
                Funded by: Prostate Cancer Foundation (PCF) 100000892
                Award ID: SU2C-AACR-DT0712
                Award Recipient : Wassim Abida Award Recipient : Joanna Cyrta Award Recipient : Glenn Heller Award Recipient : Davide Prandi Award Recipient : Joshua Armenia Award Recipient : Ilsa M Coleman Award Recipient : Marcin Cieslik Award Recipient : Matteo Benelli Award Recipient : Daniel R. Robinson Award Recipient : Eliezer M. Van Allen Award Recipient : Andrea Sboner Award Recipient : Tarcisio Fedrizzi Award Recipient : Juan Miguel Mosquera Award Recipient : Brian D. Robinson Award Recipient : Navonil De Sarkar Award Recipient : Lakshmi P Kunju Award Recipient : Scott A Tomlins Award Recipient : Yi Mi Wu Award Recipient : Daniel Nava Rodrigues Award Recipient : Massimo Loda Award Recipient : Anuradha Gopalan Award Recipient : Victor Reuter Award Recipient : Colin C. Pritchard Award Recipient : Joaquin Mateo Award Recipient : Diletta Bianchini Award Recipient : Susana Miranda Award Recipient : Suzanne Carreira Award Recipient : Pasquale Rescigno Award Recipient : Julie Filipenko Award Recipient : Jacob Vinson Award Recipient : R.Bruce Montgomery Award Recipient : Himisha Beltran Award Recipient : Elizabeth I Heath Award Recipient : Howard I Scher Award Recipient : Philip W Kantoff Award Recipient : Mary-Ellen Taplin Award Recipient : Nikolaus Schultz Award Recipient : Johann DeBono Award Recipient : Francesca Demichelis Award Recipient : Peter S Nelson Award Recipient : Mark A. Rubin Award Recipient : Charles L Sawyers
                Funded by: EIF | Stand Up To Cancer (SU2C) 100009730
                Award ID: SU2C-AACR-DT0712
                Award Recipient : Wassim Abida Award Recipient : Joanna Cyrta Award Recipient : Glenn Heller Award Recipient : Davide Prandi Award Recipient : Joshua Armenia Award Recipient : Ilsa M Coleman Award Recipient : Marcin Cieslik Award Recipient : Matteo Benelli Award Recipient : Daniel R. Robinson Award Recipient : Eliezer M. Van Allen Award Recipient : Andrea Sboner Award Recipient : Tarcisio Fedrizzi Award Recipient : Juan Miguel Mosquera Award Recipient : Brian D. Robinson Award Recipient : Navonil De Sarkar Award Recipient : Lakshmi P Kunju Award Recipient : Scott A Tomlins Award Recipient : Yi Mi Wu Award Recipient : Daniel Nava Rodrigues Award Recipient : Massimo Loda Award Recipient : Anuradha Gopalan Award Recipient : Victor Reuter Award Recipient : Colin C. Pritchard Award Recipient : Joaquin Mateo Award Recipient : Diletta Bianchini Award Recipient : Susana Miranda Award Recipient : Suzanne Carreira Award Recipient : Pasquale Rescigno Award Recipient : Julie Filipenko Award Recipient : Jacob Vinson Award Recipient : R.Bruce Montgomery Award Recipient : Himisha Beltran Award Recipient : Elizabeth I Heath Award Recipient : Howard I Scher Award Recipient : Philip W Kantoff Award Recipient : Mary-Ellen Taplin Award Recipient : Nikolaus Schultz Award Recipient : Johann DeBono Award Recipient : Francesca Demichelis Award Recipient : Peter S Nelson Award Recipient : Mark A. Rubin Award Recipient : Charles L Sawyers
                Funded by: Howard Hughes Medical Institute (HHMI) 100000011
                Award ID: none
                Award Recipient : Charles L Sawyers
                Categories
                PNAS Plus
                Biological Sciences
                Medical Sciences
                From the Cover
                PNAS Plus

                castration-resistant prostate cancer,integrative genomics,clinical outcomes,biomarkers

                Comments

                Comment on this article