Shuang (George) Zhao

Shuang (George) Zhao, MD

Assistant Professor

Department of Human Oncology

I am an assistant professor in the Department of Human Oncology. I completed my bachelor’s and master’s degrees in Computer Science & Engineering with a focus on Intelligent Systems at the University of Michigan. I then completed my MD at the University of Michigan Medical School. After a transitional year internship at Beaumont Hospital – Dearborn, I returned to the University of Michigan for residency training in radiation oncology. Under the American Board of Radiology’s Holman Pathway, I led multiple cancer genomics and biomarker studies under the mentorship of Dr. Felix Feng. As a physician-scientist, my clinical focus is on the treatment of patients with GU malignancies. My independent translational research laboratory focuses on using advances in sequencing and computational techniques to develop biomarkers of treatment response and resistance, primarily in prostate cancer. I co-direct the UWCCC Circulating Biomarker Core with Dr. Josh Lang, where we work on translating liquid biopsy technology into the clinic.

Education

Residency, University of Michigan, Radiation Oncology (2020)

MD, University of Michigan, Medicine (2015)

MSE, University of Michigan, Computer Science and Engineering (2010)

BSE, University of Michigan, Computer Science and Engineering (2009)

Academic Appointments

Assistant Professor, Human Oncology (2020)

Co-director, University of Wisconsin Carbone Cancer Center Circulating Biomarker Core (2020)

Selected Honors and Awards

Roentgen Resident/Fellow Research Award (2020)

Radiation Research Society Scholar-in-Training Travel Award (2018)

MRS Resident Conference & Research Forum Radiation Oncology Abstract Award (2017)

ASTRO Resident Clinical/Basic Science Research Award in Biology (2015)

Dean’s Merit Scholarship, University of Michigan Medical School (2010-15)

William L. Everitt Award of Excellence, University of Michigan (2009)

James B. Angell Scholar, University of Michigan (2009)

Engineering Scholarship of Honor, University of Michigan (2005-09)

Pfizer Corporate Merit Scholarship, National Merit Scholarship Corporation (2005-09 )

Michigan Merit Award, State of Michigan (2005)

Regents Merit Scholarship, University of Michigan (2005)

Research Focus

GU malignancies

Biomarkers to personalize treatment in cancer 

In the Zhao Lab, we focus on developing novel clinical biomarkers that predict response and resistance to specific therapies across cancers. These biomarkers can be used to better select which patients derive a benefit from treatment, which patients do not and can be spared potential toxicity, as well as monitor for emergence of resistance. We seek to use advanced computational approaches and liquid biopsy technologies to better personalize the care of cancer patients.

Predictive biomarkers

Treatment decisions for cancer has historically been based on tissue of origin and other pathologic and clinical variables which give prognostic information on the overall level of aggressiveness of a tumor. Predictive biomarkers on the other hand are used to predict response to specific treatments, rather than overall prognosis. Such biomarkers have high clinical utility in deciding the optimal treatment approach for an individual patient. Our laboratory seeks to leverage advances in molecular medicine to identify new predictive biomarkers for standard and emerging cancer therapies.

predictive biomarkers graphs

  • SG Zhao*, SL Chang*, DE Spratt, N Erho, M Yu, HA Ashab, M Alshalalfa, C Speers, SA Tomlins, E Davicioni, AP Dicker, PR Carroll, MR Cooperberg, SJ Freedland, RJ Karnes, A Ross, EM Schaeffer, RB Den, PL Nguyen†, FY Feng†. The development and validation of a 24-gene predictor of response to post-operative radiation therapy in prostate cancer: a matched retrospective analysis. Lancet Oncology. PMID 27743920.
  • SG Zhao*, SL Chang*, N Erho, M Yu, J Lehrer, M Alshalalfa, C Speers, MR Cooperberg, W Kim, CJ Ryan, RB Den, SJ Freedland, E Posadas, H Sandler, EA Klein, P Black, R Seiler, SA Tomlins, AM Chinnaiyan MD, RB Jenkins, E Davicioni, AE Ross, EM Schaeffer, PL Nguyen, PR Carroll, RJ Karnes, DE Spratt†, FY Feng†. Associations of luminal and basal subtyping of prostate cancer with prognosis and response to androgen deprivation therapy. JAMA Oncology. PMID 28494073.
  • SG Zhao, M Yu, DE Spratt, SL Chang, FY Feng, MM Kim, CW Speers, BL Carlson, AC Mladek, TS Lawrence, JN Sarkaria, DR Wahl. Development and validation of xenograft-based platform-independent gene signatures that predict response to alkylating chemotherapy, radiation, and combination therapy in patients with glioblastoma. Neuro-Oncology. PMID 31121035.
  • NM White*, SG Zhao*, J Zhang, EB Rozycki, HX Dang, SD McFadden, AM Eteleeb, M Alshalalfa, IA Vergara, N Erho, JM Arbeitg, RJ Karnes, RB Den, E Davicioni, CA Maher. Multi-institutional analysis shows low PCAT-14 expression associates with poor outcome in prostate cancer. European Urology. PMID 27460352.
  • R Aggarwal*, NR Rydzewski*, L Zhang, A Foye, W Kim, KT Helzer, H Bakhtiar, SL Chang, MD Perry, M Gleave, RE Reiter, J Huang, CP Evans, JJ Alumkal, JM Lang, M Yu, DA Quigley, M Sjöström, EJ Small, FY Feng, SG Zhao. Prognosis associated with luminal and basal subtypes of metastatic prostate cancer. JAMA Oncology. PMID 34554200.

Clinical next-generation sequencing

Integrated multi-omic next-generation sequencing (NGS) on clinical samples allows for a global view of the landscape of molecular alterations that drive cancer progression and treatment resistance. Our lab analyzes correlative sequencing data from clinical cohorts and trials to better understand the molecular underpinnings of the phenotypic behavior of tumors.

  • SG Zhao*, W Chen*, H Li*, A Foye, M Zhang, M Sjöstrjöm, R Aggarwal, D Playdle, A Liao, JJ Alumkal, R Das, J Chou, JT Hua, TJ Barnard, AM Bailey, E Chow, M Perry, HX Dang, R Yang, R Moussavi-Baygi, L Zhang, M Alshalalfa, SL Chang, KE Houlahan, YJ Shiah, TM Beer, G Thomas, KN Chi, M Gleave, A Zoubeidi, RE Reiter, MB Rettig, O Witte, Y Kim, L Fong, DE Spratt, TM Morgan, R Bose, FW Huang, H Li, L Chesner, T Shenoy, H Goodazi, IA Asangani, S Sandhu, JM Lang, PN Lara, N Mahajan, CP Evans, P Febbo, S Batzoglou, KE Knudsen, HH He, J Huang, W Zwart, JF Costello, J Luo, SA Tomlins, AW Wyatt, SM Dehm, A Ashworth, LA Gilbert, P Boutros, K Farh, AM Chinnaiyan†, CA Maher†, EJ Small†, DA Quigley†, FY Feng†. Hallmarks of epigenomic regulation in advanced prostate cancer. Nature Genetics. PMID 32661416.
  • DA Quigley*, HX Dang*, SG Zhao*, P Lloyd, R Aggarwal, JJ Alumkal, A Foye, V Kothari, M Perry, AM Bailey, D Playdle, TJ Barnard, L Zhang, J Zhang, JF Youngren, MP Cieslik, A Parolia, TM Beer, G Thomas, KN Chi, M Gleave, N Lack, A Zoubeidi, RE Reiter, M Rettig, O Witte, CJ Ryan, L Fong, W Kim, T Friedlander, J Chou, H Li, R Das, H Li, R Moussavi-Baygi, H Goodarzi, LA Gilbert, P Lara, CP Evans, TC Goldstein, JM Stuart, SA Tomlins, DE Spratt, RK Cheetham, DT Cheng, K Farh, JS Gehring, J Hakenberg, A Liao, P Febbo, J Shon, S Batzoglou, KE Knudsen, HH He, J Huang, AW Wyatt, SM Dehm, A Ashworth, AM Chinnaiyan†, CA Maher†, EJ Small†, FY Feng†. Genomic hallmarks and structural variation in metastatic prostate cancer. Cell. PMID 30033370.
  • SG Zhao*, WS Chen*, R Das, SL Chang, SA Tomlins, J Chou, DA Quigley, H Dang, T Barnard, BA Mahal, EA Gibb, Y Liu, E Davicioni, LR Duska, E Posadas, S Jolly, DE Spratt, PL Nguyen, CA Maher, EJ Small, FY Feng. Clinical and genomic implications of luminal and basal subtypes across carcinomas. Clinical Cancer Research. PMID 30573691.
  • E Feng*, NR Rydzewski*, M Zhang, A Lundberg, M Bootsma, KT Helzer, JM Lang, R Aggarwal, EJ Small, DA Quigley, M Sjöström, SG Zhao. Intrinsic molecular subtypes of metastatic castration-resistant prostate cancer. Clinical Cancer Research. PMID 36260524.

Computational approaches

High-throughput sequencing techniques can generate an enormous amount of data. Machine-learning is the branch of computer science specifically focused on identifying important patterns in that are too complex for humans to independently explore. Our laboratory seeks to leverage machine learning and other advanced computational approaches to develop models for clinical and biological endpoints.

  • SG Zhao, J Lehrer, SL Chang, R Das, N Erho, Y Liu, M Sjöström, RB Den, SJ Freedland, EA Klein, RJ Karnes, EM Schaeffer, M Xu, C Speers, PL Nguyen, AE Ross, JM Chan, MR Cooperberg, PR Carroll, E Davicioni, L Fong, DE Spratt, FY Feng. The immune landscape of prostate cancer and nomination of PD-L2 as a potential therapeutic target. J Natl Cancer Inst. PMID 30321406.
  • SG Zhao*, JR Evans*, V Kothari, G Sun, A Larm, V Mondine, EM Schaeffer, AE Ross, EA Klein, RB Den, AP Dicker, RJ Karnes, N Erho, PL Nguyen, E Davicioni, FY Feng. The landscape of prognostic outlier genes in high-risk prostate cancer. Clinical Cancer Research. PMID 26631616.
  • JR Evans*, SG Zhao*, SL Chang, SA Tomlins, N Erho, A Sboner, MJ Schiewer, DE Spratt, V Kothari, EA Klein, RB Den, AP Dicker, RJ Karnes, X Yu, PL Nguyen, MA Rubin, J de Bono, KE Knudsen, E Davicioni, FY Feng. Patient-level DNA damage and repair pathway profiles and prognosis after prostatectomy for High-Risk Prostate Cancer. JAMA Oncology. PMID 26746117.
  • C Speers*, S Zhao*, H Bartelink, M Liu, LJ Pierce†, FY Feng†. Development and validation of a novel radiosensitivity signature in human breast cancer. Clinical Cancer Research. PMID 25904749.
  • NR Rydzewski, E Peterson, JM Lang, M Yu, SL Chang, M Sjöström, H Bakhtiar, G Song, KT Helzer, ML Bootsma, WS Chen, RM Shrestha, M Zhang, DA Quigley, R Aggarwal, EJ Small, DR Wahl, FY Feng†, SG Zhao†. Predicting Cancer Drug TARGETS – TreAtment Response Generalized Elastic-neT Signatures. NPJ Genom Med. PMID 34548481.
  • H Bakhtiar*, KT Helzer*, Y Park, Y Chen, NR Rydzewski, ML Bootsma, Y Shi, PM Harari, M Sharifi, M Sjöström, JM Lang, M Yu, SG Zhao. Identification of phenocopies improves prediction of targeted therapy response over DNA mutations alone. NPJ Genom Med. PMID 36253482.

Liquid Biomarkers

Obtaining tissue specimens from cancer patients for molecular analysis is logistically challenging to perform at scale, especially in metastatic cancer. In collaboration with the UWCCC Circulating Biomarker Core, we work on developing liquid biopsy approaches for circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and other non-invasively assays.

Figure showing data results.

  • SG Zhao*, JM Sperger*, JL Schehr*, RR McKay, H Emamekhoo, A Singh, ZD Schultz, RM Bade, CN Stahlfeld, CS Gilsdorf, CI Hernandez, SK Wolfe, RD Mayberry, HM Krause, M Bootsma, KT Helzer, N Rydzewski, H Bakhtiar, Y Shi, G Blitzer, CE Kyriakopoulos, D Kosoff, XX Wei, J Floberg, N Sethakorn, M Sharifi, PM Harari, W Huang, H Beltran, TK Choueiri, HI Scher, DE Rathkopf, S Halabi, AJ Armstrong, DJ Beebe, M Yu, KE Sundling, ME Taplin, JM Lang. A clinical-grade liquid biomarker to detect neuroendocrine differentiation in prostate cancer. J Clin Invest. PMID 36317634.
  • JR Prensner*, S Zhao*, N Erho, M Schipper, MK Iyer, SM Dhanasekaran, C Magi-Galluzzi, R Mehra, A Sahu, J Siddiqui, E Davicioni, RB Den, AP Dicker, RJ Karnes, JT Wei, EA Klein, RB Jenkins, AM Chinnaiyan, FY Feng. RNA biomarkers associated with metastatic progression in prostate cancer: a multi-institutional high-throughput analysis of SChLAP1. Lancet Oncology. PMID 25456366.
  • M Sjöström*, SG Zhao*, S Levy*, M Zhang*, Y Ning, R Shrestha, A Lundberg, C Herberts, A Foye, R Aggarwal, JT Hua, H Li, A Bergamaschi, C Maurice-Dror, A Maheshwari, S Chen, SWS Ng, W Ye, J Petricca, M Fraser, L Chesner, MD Perry, T Moreno-Rodriguez, WS Chen, JJ Alumkal, J Chou, AK Morgans, TM Beer, GV Thomas, M Gleave, P Lloyd, T Phillips, E McCarthy, MC Haffner, A Zoubeidi, M Annala, RE Reiter, MB Rettig, O Witte, L Fong, R Bose, FW Huang, J Luo, A Bjartell, JM Lang, NP Mahajan, PN Lara, CP Evans, PT Tran, EM Posadas, C He, X Cui, J Huang, W Zwart, LA Gilbert, CA Maher, PC Boutros, KN Chi, A Ashworth, EJ Small, HH He, AW Wyatt, DA Quigley, FY Feng. The 5-hydroxymethylcytosine landscape of prostate cancer. Cancer Research. PMID 36251389.
  • JM Sperger*, H Emamekhoo*, RR McKay*, CN Stahlfeld, A Singh, XE Chen, L Kwak, CS Gilsdorf, SK Wolfe, XX Wei, R Silver, Z Zhang, MJ Morris, G Bubley, FY Feng, HI Scher, D Rathkopf, SM Dehm, TK Choueiri, S Halabi, AJ Armstrong, AW Wyatt, ME Taplin, SG Zhao, JM Lang. Prospective evaluation of clinical outcomes utilizing a multi-plex liquid biopsy targeting diverse resistance mechanisms in metastatic prostate cancer. J Clin Oncol. PMID 34197212.
  • M Bootsma*, RR McKay*, H Emamekhoo*, RM Bade, JL Schehr, MC Mannino, A Singh, SK Wolfe, ZD Schultz, J Sperger, W Xie, S Signoretti, CE Kyriakopoulos, D Kosoff, EJ Abel, KT Helzer, N Rydzewski, H Bakhtiar, Y Shi, G Blitzer, M Bassetti, J Floberg, M Yu, N Sethakorn, M Sharifi, PM Harari, TK Choueiri, JM Lang, SG Zhao. Longitudinal molecular profiling of circulating tumor cells in metastatic renal cell carcinoma. J Clin Oncol. PMID 35617646.
  • KT Helzer*, M Sharifi*, JM Sperger*, Y Shi, M Annala, ML Bootsma, SR Reese, A Taylor, KR Kaufmann, H Krause, J Schehr, N Sethakorn, D Kosoff, C Kyriakopoulos, M Burkard, NR Rydzewski, M Yu, PM Harari, M Bassetti, G Blitzer, J Floberg, M Sjöström, DA Quigley, S Dehm, AJ Armstrong, H Beltran, RR McKay, FY Feng, R O’Regan, K Wisinski, H Emamekhoo, AW Wyatt, JM Lang†, SG Zhao†. Fragmentomic analysis of circulating tumor DNA targeted cancer panels. Annals of Oncology. PMID 37330052.

*Co-first authorship, †Co-senior authorship

Funding

  • Prostate Cancer Foundation Young Investigator Award (Principal Investigator)
  • DoD Prostate Cancer Research Program Translational Science Award (Partnering Principal Investigator)
  • DoD Prostate Cancer Research Program Physician Research Award (Principal Investigator)
  • DoD Prostate Cancer Research Program Idea Award (Principal Investigator)
  • NIH DP2 Director’s New Innovator Award (Principal Investigator)
  • NCCN Young Investigator Award (Principal Investigator)
  • NIH Prostate Cancer SPORE (Biostatistics & Bioinformatics Core Co-director)
  • The role of ctDNA in endometrial cancer: A tool for risk stratification and disease monitoring Gynecologic oncology
    Blitzer GC, Zhao SG, Bradley KA, Hartenbach EM
    2023 Nov;178:170-171. doi: 10.1016/j.ygyno.2023.08.008. Epub 2023 Aug 28.
  • Co-evolution of <em>AR</em> gene copy number and structural complexity in endocrine therapy resistant prostate cancer NAR cancer
    Zivanovic A, Miller JT, Munro SA, Knutson TP, Li Y, Passow CN, Simonaitis P, Lynch M, Oseth L, Zhao SG, Feng FY, Wikström P, Corey E, Morrissey C, Henzler C, Raphael BJ, Dehm SM
    2023 Aug 24;5(3):zcad045. doi: 10.1093/narcan/zcad045. eCollection 2023 Sep.
    • More

      Androgen receptor (AR) inhibition is standard of care for advanced prostate cancer (PC). However, efficacy is limited by progression to castration-resistant PC (CRPC), usually due to AR re-activation via mechanisms that include AR amplification and structural rearrangement. These two classes of AR alterations often co-occur in CRPC tumors, but it is unclear whether this reflects intercellular or intracellular heterogeneity of AR. Resolving this is important for developing new therapies and predictive biomarkers. Here, we analyzed 41 CRPC tumors and 6 patient-derived xenografts (PDXs) using linked-read DNA-sequencing, and identified 7 tumors that developed complex, multiply-rearranged AR gene structures in conjunction with very high AR copy number. Analysis of PDX models by optical genome mapping and fluorescence in situ hybridization showed that AR residing on extrachromosomal DNA (ecDNA) was an underlying mechanism, and was associated with elevated levels and diversity of AR expression. This study identifies co-evolution of AR gene copy number and structural complexity via ecDNA as a mechanism associated with endocrine therapy resistance.

      PMID:37636316 | PMC:PMC10448862 | DOI:10.1093/narcan/zcad045


      View details for PubMedID 37636316
  • Machine Learning &amp; Molecular Radiation Tumor Biomarkers Seminars in radiation oncology
    Rydzewski NR, Helzer KT, Bootsma M, Shi Y, Bakhtiar H, Sjöström M, Zhao SG
    2023 Jul;33(3):243-251. doi: 10.1016/j.semradonc.2023.03.002.
    • More

      Developing radiation tumor biomarkers that can guide personalized radiotherapy clinical decision making is a critical goal in the effort towards precision cancer medicine. High-throughput molecular assays paired with modern computational techniques have the potential to identify individual tumor-specific signatures and create tools that can help understand heterogenous patient outcomes in response to radiotherapy, allowing clinicians to fully benefit from the technological advances in molecular profiling and computational biology including machine learning. However, the increasingly complex nature of the data generated from high-throughput and "omics" assays require careful selection of analytical strategies. Furthermore, the power of modern machine learning techniques to detect subtle data patterns comes with special considerations to ensure that the results are generalizable. Herein, we review the computational framework of tumor biomarker development and describe commonly used machine learning approaches and how they are applied for radiation biomarker development using molecular data, as well as challenges and emerging research trends.

      PMID:37331779 | PMC:PMC10287033 | DOI:10.1016/j.semradonc.2023.03.002


      View details for PubMedID 37331779
  • Fragmentomic analysis of circulating tumor DNA-targeted cancer panels Annals of oncology : official journal of the European Society for Medical Oncology
    Helzer KT, Sharifi MN, Sperger JM, Shi Y, Annala M, Bootsma ML, Reese SR, Taylor A, Kaufmann KR, Krause HK, Schehr JL, Sethakorn N, Kosoff D, Kyriakopoulos C, Burkard ME, Rydzewski NR, Yu M, Harari PM, Bassetti M, Blitzer G, Floberg J, Sjöström M, Quigley DA, Dehm SM, Armstrong AJ, Beltran H, McKay RR, Feng FY, O'Regan R, Wisinski KB, Emamekhoo H, Wyatt AW, Lang JM, Zhao SG
    2023 Sep;34(9):813-825. doi: 10.1016/j.annonc.2023.06.001. Epub 2023 Jun 16.
    • More

      BACKGROUND: The isolation of cell-free DNA (cfDNA) from the bloodstream can be used to detect and analyze somatic alterations in circulating tumor DNA (ctDNA), and multiple cfDNA-targeted sequencing panels are now commercially available for Food and Drug Administration (FDA)-approved biomarker indications to guide treatment. More recently, cfDNA fragmentation patterns have emerged as a tool to infer epigenomic and transcriptomic information. However, most of these analyses used whole-genome sequencing, which is insufficient to identify FDA-approved biomarker indications in a cost-effective manner.

      PATIENTS AND METHODS: We used machine learning models of fragmentation patterns at the first coding exon in standard targeted cancer gene cfDNA sequencing panels to distinguish between cancer and non-cancer patients, as well as the specific tumor type and subtype. We assessed this approach in two independent cohorts: a published cohort from GRAIL (breast, lung, and prostate cancers, non-cancer, n = 198) and an institutional cohort from the University of Wisconsin (UW; breast, lung, prostate, bladder cancers, n = 320). Each cohort was split 70%/30% into training and validation sets.

      RESULTS: In the UW cohort, training cross-validated accuracy was 82.1%, and accuracy in the independent validation cohort was 86.6% despite a median ctDNA fraction of only 0.06. In the GRAIL cohort, to assess how this approach performs in very low ctDNA fractions, training and independent validation were split based on ctDNA fraction. Training cross-validated accuracy was 80.6%, and accuracy in the independent validation cohort was 76.3%. In the validation cohort where the ctDNA fractions were all <0.05 and as low as 0.0003, the cancer versus non-cancer area under the curve was 0.99.

      CONCLUSIONS: To our knowledge, this is the first study to demonstrate that sequencing from targeted cfDNA panels can be utilized to analyze fragmentation patterns to classify cancer types, dramatically expanding the potential capabilities of existing clinically used panels at minimal additional cost.

      PMID:37330052 | PMC:PMC10527168 | DOI:10.1016/j.annonc.2023.06.001


      View details for PubMedID 37330052
  • Expression and Therapeutic Targeting of TROP-2 in Treatment-Resistant Prostate Cancer Clinical cancer research : an official journal of the American Association for Cancer Research
    Sperger JM, Helzer KT, Stahlfeld CN, Jiang D, Singh A, Kaufmann KR, Niles DJ, Heninger E, Rydzewski NR, Wang L, Wang L, Yang R, Ren Y, Engle JW, Huang P, Kyriakopoulos CE, Slovin SF, Soule HR, Zhao SG, Kohli M, Tagawa ST, Cai W, Dehm SM, Lang JM
    2023 Jun 13;29(12):2324-2335. doi: 10.1158/1078-0432.CCR-22-1305.
    • More

      PURPOSE: Men with metastatic castration-resistant prostate cancer (mCRPC) frequently develop resistance to androgen receptor signaling inhibitor (ARSI) treatment; therefore, new therapies are needed. Trophoblastic cell-surface antigen (TROP-2) is a transmembrane protein identified in prostate cancer and overexpressed in multiple malignancies. TROP-2 is a therapeutic target for antibody-drug conjugates (ADC).

      EXPERIMENTAL DESIGN: TROP-2 gene (TACSTD2) expression and markers of treatment resistance from prostate biopsies were analyzed using data from four previously curated cohorts of mCRPC (n = 634) and the PROMOTE study (dbGaP accession phs001141.v1.p1, n = 88). EPCAM or TROP-2-positive circulating tumor cells (CTC) were captured from peripheral blood for comparison of protein (n = 15) and gene expression signatures of treatment resistance (n = 40). We assessed the efficacy of TROP-2-targeting agents in a mouse xenograft model generated from prostate cancer cell lines.

      RESULTS: We demonstrated that TACSTD2 is expressed in mCRPC from luminal and basal tumors but at lower levels in patients with neuroendocrine prostate cancer. Patients previously treated with ARSI showed no significant difference in TACSTD2 expression, whereas patients with detectable AR-V7 expression showed increased expression. We observed that TROP-2 can serve as a cell surface target for isolating CTCs, which may serve as a predictive biomarker for ADCs. We also demonstrated that prostate cancer cell line xenografts can be targeted specifically by labeled anti-TROP-2 agents in vivo.

      CONCLUSIONS: These results support further studies on TROP-2 as a therapeutic and diagnostic target for mCRPC.

      PMID:36939530 | PMC:PMC10261916 | DOI:10.1158/1078-0432.CCR-22-1305


      View details for PubMedID 36939530
  • A clinical-grade liquid biomarker detects neuroendocrine differentiation in prostate cancer The Journal of clinical investigation
    Zhao SG, Sperger JM, Schehr JL, McKay RR, Emamekhoo H, Singh A, Schultz ZD, Bade RM, Stahlfeld CN, Gilsdorf CS, Hernandez CI, Wolfe SK, Mayberry RD, Krause HM, Bootsma M, Helzer KT, Rydzewski N, Bakhtiar H, Shi Y, Blitzer G, Kyriakopoulos CE, Kosoff D, Wei XX, Floberg J, Sethakorn N, Sharifi M, Harari PM, Huang W, Beltran H, Choueiri TK, Scher HI, Rathkopf DE, Halabi S, Armstrong AJ, Beebe DJ, Yu M, Sundling KE, Taplin M, Lang JM
    2022 Nov 1;132(21):e161858. doi: 10.1172/JCI161858.
    • More

      BackgroundNeuroendocrine prostate cancer (NEPC) is an aggressive subtype, the presence of which changes the prognosis and management of metastatic prostate cancer.MethodsWe performed analytical validation of a Circulating Tumor Cell (CTC) multiplex RNA qPCR assay to identify the limit of quantification (LOQ) in cell lines, synthetic cDNA, and patient samples. We next profiled 116 longitudinal samples from a prospectively collected institutional cohort of 17 patients with metastatic prostate cancer (7 NEPC, 10 adenocarcinoma) as well as 265 samples from 139 patients enrolled in 3 adenocarcinoma phase II trials of androgen receptor signaling inhibitors (ARSIs). We assessed a NEPC liquid biomarker via the presence of neuroendocrine markers and the absence of androgen receptor (AR) target genes.ResultsUsing the analytical validation LOQ, liquid biomarker NEPC detection in the longitudinal cohort had a per-sample sensitivity of 51.35% and a specificity of 91.14%. However, when we incorporated the serial information from multiple liquid biopsies per patient, a unique aspect of this study, the per-patient predictions were 100% accurate, with a receiver-operating-curve (ROC) AUC of 1. In the adenocarcinoma ARSI trials, the presence of neuroendocrine markers, even while AR target gene expression was retained, was a strong negative prognostic factor.ConclusionOur analytically validated CTC biomarker can detect NEPC with high diagnostic accuracy when leveraging serial samples that are only feasible using liquid biopsies. Patients with expression of NE genes while retaining AR-target gene expression may indicate the transition to neuroendocrine differentiation, with clinical characteristics consistent with this phenotype.FundingNIH (DP2 OD030734, 1UH2CA260389, R01CA247479, and P30 CA014520), Department of Defense (PC190039 and PC200334), and Prostate Cancer Foundation (Movember Foundation - PCF Challenge Award).

      PMID:36317634 | PMC:PMC9621140 | DOI:10.1172/JCI161858


      View details for PubMedID 36317634
  • Intrinsic Molecular Subtypes of Metastatic Castration-Resistant Prostate Cancer Clinical cancer research : an official journal of the American Association for Cancer Research
    Feng E, Rydzewski NR, Zhang M, Lundberg A, Bootsma M, Helzer KT, Lang JM, Aggarwal R, Small EJ, Quigley DA, Sjöström M, Zhao SG
    2022 Dec 15;28(24):5396-5404. doi: 10.1158/1078-0432.CCR-22-2567.
    • More

      PURPOSE: Although numerous biology-driven subtypes have been described previously in metastatic castration-resistant prostate cancer (mCRPC), unsupervised molecular subtyping based on gene expression has been less studied, especially using large cohorts. Thus, we sought to identify the intrinsic molecular subtypes of mCRPC and assess molecular and clinical correlates in the largest combined cohort of mCRPC samples with gene expression data available to date.

      EXPERIMENTAL DESIGN: We combined and batch-effect corrected gene expression data from four mCRPC cohorts from the Fred Hutchinson Cancer Research Center (N = 157), a small-cell neuroendocrine (NE) prostate cancer (SCNC)-enriched cohort from Weill Cornell Medicine (N = 49), and cohorts from the Stand Up 2 Cancer/Prostate Cancer Foundation East Coast Dream Team (N = 266) and the West Coast Dream Team (N = 162).

      RESULTS: Hierarchical clustering of RNA-sequencing data from these 634 mCRPC samples identified two distinct adenocarcinoma subtypes, one of which (adeno-immune) was characterized by higher gene expression of immune pathways, higher CIBERSORTx immune scores, diminished ASI benefit, and non-lymph node metastasis tropism compared with an adeno-classic subtype. We also identified two distinct subtypes with enrichment for an NE phenotype, including an NE-liver subgroup characterized by liver metastasis tropism, PTEN loss, and APC and SPOP mutations compared with an NE-classic subgroup.

      CONCLUSIONS: Our results emphasize the heterogeneity of mCRPC beyond currently accepted molecular phenotypes, and suggest that future studies should consider incorporating transcriptome-wide profiling to better understand how these differences impact treatment responses and outcomes.

      PMID:36260524 | PMC:PMC9890931 | DOI:10.1158/1078-0432.CCR-22-2567


      View details for PubMedID 36260524
  • Identification of phenocopies improves prediction of targeted therapy response over DNA mutations alone NPJ genomic medicine
    Bakhtiar H, Helzer KT, Park Y, Chen Y, Rydzewski NR, Bootsma ML, Shi Y, Harari PM, Sharifi M, Sjöström M, Lang JM, Yu M, Zhao SG
    2022 Oct 17;7(1):58. doi: 10.1038/s41525-022-00328-7.
    • More

      DNA mutations in specific genes can confer preferential benefit from drugs targeting those genes. However, other molecular perturbations can "phenocopy" pathogenic mutations, but would not be identified using standard clinical sequencing, leading to missed opportunities for other patients to benefit from targeted treatments. We hypothesized that RNA phenocopy signatures of key cancer driver gene mutations could improve our ability to predict response to targeted therapies, despite not being directly trained on drug response. To test this, we built gene expression signatures in tissue samples for specific mutations and found that phenocopy signatures broadly increased accuracy of drug response predictions in-vitro compared to DNA mutation alone, and identified additional cancer cell lines that respond well with a positive/negative predictive value on par or better than DNA mutations. We further validated our results across four clinical cohorts. Our results suggest that routine RNA sequencing of tumors to identify phenocopies in addition to standard targeted DNA sequencing would improve our ability to accurately select patients for targeted therapies in the clinic.

      PMID:36253482 | PMC:PMC9576758 | DOI:10.1038/s41525-022-00328-7


      View details for PubMedID 36253482
  • The 5-Hydroxymethylcytosine Landscape of Prostate Cancer Cancer research
    Sjöström M, Zhao SG, Levy S, Zhang M, Ning Y, Shrestha R, Lundberg A, Herberts C, Foye A, Aggarwal R, Hua JT, Li H, Bergamaschi A, Maurice-Dror C, Maheshwari A, Chen S, Ng WS, Ye W, Petricca J, Fraser M, Chesner L, Perry MD, Moreno-Rodriguez T, Chen WS, Alumkal JJ, Chou J, Morgans AK, Beer TM, Thomas GV, Gleave M, Lloyd P, Phillips T, McCarthy E, Haffner MC, Zoubeidi A, Annala M, Reiter RE, Rettig MB, Witte ON, Fong L, Bose R, Huang FW, Luo J, Bjartell A, Lang JM, Mahajan NP, Lara PN, Evans CP, Tran PT, Posadas EM, He C, Cui X, Huang J, Zwart W, Gilbert LA, Maher CA, Boutros PC, Chi KN, Ashworth A, Small EJ, He HH, Wyatt AW, Quigley DA, Feng FY
    2022 Nov 2;82(21):3888-3902. doi: 10.1158/0008-5472.CAN-22-1123.
    • More

      Analysis of DNA methylation is a valuable tool to understand disease progression and is increasingly being used to create diagnostic and prognostic clinical biomarkers. While conversion of cytosine to 5-methylcytosine (5mC) commonly results in transcriptional repression, further conversion to 5-hydroxymethylcytosine (5hmC) is associated with transcriptional activation. Here we perform the first study integrating whole-genome 5hmC with DNA, 5mC, and transcriptome sequencing in clinical samples of benign, localized, and advanced prostate cancer. 5hmC is shown to mark activation of cancer drivers and downstream targets. Furthermore, 5hmC sequencing revealed profoundly altered cell states throughout the disease course, characterized by increased proliferation, oncogenic signaling, dedifferentiation, and lineage plasticity to neuroendocrine and gastrointestinal lineages. Finally, 5hmC sequencing of cell-free DNA from patients with metastatic disease proved useful as a prognostic biomarker able to identify an aggressive subtype of prostate cancer using the genes TOP2A and EZH2, previously only detectable by transcriptomic analysis of solid tumor biopsies. Overall, these findings reveal that 5hmC marks epigenomic activation in prostate cancer and identify hallmarks of prostate cancer progression with potential as biomarkers of aggressive disease.

      SIGNIFICANCE: In prostate cancer, 5-hydroxymethylcytosine delineates oncogene activation and stage-specific cell states and can be analyzed in liquid biopsies to detect cancer phenotypes. See related article by Wu and Attard, p. 3880.

      PMID:36251389 | PMC:PMC9627125 | DOI:10.1158/0008-5472.CAN-22-1123


      View details for PubMedID 36251389
  • Radiomic Modeling of Bone Density and Rib Fracture Risk After Stereotactic Body Radiation Therapy for Early-Stage Non-Small Cell Lung Cancer Advances in radiation oncology
    Rydzewski NR, Yadav P, Musunuru HB, Condit KM, Francis D, Zhao SG, Baschnagel AM
    2021 Dec 29;7(3):100884. doi: 10.1016/j.adro.2021.100884. eCollection 2022 May-Jun.
    • More

      PURPOSE: Our purpose was to determine whether bone density and bone-derived radiomic metrics in combination with dosimetric variables could improve risk stratification of rib fractures after stereotactic body radiation therapy (SBRT) for early-stage non-small cell lung cancer (NSCLC).

      METHODS AND MATERIALS: A retrospective analysis was conducted of patients with early-stage NSCLC treated with SBRT. Dosimetric data and rib radiomic data extracted using PyRadiomics were used for the analysis. A subset of patients had bone density scans that were used to create a predicted bone density score for all patients. A 10-fold cross validated approach with 10 resamples was used to find the top univariate logistic models and elastic net regression models that predicted for rib fracture.

      RESULTS: A total of 192 treatment plans were included in the study with a rib fracture rate of 16.1%. A predicted bone density score was created from a multivariate model with vertebral body Hounsfield units and patient weight, with an R-squared of 0.518 compared with patient dual-energy x-ray absorptiometry T-scores. When analyzing all patients, a low predicted bone density score approached significance for increased risk of rib fracture (P = .07). On competing risk analysis, when stratifying patients based on chest wall V30 Gy and bone density score, those with a V30 Gy ≥30 cc and a low bone density score had a significantly higher risk of rib fracture compared with all other patients (P < .001), with a predicted 2-year risk of rib fracture of 28.6% (95% confidence interval, 17.2%-41.1%) and 4.9% (95% confidence interval, 2.3%-9.0%), respectively. Dosimetric variables were the primary drivers of fracture risk. A multivariate elastic net regression model including all dosimetric variables was the best predictor of rib fracture (area under the curve [AUC], 0.864). Bone density variables (AUC, 0.618) and radiomic variables (AUC, 0.617) have better predictive power than clinical variables that exclude bone density (AUC, 0.538).

      CONCLUSION: Radiomic features, including a bone density score that includes vertebral body Hounsfield units and radiomic signatures from the ribs, can be used to stratify risk of rib fracture after SBRT for NSCLC.

      PMID:35647405 | PMC:PMC9133372 | DOI:10.1016/j.adro.2021.100884


      View details for PubMedID 35647405
  • Longitudinal Molecular Profiling of Circulating Tumor Cells in Metastatic Renal Cell Carcinoma Journal of clinical oncology : official journal of the American Society of Clinical Oncology
    Bootsma M, McKay RR, Emamekhoo H, Bade RM, Schehr JL, Mannino MC, Singh A, Wolfe SK, Schultz ZD, Sperger J, Xie W, Signoretti S, Kyriakopoulos CE, Kosoff D, Abel EJ, Helzer KT, Rydzewski N, Bakhtiar H, Shi Y, Blitzer G, Bassetti M, Floberg J, Yu M, Sethakorn N, Sharifi M, Harari PM, Choueiri TK, Lang JM, Zhao SG
    2022 Nov 1;40(31):3633-3641. doi: 10.1200/JCO.22.00219. Epub 2022 May 26.
    • More

      PURPOSE: Liquid biopsies in metastatic renal cell carcinoma (mRCC) provide a unique approach to understand the molecular basis of treatment response and resistance. This is particularly important in the context of immunotherapies, which target key immune-tumor interactions. Unlike metastatic tissue biopsies, serial real-time profiling of mRCC is feasible with our noninvasive circulating tumor cell (CTC) approach.

      METHODS: We collected 457 longitudinal liquid biopsies from 104 patients with mRCC enrolled in one of two studies, either a prospective cohort or a phase II multicenter adaptive immunotherapy trial. Using a novel CTC capture and automated microscopy platform, we profiled CTC enumeration and expression of HLA I and programmed cell death-ligand 1 (PD-L1). Given their diametric immunological roles, we focused on the HLA I to PD-L1 ratio (HP ratio).

      RESULTS: Patients with radiographic responses showed significantly lower CTC abundances throughout treatment. Furthermore, patients whose CTC enumeration trajectory was in the highest quartile (> 0.12 CTCs/mL annually) had shorter overall survival (median 17.0 months v 21.1 months, P < .001). Throughout treatment, the HP ratio decreased in patients receiving immunotherapy but not in patients receiving tyrosine kinase inhibitors. Patients with an HP ratio trajectory in the highest quartile (≥ 0.0012 annually) displayed significantly shorter overall survival (median 18.4 months v 21.2 months, P = .003).

      CONCLUSION: In the first large longitudinal CTC study in mRCC to date to our knowledge, we identified the prognostic importance of CTC enumeration and the change over time of both CTC enumeration and the HP ratio. These insights into changes in both tumor burden and the molecular profile of tumor cells in response to different treatments provide potential biomarkers to predict and monitor response to immunotherapy in mRCC.

      PMID:35617646 | PMC:PMC9622626 | DOI:10.1200/JCO.22.00219


      View details for PubMedID 35617646
  • A non-coding RNA balancing act: miR-346-induced DNA damage is limited by the long non-coding RNA NORAD in prostate cancer Molecular cancer
    Fletcher CE, Deng L, Orafidiya F, Yuan W, Lorentzen GS, Cyran OW, Varela-Carver A, Constantin TA, Leach DA, Dobbs FM, Figueiredo I, Gurel B, Parkes E, Bogdan D, Pereira RR, Zhao SG, Neeb A, Issa F, Hester J, Kudo H, Liu Y, Philippou Y, Bristow R, Knudsen K, Bryant RJ, Feng FY, Reed SH, Mills IG, Bono Jd, Bevan CL
    2022 Mar 22;21(1):82. doi: 10.1186/s12943-022-01540-w.
    • More

      BACKGROUND: miR-346 was identified as an activator of Androgen Receptor (AR) signalling that associates with DNA damage response (DDR)-linked transcripts in prostate cancer (PC). We sought to delineate the impact of miR-346 on DNA damage, and its potential as a therapeutic agent.

      METHODS: RNA-IP, RNA-seq, RNA-ISH, DNA fibre assays, in vivo xenograft studies and bioinformatics approaches were used alongside a novel method for amplification-free, single nucleotide-resolution genome-wide mapping of DNA breaks (INDUCE-seq).

      RESULTS: miR-346 induces rapid and extensive DNA damage in PC cells - the first report of microRNA-induced DNA damage. Mechanistically, this is achieved through transcriptional hyperactivation, R-loop formation and replication stress, leading to checkpoint activation and cell cycle arrest. miR-346 also interacts with genome-protective lncRNA NORAD to disrupt its interaction with PUM2, leading to PUM2 stabilisation and its increased turnover of DNA damage response (DDR) transcripts. Confirming clinical relevance, NORAD expression and activity strongly correlate with poor PC clinical outcomes and increased DDR in biopsy RNA-seq studies. In contrast, miR-346 is associated with improved PC survival. INDUCE-seq reveals that miR-346-induced DSBs occur preferentially at binding sites of the most highly-transcriptionally active transcription factors in PC cells, including c-Myc, FOXA1, HOXB13, NKX3.1, and importantly, AR, resulting in target transcript downregulation. Further, RNA-seq reveals widespread miR-346 and shNORAD dysregulation of DNA damage, replication and cell cycle processes. NORAD drives target-directed miR decay (TDMD) of miR-346 as a novel genome protection mechanism: NORAD silencing increases mature miR-346 levels by several thousand-fold, and WT but not TDMD-mutant NORAD rescues miR-346-induced DNA damage. Importantly, miR-346 sensitises PC cells to DNA-damaging drugs including PARP inhibitor and chemotherapy, and induces tumour regression as a monotherapy in vivo, indicating that targeting miR-346:NORAD balance is a valid therapeutic strategy.

      CONCLUSIONS: A balancing act between miR-346 and NORAD regulates DNA damage and repair in PC. miR-346 may be particularly effective as a therapeutic in the context of decreased NORAD observed in advanced PC, and in transcriptionally-hyperactive cancer cells.

      PMID:35317841 | PMC:PMC8939142 | DOI:10.1186/s12943-022-01540-w


      View details for PubMedID 35317841
  • SEEMLIS: a flexible semi-automated method for enrichment of methylated DNA from low-input samples Clinical epigenetics
    Rodems TS, Juang DS, Stahlfeld CN, Gilsdorf CS, Krueger EG, Heninger E, Zhao SG, Sperger JM, Beebe DJ, Haffner MC, Lang JM
    2022 Mar 10;14(1):37. doi: 10.1186/s13148-022-01252-4.
    • More

      BACKGROUND: DNA methylation alterations have emerged as hallmarks of cancer and have been proposed as screening, prognostic, and predictive biomarkers. Traditional approaches for methylation analysis have relied on bisulfite conversion of DNA, which can damage DNA and is not suitable for targeted gene analysis in low-input samples. Here, we have adapted methyl-CpG-binding domain protein 2 (MBD2)-based DNA enrichment for use on a semi-automated exclusion-based sample preparation (ESP) platform for robust and scalable enrichment of methylated DNA from low-input samples, called SEEMLIS.

      RESULTS: We show that combining methylation-sensitive enzyme digestion with ESP-based MBD2 enrichment allows for single gene analysis with high sensitivity for GSTP1 in highly impure, heterogenous samples. We also show that ESP-based MBD2 enrichment coupled with targeted pre-amplification allows for analysis of multiple genes with sensitivities approaching the single cell level in pure samples for GSTP1 and RASSF1 and sensitivity down to 14 cells for these genes in highly impure samples. Finally, we demonstrate the potential clinical utility of SEEMLIS by successful detection of methylated gene signatures in circulating tumor cells (CTCs) from patients with prostate cancer with varying CTC number and sample purity.

      CONCLUSIONS: SEEMLIS is a robust assay for targeted DNA methylation analysis in low-input samples, with flexibility at multiple steps. We demonstrate the feasibility of this assay to analyze DNA methylation in prostate cancer cells using CTCs from patients with prostate cancer as a real-world example of a low-input analyte of clinical importance. In summary, this novel assay provides a platform for determining methylation signatures in rare cell populations with broad implications for research as well as clinical applications.

      PMID:35272673 | PMC:PMC8908705 | DOI:10.1186/s13148-022-01252-4


      View details for PubMedID 35272673
  • Reply to M. K. Bos et al Journal of clinical oncology : official journal of the American Society of Clinical Oncology
    Sperger JM, Feng FY, Armstrong AJ, Zhao SG, Lang JM
    2022 Feb 10;40(5):520-522. doi: 10.1200/JCO.21.02238. Epub 2021 Dec 8.
  • Prostate cancer research in the 21st century; report from the 2021 Coffey-Holden prostate cancer academy meeting The Prostate
    Miyahira AK, Zarif JC, Coombs CC, Flavell RR, Russo JW, Zaidi S, Zhao D, Zhao SG, Pienta KJ, Soule HR
    2022 Feb;82(2):169-181. doi: 10.1002/pros.24262. Epub 2021 Nov 3.
    • More

      INTRODUCTION: The 2021 Coffey-Holden Prostate Cancer Academy (CHPCA) Meeting, "Prostate Cancer Research in the 21st Century," was held virtually, from June 24-25, 2021.

      METHODS: The CHPCA Meeting is organized by the Prostate Cancer Foundation as a unique discussion-oriented meeting focusing on critical topics in prostate cancer research envisioned to bridge the next major advances in prostate cancer biology and treatment. The 2021 CHPCA Meeting was virtually attended by 89 investigators and included 31 talks over nine sessions.

      RESULTS: Major topic areas discussed at the meeting included: cancer genomics and sequencing, functional genomic approaches to studying mediators of plasticity, emerging signaling pathways in metastatic castration resistant prostate cancer, Wnt signaling biology and the challenges of targeted therapy, clonal hematopoiesis, neuroendocrine cell plasticity and antitumor immunity, cancer immunotherapy and its synergizers, and imaging the tumor microenvironment and metabolism.

      DISCUSSION: This meeting report summarizes the research presented at the 2021 CHPCA Meeting. We hope that publication of this knowledge will accelerate new understandings and the development of new biomarkers and treatments for prostate cancer.

      PMID:34734426 | PMC:PMC8688282 | DOI:10.1002/pros.24262


      View details for PubMedID 34734426
  • Prognosis Associated With Luminal and Basal Subtypes of Metastatic Prostate Cancer JAMA oncology
    Aggarwal R, Rydzewski NR, Zhang L, Foye A, Kim W, Helzer KT, Bakhtiar H, Chang SL, Perry MD, Gleave M, Reiter RE, Huang J, Evans CP, Alumkal JJ, Lang JM, Yu M, Quigley DA, Sjöström M, Small EJ, Feng FY, Zhao SG
    2021 Nov 1;7(11):1644-1652. doi: 10.1001/jamaoncol.2021.3987.
    • More

      IMPORTANCE: Luminal and basal subtypes of primary prostate cancer have been shown to be molecularly distinct and clinically important in predicting response to therapy. These subtypes have not been described in metastatic prostate cancer.

      OBJECTIVES: To identify clinical and molecular correlates of luminal and basal subtypes in metastatic castration-resistant prostate cancer (mCRPC) and investigate differences in survival, particularly after treatment with androgen-signaling inhibitors (ASIs).

      DESIGN, SETTING, AND PARTICIPANTS: In this cohort study, a retrospective analysis was conducted of 4 cohorts with mCRPC (N = 634) across multiple academic centers. Treatment was at the physicians' discretion. Details of the study cohorts have been published elsewhere between 2016 and 2019. Data were analyzed from March 2018 to February 2021.

      MAIN OUTCOMES AND MEASURES: The primary clinical end point was overall survival from the date of tissue biopsy/molecular profiling. Luminal and basal subtypes were also stratified by postbiopsy ASI treatment. The primary molecular analyses included associations with small cell/neuroendocrine prostate cancer (SCNC), molecular pathways, and DNA alterations.

      RESULTS: In the 634 patients, 288 (45%) had tumors classified as luminal, and 346 (55%) had tumors classified as basal. However, 53 of 59 (90%) SCNC tumors were basal (P < .001). Similar to primary prostate cancer, luminal tumors exhibited overexpression of AR pathway genes. In basal tumors, a significantly higher rate of RB1 loss (23% basal vs 4% luminal; P < .001), FOXA1 alterations (36% basal vs 27% luminal; P = .03) and MYC alterations (73% basal vs 56% luminal; P < .001) were identified. Patients with basal tumors had worse overall survival compared with those with luminal tumors only in patients treated with an ASI postbiopsy (East Coast Dream Team: hazard ratio [HR], 0.39; 95% CI, 0.20-0.74; P = .004; West Coast Dream Team: HR, 0.57; 95% CI, 0.33-0.97; P = .04). Among patients with luminal tumors, those treated with an ASI had significantly better survival (HR, 0.27; 95% CI, 0.14-0.53; P < .001), whereas patients with basal tumors did not (HR, 0.62; 95% CI, 0.36-1.04, P = .07). The interaction term between subtype and ASI treatment was statistically significant (HR, 0.42; 95% CI, 0.20-0.89; P = .02).

      CONCLUSIONS AND RELEVANCE: These findings represent the largest integrated clinical, transcriptomic, and genomic analysis of mCRPC samples to date, and suggest that mCRPC can be classified as luminal and basal tumors. Analogous to primary prostate cancer, these data suggest that the benefit of ASI treatment is more pronounced in luminal tumors and support the use of ASIs in this population. In the basal tumors, a chemotherapeutic approach could be considered in some patients given the similarity to SCNC and the diminished benefit of ASI therapy. Further validation in prospective clinical trials is warranted.

      PMID:34554200 | PMC:PMC8461554 | DOI:10.1001/jamaoncol.2021.3987


      View details for PubMedID 34554200
  • Novel genomic signature predictive of response to immune checkpoint blockade: A pan-cancer analysis from project Genomics Evidence Neo-plasia Information Exchange (GENIE) Cancer genetics
    Swami N, Hwang WL, Guo JA, Hoffman H, Abramowitz MC, Elbakouny Z, Beltran H, Chipidza F, Choueiri T, Pra AD, Huang F, Kaochar S, Kantoff P, Kim DW, Kishan AU, Kobetz E, Marinac C, Mucci LA, Muralidhar V, Pollack A, Sanford NN, Schaeffer EM, Spratt DE, Zhao SG, Rebbeck TR, Nguyen PL, Feng FY, Mahal BA, Alshalalfa M
    2021 Nov;258-259:61-68. doi: 10.1016/j.cancergen.2021.08.004. Epub 2021 Aug 28.
    • More

      BACKGROUND: High tumor mutation burden (TMB) and total mutation count (TMC) can be predictive of better response to immune checkpoint blockade (ICB). Nevertheless, TMB and TMC are limited by variation across cancers and inconsistent definitions due to different profiling methods (targeted vs whole genome sequencing). Our objective was to identify genomic alterations (GAs) associated with ICB response and builds a novel genomic signature predictive of ICB response, independent of TMB/TMC.

      METHODS: This was a pan-cancer next generation sequencing (NGS)-association study using January 2014-May 2016 data from AACR Project Genomics Evidence Neo-plasia Information Exchange (GENIE). Participants included 6619 patients with metastatic or un-resectable cancer across 9 cancer types (including 1572 ICB-treated patients). GA data was collected using next-generation sequencing (NGS) assays and downloaded from cbioportal.org. Predictive analyses for ICB response were performed to develop the signature (ImmGA).

      RESULTS: GAs in 16 genes were associated with improved OS in ICB-treated patients (p < 0.005). 13 GAs were associated with an OS benefit in ICB-treated patients (Pinteraction < 0.05); these genes composed the ImmGA signature. High ImmGA score (≥2 alterations out of 13 predictive GAs) was associated with better OS in ICB-treated patients (AHR:0.67, 95%CI [0.6-0.75], p = 1.4e-12), even after accounting for TMC (Pinteraction = 8e-16). High ImmGA was associated with better OS in ICB-treated patients across most cancers and across different ICB treatment modalities.

      CONCLUSION: A novel signature predictive of ICB response (ImmGA) was developed from 13 GAs. Further investigation of the utility of ImmGA for treatment and trial selection is warranted.

      PMID:34551377 | DOI:10.1016/j.cancergen.2021.08.004


      View details for PubMedID 34551377
  • Predicting cancer drug TARGETS - TreAtment Response Generalized Elastic-neT Signatures NPJ genomic medicine
    Rydzewski NR, Peterson E, Lang JM, Yu M, Chang SL, Sjöström M, Bakhtiar H, Song G, Helzer KT, Bootsma ML, Chen WS, Shrestha RM, Zhang M, Quigley DA, Aggarwal R, Small EJ, Wahl DR, Feng FY, Zhao SG
    2021 Sep 21;6(1):76. doi: 10.1038/s41525-021-00239-z.
    • More

      We are now in an era of molecular medicine, where specific DNA alterations can be used to identify patients who will respond to specific drugs. However, there are only a handful of clinically used predictive biomarkers in oncology. Herein, we describe an approach utilizing in vitro DNA and RNA sequencing and drug response data to create TreAtment Response Generalized Elastic-neT Signatures (TARGETS). We trained TARGETS drug response models using Elastic-Net regression in the publicly available Genomics of Drug Sensitivity in Cancer (GDSC) database. Models were then validated on additional in-vitro data from the Cancer Cell Line Encyclopedia (CCLE), and on clinical samples from The Cancer Genome Atlas (TCGA) and Stand Up to Cancer/Prostate Cancer Foundation West Coast Prostate Cancer Dream Team (WCDT). First, we demonstrated that all TARGETS models successfully predicted treatment response in the separate in-vitro CCLE treatment response dataset. Next, we evaluated all FDA-approved biomarker-based cancer drug indications in TCGA and demonstrated that TARGETS predictions were concordant with established clinical indications. Finally, we performed independent clinical validation in the WCDT and found that the TARGETS AR signaling inhibitors (ARSI) signature successfully predicted clinical treatment response in metastatic castration-resistant prostate cancer with a statistically significant interaction between the TARGETS score and PSA response (p = 0.0252). TARGETS represents a pan-cancer, platform-independent approach to predict response to oncologic therapies and could be used as a tool to better select patients for existing therapies as well as identify new indications for testing in prospective clinical trials.

      PMID:34548481 | PMC:PMC8455625 | DOI:10.1038/s41525-021-00239-z


      View details for PubMedID 34548481
  • ATR Inhibitor M6620 (VX-970) Enhances the Effect of Radiation in Non-Small Cell Lung Cancer Brain Metastasis Patient-Derived Xenografts Molecular cancer therapeutics
    Baschnagel AM, Elnaggar JH, VanBeek HJ, Kromke AC, Skiba JH, Kaushik S, Abel L, Clark PA, Longhurst CA, Nickel KP, Leal TA, Zhao SG, Kimple RJ
    2021 Nov;20(11):2129-2139. doi: 10.1158/1535-7163.MCT-21-0305. Epub 2021 Aug 19.
    • More

      M6620, a selective ATP-competitive inhibitor of the ATM and RAD3-related (ATR) kinase, is currently under investigation with radiation in patients with non-small cell lung cancer (NSCLC) brain metastases. We evaluated the DNA damage response (DDR) pathway profile of NSCLC and assessed the radiosensitizing effects of M6620 in a preclinical NSCLC brain metastasis model. Mutation analysis and transcriptome profiling of DDR genes and pathways was performed on NSCLC patient samples. NSCLC cell lines were assessed with proliferation, clonogenic survival, apoptosis, cell cycle, and DNA damage signaling and repair assays. NSCLC brain metastasis patient-derived xenograft models were used to assess intracranial response and overall survival. In vivo IHC was performed to confirm in vitro results. A significant portion of NSCLC patient tumors demonstrated enrichment of DDR pathways. DDR pathways correlated with lung squamous cell histology; and mutations in ATR, ATM, BRCA1, BRCA2, CHEK1, and CHEK2 correlated with enrichment of DDR pathways in lung adenocarcinomas. M6620 reduced colony formation after radiotherapy and resulted in inhibition of DNA DSB repair, abrogation of the radiation-induced G2 cell checkpoint, and formation of dysfunctional micronuclei, leading to enhanced radiation-induced mitotic death. The combination of M6620 and radiation resulted in improved overall survival in mice compared with radiation alone. In vivo IHC revealed inhibition of pChk1 in the radiation plus M6620 group. M6620 enhances the effect of radiation in our preclinical NSCLC brain metastasis models, supporting the ongoing clinical trial (NCT02589522) evaluating M6620 in combination with whole brain irradiation in patients with NSCLC brain metastases.

      PMID:34413128 | PMC:PMC8571002 | DOI:10.1158/1535-7163.MCT-21-0305


      View details for PubMedID 34413128
  • Prospective Evaluation of Clinical Outcomes Using a Multiplex Liquid Biopsy Targeting Diverse Resistance Mechanisms in Metastatic Prostate Cancer Journal of clinical oncology : official journal of the American Society of Clinical Oncology
    Sperger JM, Emamekhoo H, McKay RR, Stahlfeld CN, Singh A, Chen XE, Kwak L, Gilsdorf CS, Wolfe SK, Wei XX, Silver R, Zhang Z, Morris MJ, Bubley G, Feng FY, Scher HI, Rathkopf D, Dehm SM, Choueiri TK, Halabi S, Armstrong AJ, Wyatt AW, Taplin M, Zhao SG, Lang JM
    2021 Sep 10;39(26):2926-2937. doi: 10.1200/JCO.21.00169. Epub 2021 Jul 1.
    • More

      PURPOSE: Nearly all men with prostate cancer treated with androgen receptor (AR) signaling inhibitors (ARSIs) develop resistance via diverse mechanisms including constitutive activation of the AR pathway, driven by AR genomic structural alterations, expression of AR splice variants (AR-Vs), or loss of AR dependence and lineage plasticity termed neuroendocrine prostate cancer. Understanding these de novo acquired ARSI resistance mechanisms is critical for optimizing therapy.

      MATERIALS AND METHODS: A novel liquid biopsy technology was used to collect mRNA from circulating tumor cells (CTCs) to measure expression of AR-Vs, AR targets, and neuroendocrine prostate cancer markers. An institutional review board-approved prospective cohort (N = 99) was used to identify patterns of gene expression. Two prospective multicenter phase II clinical trials of ARSIs for men with castration-resistant prostate cancer (ClinicalTrials.gov: NCT01942837 [enzalutamide, N = 21] and NCT02025010 [abiraterone, N = 27]) were used to further validate these findings.

      RESULTS: Hierarchical clustering of CTC transcripts identified two distinct clusters. Cluster 2 (C2) exhibited increased expression of AR-regulated genes and was associated with worse overall survival (median 8.6 v 22.4 months; P < .01; hazard ratio [HR] = 3.45 [1.9 to 6.14]). In multivariable analysis, C2 was prognostic independent of other clinicopathologic variables. AR-V status was not significant when accounting for C2. Upon further validation in pooled multicenter phase II trials, C2 was associated with worse overall survival (15.2 months v not reached; P < .01; HR = 8.43 [2.74 to 25.92]), prostate-specific antigen progression-free survival (3.6 v 12 months; P < .01; HR = 4.64 [1.53 to 14.11]), and radiographic progression-free survival (2.7 v 40.6 months; P < .01; HR = 4.64 [1.82 to 17.41]).

      CONCLUSION: We demonstrate that a transcriptional profile detectable in CTCs obtained from liquid biopsies can serve as an independent prognostic marker beyond AR-V7 in patients with metastatic prostate cancer and can be used to identify the emergence of multiple ARSI resistance mechanisms. This is currently being investigated in additional prospective trials.

      PMID:34197212 | PMC:PMC8425833 | DOI:10.1200/JCO.21.00169


      View details for PubMedID 34197212
  • Phase II Multicenter Study of Enzalutamide in Metastatic Castration-Resistant Prostate Cancer to Identify Mechanisms Driving Resistance Clinical cancer research : an official journal of the American Association for Cancer Research
    McKay RR, Kwak L, Crowdis JP, Sperger JM, Zhao SG, Xie W, Werner L, Lis RT, Zhang Z, Wei XX, Lang JM, Van Allen M, Bhatt RS, Yu EY, Nelson PS, Bubley GJ, Montgomery RB, Taplin M
    2021 Jul 1;27(13):3610-3619. doi: 10.1158/1078-0432.CCR-20-4616. Epub 2021 Apr 13.
    • More

      PURPOSE: Enzalutamide is a second-generation androgen receptor (AR) inhibitor that has improved overall survival (OS) in metastatic castration-resistant prostate cancer (CRPC). However, nearly all patients develop resistance. We designed a phase II multicenter study of enzalutamide in metastatic CRPC incorporating tissue and blood biomarkers to dissect mechanisms driving resistance.

      PATIENTS AND METHODS: Eligible patients with metastatic CRPC underwent a baseline metastasis biopsy and then initiated enzalutamide 160 mg daily. A repeat metastasis biopsy was obtained at radiographic progression from the same site when possible. Blood for circulating tumor cell (CTC) analysis was collected at baseline and progression. The primary objective was to analyze mechanisms of resistance in serial biopsies. Whole-exome sequencing was performed on tissue biopsies. CTC samples underwent RNA sequencing.

      RESULTS: A total of 65 patients initiated treatment, of whom 22 (33.8%) had received prior abiraterone. Baseline biopsies were enriched for alterations in AR (mutations, amplifications) and tumor suppression genes (PTEN, RB1, and TP53), which were observed in 73.1% and 92.3% of baseline biopsies, respectively. Progression biopsies revealed increased AR amplifications (64.7% at progression vs. 53.9% at baseline) and BRCA2 alterations (64.7% at progression vs. 38.5% at baseline). Genomic analysis of baseline and progression CTC samples demonstrated increased AR splice variants, AR-regulated genes, and neuroendocrine markers at progression.

      CONCLUSIONS: Our results demonstrate that a large proportion of enzalutamide-treated patients have baseline and progression alterations in the AR pathway and tumor suppressor genes. We demonstrate an increased number of BRCA2 alterations post-enzalutamide, highlighting the importance of serial tumor sampling in CRPC.

      PMID:33849963 | PMC:PMC8254786 | DOI:10.1158/1078-0432.CCR-20-4616


      View details for PubMedID 33849963
  • Prostate-specific Membrane Antigen and Fluciclovine Transporter Genes are Associated with Variable Clinical Features and Molecular Subtypes of Primary Prostate Cancer European urology
    Chu CE, Alshalalfa M, Sjöström M, Zhao SG, Liu Y, Chou J, Herlemann A, Mahal B, Kishan AU, Spratt DE, Cooperberg M, Small E, Wong A, Porten S, Hope TA, Ross AE, Davicioni E, Nguyen P, Karnes RJ, Carroll PR, Schaeffer E, Feng FY
    2021 Jun;79(6):717-721. doi: 10.1016/j.eururo.2021.03.017. Epub 2021 Apr 8.
    • More

      18F-Fluciclovine-based positron emission tomography (PET) imaging is recommended in the USA for biochemical recurrence (BCR) after prostate cancer treatment. However, prostate-specific membrane antigen (PSMA)-based PET imaging is more common worldwide, supported by international guidelines, and is now approved by the Food and Drug Administration in the USA for initial staging of primary prostate cancer. Little is known about the molecular profiles of lesions detected by PSMA-targeted PET/computed tomography (CT) versus 18F-fluciclovine PET/CT. We examined the expression of PSMA (FOLH1) and the fluciclovine transporter genes LAT1-4 and ASCT1/2 in a combined cohort of more than 18 000 radical prostatectomy specimens and their associations with clinical outcomes. Expression of PSMA and all but one fluciclovine transporter gene was higher in prostate cancer than in benign tissue. PSMA expression was associated with Gleason score (GS) ≥8 and lymph node involvement (LNI), and had a positive linear correlation with Decipher risk score. By contrast, expression of the fluciclovine transporters LAT2, LAT3, and ASCT2 was negatively associated with GS ≥ 8, LNI, and high Decipher score. The top decile of PSMA expression was associated with poorest metastasis-free survival (MFS), while the bottom deciles of LAT3 and ASCT2 expression were associated with poorest MFS. PATIENT SUMMARY: We measured the expression of genes that encode the targets for two different radiotracers in PET (positron emission tomography) scans of the prostate. We found that PSMA gene expression (PSMA-based tracer) is associated with worse clinical outcomes, while expression of ASCT2, LAT2, and LAT3 genes (fluciclovine tracer) is associated with better outcomes.

      PMID:33840559 | DOI:10.1016/j.eururo.2021.03.017


      View details for PubMedID 33840559
  • Recent Advances in Epigenetic Biomarkers and Epigenetic Targeting in Prostate Cancer European urology
    Kumaraswamy A, Leng RW, Westbrook TC, Yates JA, Zhao SG, Evans CP, Feng FY, Morgan TM, Alumkal JJ
    2021 Jul;80(1):71-81. doi: 10.1016/j.eururo.2021.03.005. Epub 2021 Mar 27.
    • More

      CONTEXT: In addition to genetic alterations, epigenetic alterations play a crucial role during prostate cancer progression. A better understanding of the epigenetic factors that promote prostate cancer progression may lead to the design of rational therapeutic strategies to target prostate cancer more effectively.

      OBJECTIVE: To systematically review recent literature on the role of epigenetic factors in prostate cancer and highlight key preclinical and translational data with epigenetic therapies.

      EVIDENCE ACQUISITION: We performed a systemic literature search in PubMed. At the request of the editors, we limited our search to articles published between January 2015 and August 2020 in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. Clinical trials targeting epigenetic factors were retrieved from clinicaltrials.gov.

      EVIDENCE SYNTHESIS: We retrieved 1451 articles, and 62 were finally selected for review. Twelve additional foundational studies outside this time frame were also included. Findings from both preclinical and clinical studies were reviewed and summarized. We also discuss 12 ongoing clinical studies with epigenetic targeted therapies.

      CONCLUSIONS: Epigenetic mechanisms impact prostate cancer progression. Understanding the role of specific epigenetic factors is critical to determine how we may improve prostate cancer treatment and modulate resistance to standard therapies. Recent preclinical studies and ongoing or completed clinical studies with epigenetic therapies provide a useful roadmap for how to best deploy epigenetic therapies clinically to target prostate cancer.

      PATIENT SUMMARY: Epigenetics is a process by which gene expression is regulated without changes in the DNA sequence itself. Oftentimes, epigenetic changes influence cellular behavior and contribute to cancer development or progression. Understanding how epigenetic changes occur in prostate cancer is the first step toward therapeutic targeting in patients. Importantly, laboratory-based studies and recently completed and ongoing clinical trials suggest that drugs targeting epigenetic factors are promising. More work is necessary to determine whether this class of drugs will add to our existing treatment arsenal in prostate cancer.

      PMID:33785255 | PMC:PMC8547521 | DOI:10.1016/j.eururo.2021.03.005


      View details for PubMedID 33785255
  • Validation of a 22-Gene Genomic Classifier in Patients With Recurrent Prostate Cancer: An Ancillary Study of the NRG/RTOG 9601 Randomized Clinical Trial JAMA oncology
    Feng FY, Huang H, Spratt DE, Zhao SG, Sandler HM, Simko JP, Davicioni E, Nguyen PL, Pollack A, Efstathiou JA, Dicker AP, Todorovic T, Margrave J, Liu YS, Dabbas B, Thompson JS, Das R, Dignam JJ, Sweeney C, Attard G, Bahary J, Lukka HR, Hall WA, Pisansky TM, Shah AB, Pugh SL, Shipley WU, Tran PT
    2021 Apr 1;7(4):544-552. doi: 10.1001/jamaoncol.2020.7671.
    • More

      IMPORTANCE: Decipher (Decipher Biosciences Inc) is a genomic classifier (GC) developed to estimate the risk of distant metastasis (DM) after radical prostatectomy (RP) in patients with prostate cancer.

      OBJECTIVE: To validate the GC in the context of a randomized phase 3 trial.

      DESIGN, SETTING, AND PARTICIPANTS: This ancillary study used RP specimens from the phase 3 placebo-controlled NRG/RTOG 9601 randomized clinical trial conducted from March 1998 to March 2003. The specimens were centrally reviewed, and RNA was extracted from the highest-grade tumor available in 2019 with a median follow-up of 13 years. Clinical-grade whole transcriptomes from samples passing quality control were assigned GC scores (scale, 0-1). A National Clinical Trials Network-approved prespecified statistical plan included the primary objective of validating the independent prognostic ability of GC for DM, with secondary end points of prostate cancer-specific mortality (PCSM) and overall survival (OS). Data were analyzed from September 2019 to December 2019.

      INTERVENTION: Salvage radiotherapy (sRT) with or without 2 years of bicalutamide.

      MAIN OUTCOMES AND MEASURES: The preplanned primary end point of this study was the independent association of the GC with the development of DM.

      RESULTS: In this ancillary study of specimens from a phase 3 randomized clinical trial, GC scores were generated from 486 of 760 randomized patients with a median follow-up of 13 years; samples from a total of 352 men (median [interquartile range] age, 64.5 (60-70) years; 314 White [89.2%] participants) passed microarray quality control and comprised the final cohort for analysis. On multivariable analysis, the GC (continuous variable, per 0.1 unit) was independently associated with DM (hazard ratio [HR], 1.17; 95% CI, 1.05-1.32; P = .006), PCSM (HR, 1.39; 95% CI, 1.20-1.63; P < .001), and OS (HR, 1.17; 95% CI, 1.06-1.29; P = .002) after adjusting for age, race/ethnicity, Gleason score, T stage, margin status, entry prostate-specific antigen, and treatment arm. Although the original planned analysis was not powered to detect a treatment effect interaction by GC score, the estimated absolute effect of bicalutamide on 12-year OS was less when comparing patients with lower vs higher GC scores (2.4% vs 8.9%), which was further demonstrated in men receiving early sRT at a prostate-specific antigen level lower than 0.7 ng/mL (-7.8% vs 4.6%).

      CONCLUSIONS AND RELEVANCE: This ancillary validation study of the Decipher GC in a randomized trial cohort demonstrated association of the GC with DM, PCSM, and OS independent of standard clinicopathologic variables. These results suggest that not all men with biochemically recurrent prostate cancer after surgery benefit equally from the addition of hormone therapy to sRT.

      TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT00002874.

      PMID:33570548 | PMC:PMC7879385 | DOI:10.1001/jamaoncol.2020.7671


      View details for PubMedID 33570548
  • Novel Transcriptomic Interactions Between Immune Content and Genomic Classifier Predict Lethal Outcomes in High-grade Prostate Cancer European urology
    Yamoah K, Awasthi S, Mahal BA, Zhao SG, Grass GD, Berglund A, Abraham-Miranda J, Gerke T, Rounbehler RJ, Davicioni E, Liu Y, Park J, Cleveland JL, Pow-Sang JM, Fernandez D, Torres-Roca J, Karnes RJ, Schaeffer E, Freedland SJ, Spratt DE, Den RB, Rebbeck TR, Feng F
    2022 Apr;81(4):325-330. doi: 10.1016/j.eururo.2020.11.038. Epub 2020 Dec 7.
    • More

      Grade group 4 and 5 (GG-45) prostate cancer (PCa) patients are at the highest risk of lethal outcomes, yet lack genomic risk stratification for prognosis and treatment selection. Here, we assess whether transcriptomic interactions between tumor immune content score (ICS) and the Decipher genomic classifier can identify most lethal subsets of GG-45 PCa. We utilized whole transcriptome data from 8071 tumor tissue (6071 prostatectomy and 2000 treatment-naïve biopsy samples) to derive four immunogenomic subtypes using ICS and Decipher. When compared across all grade groups, GG-45 samples had the highest proportion of most aggressive subtype-ICSHigh/DecipherHigh. Subsequent analyses within the GG-45 patient samples (n = 1420) revealed that the ICSHigh/DecipherHigh subtype was associated with increased genomic radiosensitivity. Additionally, in a multivariable model (n = 335), ICSHigh/DecipherHigh subtype had a significantly higher risk of distant metastasis (hazard ratio [HR] = 5.41; 95% confidence interval [CI], 2.76-10.6; p ≤ 0.0001) and PCa-specific mortality (HR = 10.6; 95% CI, 4.18-26.94; p ≤ 0.0001) as compared with ICSLow/DecipherLow. The novel immunogenomic subtypes establish a very strong synergistic interaction between ICS and Decipher in identifying GG-45 patients who experience the most lethal outcomes. PATIENT SUMMARY: In this analysis, we identified a novel interaction between the total immune content of prostate tumors and genomic classifier to identify the most lethal subset of patients with grade groups 4 and 5. Our results will aid in the subtyping of aggressive prostate cancer patients who may benefit from combined immune-radiotherapy modalities.

      PMID:33303244 | PMC:PMC9838823 | DOI:10.1016/j.eururo.2020.11.038


      View details for PubMedID 33303244
  • A Systematic Review of the Evidence for the Decipher Genomic Classifier in Prostate Cancer European urology
    Jairath NK, Pra AD, Vince R, Dess RT, Jackson WC, Tosoian JJ, McBride SM, Zhao SG, Berlin A, Mahal BA, Kishan AU, Den RB, Freedland SJ, Salami SS, Kaffenberger SD, Pollack A, Tran P, Mehra R, Morgan TM, Weiner AB, Mohamad O, Carroll PR, Cooperberg MR, Karnes RJ, Nguyen PL, Michalski JM, Tward JD, Feng FY, Schaeffer EM, Spratt DE
    2021 Mar;79(3):374-383. doi: 10.1016/j.eururo.2020.11.021. Epub 2020 Dec 5.
    • More

      CONTEXT: Molecular biomarkers aim to address the established limitations of clinicopathologic factors to accurately risk stratify patients with prostate cancer (PCa). Questions remain as to whether sufficient evidence supports adoption of these biomarkers for clinical use.

      OBJECTIVE: To perform a systematic review of the available evidence supporting the clinical utility of the Decipher genomic classifier (GC).

      EVIDENCE ACQUISITION: The review was performed as per the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines by searching PubMed and conference abstracts from January 2010 to June 2020. Evidence was then graded using the criteria of Simon et al (Simon RM, Paik S, Hayes DF. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst 2009;101:1446-52) and American Urology Association (AUA) criteria.

      EVIDENCE SYNTHESIS: In total, 42 studies and 30407 patients were included. GC performance data were available for localized, postprostatectomy, nonmetastatic castration-resistant, and metastatic hormone-sensitive PCa as part of retrospective studies (n=12141), prospective registries (n=17053), and prospective and post hoc randomized trial analyses (n=1213). In 32 studies (n=12600), the GC was independently prognostic for all study endpoints (adverse pathology, biochemical failure, metastasis, and cancer-specific and overall survival) on multivariable analysis and improved the discrimination over standard of care in 24 studies (n=8543). GC use changed the management in active surveillance (number needed to test [NNT]=9) and postprostatectomy (NNT=1.5-4) settings in five studies (n=4331). Evidence strength was levels 1 and 2 by the Simon criteria for all disease states other than high-risk PCa, and grades A and B by AUA criteria depending on disease state.

      CONCLUSIONS: Consistent data are now present from diverse levels of evidence, which when viewed together, have demonstrated clinical utility of the GC in PCa. The utility of the GC is strongest for intermediate-risk PCa and postprostatectomy decision-making.

      PATIENT SUMMARY: In this paper, we review the evidence of the Decipher genomic classification tool for men with prostate cancer. We found consistent evidence that the test helps identify which cancers are more or less aggressive, which in turn aids in personalized treatment decision-making.

      PMID:33293078 | DOI:10.1016/j.eururo.2020.11.021


      View details for PubMedID 33293078
  • SV-HotSpot: detection and visualization of hotspots targeted by structural variants associated with gene expression Scientific reports
    Eteleeb AM, Quigley DA, Zhao SG, Pham D, Yang R, Dehm SM, Luo J, Feng FY, Dang HX, Maher CA
    2020 Sep 28;10(1):15890. doi: 10.1038/s41598-020-71168-7.
    • More

      Whole genome sequencing (WGS) has enabled the discovery of genomic structural variants (SVs), including those targeting intergenic and intronic non-coding regions that eluded previous exome focused strategies. However, the field currently lacks an automated tool that analyzes SV candidates to identify recurrent SVs and their targeted sites (hotspot regions), visualizes these genomic events within the context of various functional elements, and evaluates their potential effect on gene expression. To address this, we developed SV-HotSpot, an automated tool that integrates SV candidates, copy number alterations, gene expression, and genome annotations (e.g. gene and regulatory elements) to discover, annotate, and visualize recurrent SVs and their targeted hotspot regions that may affect gene expression. We applied SV-HotSpot to WGS and matched transcriptome data from metastatic castration resistant prostate cancer patients and rediscovered recurrent SVs targeting coding and non-coding functional elements known to promote prostate cancer progression and metastasis. SV-HotSpot provides a valuable resource to integrate SVs, gene expression, and genome annotations for discovering biologically relevant SVs altering coding and non-coding genome. SV-HotSpot is available at https://github.com/ChrisMaherLab/SV-HotSpot .

      PMID:32985524 | PMC:PMC7522247 | DOI:10.1038/s41598-020-71168-7


      View details for PubMedID 32985524
  • Autoantibody Landscape in Patients with Advanced Prostate Cancer Clinical cancer research : an official journal of the American Association for Cancer Research
    Chen WS, Haynes WA, Waitz R, Kamath K, Vega-Crespo A, Shrestha R, Zhang M, Foye A, Carretero IB, Garcilazo IP, Zhang M, Zhao SG, Sjöström M, Quigley DA, Chou J, Beer TM, Rettig M, Gleave M, Evans CP, Lara P, Chi KN, Reiter RE, Alumkal JJ, Ashworth A, Aggarwal R, Small EJ, Daugherty PS, Ribas A, Oh DY, Shon JC, Feng FY
    2020 Dec 1;26(23):6204-6214. doi: 10.1158/1078-0432.CCR-20-1966. Epub 2020 Sep 23.
    • More

      PURPOSE: Autoantibody responses in cancer are of great interest, as they may be concordant with T-cell responses to cancer antigens or predictive of response to cancer immunotherapies. Thus, we sought to characterize the antibody landscape of metastatic castration-resistant prostate cancer (mCRPC).

      EXPERIMENTAL DESIGN: Serum antibody epitope repertoire analysis (SERA) was performed on patient serum to identify tumor-specific neoepitopes. Somatic mutation-specific neoepitopes were investigated by associating serum epitope enrichment scores with whole-genome sequencing results from paired solid tumor metastasis biopsies and germline blood samples. A protein-based immunome-wide association study (PIWAS) was performed to identify significantly enriched epitopes, and candidate serum antibodies enriched in select patients were validated by ELISA profiling. A distinct cohort of patients with melanoma was evaluated to validate the top cancer-specific epitopes.

      RESULTS: SERA was performed on 1,229 serum samples obtained from 72 men with mCRPC and 1,157 healthy control patients. Twenty-nine of 6,636 somatic mutations (0.44%) were associated with an antibody response specific to the mutated peptide. PIWAS analyses identified motifs in 11 proteins, including NY-ESO-1 and HERVK-113, as immunogenic in mCRPC, and ELISA confirmed serum antibody enrichment in candidate patients. Confirmatory PIWAS, Identifying Motifs Using Next-generation sequencing Experiments (IMUNE), and ELISA analyses performed on serum samples from 106 patients with melanoma similarly revealed enriched cancer-specific antibody responses to NY-ESO-1.

      CONCLUSIONS: We present the first large-scale profiling of autoantibodies in advanced prostate cancer, utilizing a new antibody profiling approach to reveal novel cancer-specific antigens and epitopes. Our study recovers antigens of known importance and identifies novel tumor-specific epitopes of translational interest.

      PMID:32967941 | PMC:PMC7710628 | DOI:10.1158/1078-0432.CCR-20-1966


      View details for PubMedID 32967941
  • Purine metabolism regulates DNA repair and therapy resistance in glioblastoma Nature communications
    Zhou W, Yao Y, Scott AJ, Wilder-Romans K, Dresser JJ, Werner CK, Sun H, Pratt D, Sajjakulnukit P, Zhao SG, Davis M, Nelson BS, Halbrook CJ, Zhang L, Gatto F, Umemura Y, Walker AK, Kachman M, Sarkaria JN, Xiong J, Morgan MA, Rehemtualla A, Castro MG, Lowenstein P, Chandrasekaran S, Lawrence TS, Lyssiotis CA, Wahl DR
    2020 Jul 30;11(1):3811. doi: 10.1038/s41467-020-17512-x.
    • More

      Intratumoral genomic heterogeneity in glioblastoma (GBM) is a barrier to overcoming therapy resistance. Treatments that are effective independent of genotype are urgently needed. By correlating intracellular metabolite levels with radiation resistance across dozens of genomically-distinct models of GBM, we find that purine metabolites, especially guanylates, strongly correlate with radiation resistance. Inhibiting GTP synthesis radiosensitizes GBM cells and patient-derived neurospheres by impairing DNA repair. Likewise, administration of exogenous purine nucleosides protects sensitive GBM models from radiation by promoting DNA repair. Neither modulating pyrimidine metabolism nor purine salvage has similar effects. An FDA-approved inhibitor of GTP synthesis potentiates the effects of radiation in flank and orthotopic patient-derived xenograft models of GBM. High expression of the rate-limiting enzyme of de novo GTP synthesis is associated with shorter survival in GBM patients. These findings indicate that inhibiting purine synthesis may be a promising strategy to overcome therapy resistance in this genomically heterogeneous disease.

      PMID:32732914 | PMC:PMC7393131 | DOI:10.1038/s41467-020-17512-x


      View details for PubMedID 32732914
  • The DNA methylation landscape of advanced prostate cancer Nature genetics
    Zhao SG, Chen WS, Li H, Foye A, Zhang M, Sjöström M, Aggarwal R, Playdle D, Liao A, Alumkal JJ, Das R, Chou J, Hua JT, Barnard TJ, Bailey AM, Chow ED, Perry MD, Dang HX, Yang R, Moussavi-Baygi R, Zhang L, Alshalalfa M, Chang SL, Houlahan KE, Shiah Y, Beer TM, Thomas G, Chi KN, Gleave M, Zoubeidi A, Reiter RE, Rettig MB, Witte O, Kim MY, Fong L, Spratt DE, Morgan TM, Bose R, Huang FW, Li H, Chesner L, Shenoy T, Goodarzi H, Asangani IA, Sandhu S, Lang JM, Mahajan NP, Lara PN, Evans CP, Febbo P, Batzoglou S, Knudsen KE, He HH, Huang J, Zwart W, Costello JF, Luo J, Tomlins SA, Wyatt AW, Dehm SM, Ashworth A, Gilbert LA, Boutros PC, Farh K, Chinnaiyan AM, Maher CA, Small EJ, Quigley DA, Feng FY
    2020 Aug;52(8):778-789. doi: 10.1038/s41588-020-0648-8. Epub 2020 Jul 13.
    • More

      Although DNA methylation is a key regulator of gene expression, the comprehensive methylation landscape of metastatic cancer has never been defined. Through whole-genome bisulfite sequencing paired with deep whole-genome and transcriptome sequencing of 100 castration-resistant prostate metastases, we discovered alterations affecting driver genes that were detectable only with integrated whole-genome approaches. Notably, we observed that 22% of tumors exhibited a novel epigenomic subtype associated with hypermethylation and somatic mutations in TET2, DNMT3B, IDH1 and BRAF. We also identified intergenic regions where methylation is associated with RNA expression of the oncogenic driver genes AR, MYC and ERG. Finally, we showed that differential methylation during progression preferentially occurs at somatic mutational hotspots and putative regulatory regions. This study is a large integrated study of whole-genome, whole-methylome and whole-transcriptome sequencing in metastatic cancer that provides a comprehensive overview of the important regulatory role of methylation in metastatic castration-resistant prostate cancer.

      PMID:32661416 | PMC:PMC7454228 | DOI:10.1038/s41588-020-0648-8


      View details for PubMedID 32661416
  • Tumor Immune Microenvironment Clusters in Localized Prostate Adenocarcinoma: Prognostic Impact of Macrophage Enriched/Plasma Cell Non-Enriched Subtypes Journal of clinical medicine
    Jairath NK, Farha MW, Srinivasan S, Jairath R, Green MD, Dess RT, Jackson WC, Weiner AB, Schaeffer EM, Zhao SG, Feng FY, Naqa IE, Spratt DE
    2020 Jun 24;9(6):1973. doi: 10.3390/jcm9061973.
    • More

      BACKGROUND: Prostate cancer (PCa) is characterized by significant heterogeneity in its molecular, genomic, and immunologic characteristics.

      METHODS: Whole transcriptome RNAseq data from The Cancer Genome Atlas of prostate adenocarcinomas (n = 492) was utilized. The immune microenvironment was characterized using the CIBERSORTX tool to identify immune cell type composition. Unsupervised hierarchical clustering was performed based on immune cell type content. Analyses of progression-free survival (PFS), distant metastases, and overall survival (OS) were performed using Kaplan-Meier estimates and Cox regression multivariable analyses.

      RESULTS: Four immune clusters were identified, largely defined by plasma cell, CD4+ Memory Resting T Cells (CD4 MR), and M0 and M2 macrophage content (CD4 MRHighPlasma CellHighM0LowM2Mid, CD4 MRLowPlasma CellHighM0LowM2Low, CD4 MRHighPlasma CellLowM0HighM2Low, and CD4 MRHighPlasma CellLowM0LowM2High). The two macrophage-enriched/plasma cell non-enriched clusters (3 and 4) demonstrated worse PFS (HR 2.24, 95% CI 1.46-3.45, p = 0.0002) than the clusters 1 and 2. No metastatic events occurred in the plasma cell enriched, non-macrophage-enriched clusters. Comparing clusters 3 vs. 4, in patients treated by surgery alone, cluster 3 had zero progression events (p < 0.0001). However, cluster 3 patients had worse outcomes after post-operative radiotherapy (p = 0.018).

      CONCLUSION: Distinct tumor immune clusters with a macrophage-enriched, plasma cell non-enriched phenotype and reduced plasma cell enrichment independently characterize an aggressive phenotype in localized prostate cancer that may differentially respond to treatment.

      PMID:32599760 | PMC:PMC7356642 | DOI:10.3390/jcm9061973


      View details for PubMedID 32599760
  • A Signature That May Be Predictive of Early Versus Late Recurrence After Radiation Treatment for Breast Cancer That May Inform the Biology of Early, Aggressive Recurrences International journal of radiation oncology, biology, physics
    Speers C, Chang SL, Pesch A, Ritter C, Olsen E, Chandler B, Moubadder L, Liu M, Cameron M, Michmerhuizen A, Wilder-Romans K, Zhao SG, Nyati S, Bartelink H, Feng FY, Pierce LJ
    2020 Nov 1;108(3):686-696. doi: 10.1016/j.ijrobp.2020.05.015. Epub 2020 May 17.
    • More

      PURPOSE: Unmet clinical needs in breast cancer (BC) management include the identification of patients at high risk of local failure despite adjuvant radiation and an understanding of the biology of these recurrences. We previously reported a radiation response signature and here extend those studies to identify a signature predictive of recurrence timing (before or after 3 years).

      METHODS AND MATERIALS: Two independent patient cohorts were used. The training cohort included 119 patients with in-breast tumor recurrence (343 total), and the validation testing cohort had 16 patients with recurrences (112 total). All patients received radiation treatment after breast-conserving surgery. Initial feature selection used Spearman rank correlation, and a linear model was trained and locked before testing and validation. Cox regression was used for univariate and multivariable analyses (UVA and MVA, respectively). Biologically related concepts were identified using gene set enrichment analysis.

      RESULTS: Spearman correlation identified 485 genes whose expression was significantly associated with recurrence time (early vs late). Feature reduction further refined the list to 41 genes retained within the signature. In training, the correlation of score to recurrence time was 0.85 (P value < 1.3 × 10-31) with an area under the curve (AUC) of 0.91. Application of this early versus late signature to an independent BC testing and validation set accurately identified patients with early versus late recurrences (Spearman correlation = 0.75, P value = .001, AUC = 0.92, sensitivity = 0.75, specificity = 1.0, positive predictive value = 1.0, and negative predictive value = 0.8). Unique associations of breast cancer intrinsic subtype to timing of local recurrence were identified. In UVA and MVA the early versus late recurrence signature remained the most significant factor associated with recurrence. Gene set enrichment analysis identified proliferation and epidermal growth factor receptor concepts associated with early recurrences and luminal and ER-signaling pathways associated with late recurrences. Knockdown of genes associated with the early and late recurrences demonstrated novel effects on proliferation and clonogenic survival, respectively.

      CONCLUSIONS: We report a breast cancer gene signature that may identify patients unlikely to respond to adjuvant radiation and may be used to predict timing of recurrences with implications for potential treatment intensification and duration of follow-up for women with breast cancer treated with radiation.

      PMID:32434041 | DOI:10.1016/j.ijrobp.2020.05.015


      View details for PubMedID 32434041
  • Performance of clinicopathologic models in men with high risk localized prostate cancer: impact of a 22-gene genomic classifier Prostate cancer and prostatic diseases
    Tosoian JJ, Birer SR, Karnes RJ, Zhang J, Davicioni E, Klein EE, Freedland SJ, Weinmann S, Trock BJ, Dess RT, Zhao SG, Jackson WC, Yamoah K, Pra AD, Mahal BA, Morgan TM, Mehra R, Kaffenberger S, Salami SS, Kane C, Pollack A, Den RB, Berlin A, Schaeffer EM, Nguyen PL, Feng FY, Spratt DE
    2020 Dec;23(4):646-653. doi: 10.1038/s41391-020-0226-2. Epub 2020 Mar 30.
    • More

      BACKGROUND: Prostate cancer exhibits biological and clinical heterogeneity even within established clinico-pathologic risk groups. The Decipher genomic classifier (GC) is a validated method to further risk-stratify disease in patients with prostate cancer, but its performance solely within National Comprehensive Cancer Network (NCCN) high-risk disease has not been undertaken to date.

      METHODS: A multi-institutional retrospective study of 405 men with high-risk prostate cancer who underwent primary treatment with radical prostatectomy (RP) or radiation therapy (RT) with androgen-deprivation therapy (ADT) at 11 centers from 1995 to 2005 was performed. Cox proportional hazards models were used to determine the hazard ratios (HR) for the development of metastatic disease based on clinico-pathologic variables, risk groups, and GC score. The area under the receiver operating characteristic curve (AUC) was determined for regression models without and with the GC score.

      RESULTS: Over a median follow-up of 82 months, 104 patients (26%) developed metastatic disease. On univariable analysis, increasing GC score was significantly associated with metastatic disease ([HR]: 1.34 per 0.1 unit increase, 95% confidence interval [CI]: 1.19-1.50, p < 0.001), while age, serum PSA, biopsy GG, and clinical T-stage were not (all p > 0.05). On multivariable analysis, GC score (HR: 1.33 per 0.1 unit increase, 95% CI: 1.19-1.48, p < 0.001) and GC high-risk (vs low-risk, HR: 2.95, 95% CI: 1.79-4.87, p < 0.001) were significantly associated with metastasis. The addition of GC score to regression models based on NCCN risk group improved model AUC from 0.46 to 0.67, and CAPRA from 0.59 to 0.71.

      CONCLUSIONS: Among men with high-risk prostate cancer, conventional clinico-pathologic data had poor discrimination to risk stratify development of metastatic disease. GC score was a significant and independent predictor of metastasis and may help identify men best suited for treatment intensification/de-escalation.

      PMID:32231245 | PMC:PMC10184170 | DOI:10.1038/s41391-020-0226-2


      View details for PubMedID 32231245
  • Genomic and clinical characterization of stromal infiltration markers in prostate cancer Cancer
    Mahal BA, Alshalalfa M, Zhao SG, Beltran H, Chen WS, Chipidza F, Davicioni E, Karnes RJ, Ku S, Lotan TL, Muralidhar V, Rebbeck TR, Schaeffer EM, Spratt DE, Feng FY, Nguyen PL
    2020 Apr 1;126(7):1407-1412. doi: 10.1002/cncr.32688. Epub 2020 Jan 6.
    • More

      BACKGROUND: The progression of prostate cancer is a complex, multistep process that involves molecular alterations in cells of the tumor and the microenvironment, with associated interactions between the stroma and epithelium. Genomic expression analyses of stromal infiltration markers were performed to determine the significance thereof in prostate cancer.

      METHODS: Genome-wide expression profiles of formalin-fixed, paraffin-embedded radical prostatectomy samples were evaluated from a prospective registry cohort (n = 5239) and 3 retrospective institutional cohorts (n = 1135). Two independent stromal gene expression signatures implied stromal infiltration. Cox proportional hazards regression defined the association between stromal infiltration expression and metastasis-free survival (MFS).

      RESULTS: Stromal expression scores were correlated with stromal signature genes and with other key stromal markers (CAV1, VIM, and TAGLN), basal activity, and CD3 and CD4 immune biomarkers (r > 0.5 for all). The top decile of stromal expression was associated with high genomic risk scores (Decipher ≥ 0.6) , high Cancer of the Prostate Risk Assessment-Postsurgical scores, Gleason 9 to 10 disease, and a higher risk for metastasis (hazard ratio, 2.35; 95% CI, 1.37-4.02; P = .001). A higher stromal infiltration score was also associated with decreased expression of DNA repair genes and higher radiation sensitivity genomic scores. Postoperative radiation therapy (RT) was associated with an MFS benefit for patients with high stromal scores, but not for patients with low stromal scores (Pinteraction = .02).

      CONCLUSIONS: Expression of stromal infiltration markers is correlated with prostate cancer aggressiveness/progression and may be predictive of a response to RT. Stromal infiltration markers should be studied and considered for incorporation into clinical prognostication and decision making.

      PMID:31905251 | PMC:PMC7332205 | DOI:10.1002/cncr.32688


      View details for PubMedID 31905251
  • Prostate cancer incidence across stage, NCCN risk groups, and age before and after USPSTF Grade D recommendations against prostate-specific antigen screening in 2012 Cancer
    Butler SS, Muralidhar V, Zhao SG, Sanford NN, Franco I, Fullerton ZH, Chavez J, D'Amico AV, Feng FY, Rebbeck TR, Nguyen PL, Mahal BA
    2020 Feb 15;126(4):717-724. doi: 10.1002/cncr.32604. Epub 2019 Dec 3.
    • More

      BACKGROUND: We sought to determine the extent to which US Preventive Services Task Force (USPSTF) 2012 Grade D recommendations against prostate-specific antigen screening may have impacted recent prostate cancer disease incidence patterns in the United States across stage, National Comprehensive Cancer Network (NCCN) risk groups, and age groups.

      METHODS: SEER*Stat version 8.3.4 was used to calculate annual prostate cancer incidence rates from 2010 to 2015 for men aged ≥50 years according to American Joint Committee on Cancer stage at diagnosis (localized vs metastatic), NCCN risk group (low vs unfavorable [intermediate or high-risk]), and age group (50-74 years vs ≥75 years). Age-adjusted incidences per 100,000 persons with corresponding year-by-year incidence ratios (IRs) were calculated using the 2000 US Census population.

      RESULTS: From 2010 to 2015, the incidence (per 100,000 persons) of localized prostate cancer decreased from 195.4 to 131.9 (Ptrend < .001) and from 189.0 to 123.4 (Ptrend < .001) among men aged 50-74 and ≥75 years, respectively. The largest relative year-by-year decline occurred between 2011 and 2012 in NCCN low-risk disease (IR, 0.77 [0.75-0.79, P < .0001] and IR 0.68 [0.62-0.74, P < .0001] for men aged 50-74 and ≥75 years, respectively). From 2010-2015, the incidence of metastatic disease increased from 6.2 to 7.1 (Ptrend < .001) and from 16.8 to 22.6 (Ptrend < .001) among men aged 50-74 and ≥75 years, respectively.

      CONCLUSIONS: This report illustrates recent prostate cancer "reverse migration" away from indolent disease and toward more aggressive disease beginning in 2012. The incidence of localized disease declined across age groups from 2012 to 2015, with the greatest relative declines occurring in low-risk disease. Additionally, the incidence of distant metastatic disease increased gradually throughout the study period.

      PMID:31794057 | DOI:10.1002/cncr.32604


      View details for PubMedID 31794057
  • Germline polymorphisms associated with impaired survival outcomes and somatic tumor alterations in advanced prostate cancer Prostate cancer and prostatic diseases
    Chen WS, Feng EL, Aggarwal R, Foye A, Beer TM, Alumkal JJ, Gleave M, Chi KN, Reiter RE, Rettig MB, Evans CP, Small EJ, Sharifi N, Zhao SG
    2020 Jun;23(2):316-323. doi: 10.1038/s41391-019-0188-4. Epub 2019 Nov 19.
    • More

      INTRODUCTION: Germline variants in androgen metabolism genes may influence clinical response to androgen deprivation therapy (ADT) in advanced prostate cancer. We sought to investigate the prognostic significance of germline variants in androgen metabolism genes with respect to overall survival (OS) after ADT, and to associate germline variants with tumor genomic features.

      METHODS: Germline and somatic whole-genome sequencing (WGS) data were evaluated in a cohort of 101 men with metastatic castration-resistant prostate cancer (mCRPC). Survival analyses were performed to identify polymorphisms associated with impaired OS after primary ADT. Germline variants found to be prognostic of OS were associated with tumor somatic DNA-sequence alterations based on WGS performed on paired metastasis biopsies from the same 101 patients. Gene set enrichment analysis was performed based on tumor RNA-sequencing data to identify genomic pathways differentially expressed in patients with germline variants.

      RESULTS: A comprehensive literature review identified 17 candidate polymorphisms in nine androgen metabolism genes that have been previously shown to have an association with response to ADT in prostate cancer. Of these, the variant rs1856888 allele located 13 kb upstream of HSD3B1 was found to be significantly associated with impaired OS (P = 0.029). Variant rs1856888 was commonly co-inherited with the well-characterized HSD3B1(1245A>C) polymorphism, and there was a trend toward shorter median OS in patients with HSD3B1(1245A>C) compared with homozygous wild-type patients (P = 0.052). While HSD3B1 germline variants were not associated with common somatic tumor DNA alterations, they were associated with increased tumor expression of cell proliferation and cell cycle genes.

      CONCLUSIONS: This study presents a comprehensive assessment of germline variants in androgen metabolism genes and highlights HSD3B1 polymorphisms as prognostic of OS after ADT and associated with an aggressive gene expression tumor profile in mCRPC.

      PMID:31745256 | PMC:PMC7529063 | DOI:10.1038/s41391-019-0188-4


      View details for PubMedID 31745256
  • Clinicogenomic Radiotherapy Classifier Predicting the Need for Intensified Locoregional Treatment After Breast-Conserving Surgery for Early-Stage Breast Cancer Journal of clinical oncology : official journal of the American Society of Clinical Oncology
    Sjöström M, Chang SL, Fishbane N, Davicioni E, Zhao SG, Hartman L, Holmberg E, Feng FY, Speers CW, Pierce LJ, Malmström P, Fernö M, Karlsson P
    2019 Dec 10;37(35):3340-3349. doi: 10.1200/JCO.19.00761. Epub 2019 Oct 16.
    • More

      PURPOSE: Most patients with early-stage breast cancer are treated with adjuvant radiotherapy (RT) after breast-conserving surgery (BCS) to prevent locoregional recurrence (LRR). However, no genomic tools are used currently to select the optimal RT strategy.

      METHODS: We profiled the transcriptome of primary tumors on a clinical grade assay from the SweBCG91-RT trial, in which patients with node-negative breast cancer were randomly assigned to either whole-breast RT after BCS or no RT. We derived a new classifier, Adjuvant Radiotherapy Intensification Classifier (ARTIC), comprising 27 genes and patient age, in three publicly available cohorts, then independently validated ARTIC for LRR in 748 patients in SweBCG91-RT. We also compared previously published genomic signatures for ability to predict benefit from RT in SweBCG91-RT.

      RESULTS: ARTIC was highly prognostic for LRR in patients treated with RT (hazard ratio [HR], 3.4; 95% CI, 2.0 to 5.9; P < .001) and predictive of RT benefit (Pinteraction = .005). Patients with low ARTIC scores had a large benefit from RT (HR, 0.33 [95% CI, 0.21 to 0.52], P < .001; 10-year cumulative incidence of LRR, 6% v 21%), whereas those with high ARTIC scores benefited less from RT (HR, 0.73 [95% CI, 0.44 to 1.2], P = .23; 10-year cumulative incidence of LRR, 25% v 32%). In contrast, none of the eight previously published signatures were predictive of benefit from RT in SweBCG91-RT.

      CONCLUSION: ARTIC identified women with a substantial benefit from RT as well as women with a particularly elevated LRR risk in whom whole-breast RT was not sufficiently effective and, thus, in whom intensified treatment strategies such as tumor-bed boost, and possibly regional nodal RT, should be considered. To our knowledge, ARTIC is the first classifier validated as predictive of benefit from RT in a phase III clinical trial with patients randomly assigned to receive or not receive RT.

      PMID:31618132 | PMC:PMC6901281 | DOI:10.1200/JCO.19.00761


      View details for PubMedID 31618132
  • Transcriptomic Heterogeneity of Androgen Receptor Activity Defines a <em>de novo</em> low AR-Active Subclass in Treatment Naïve Primary Prostate Cancer Clinical cancer research : an official journal of the American Association for Cancer Research
    Spratt DE, Alshalalfa M, Fishbane N, Weiner AB, Mehra R, Mahal BA, Lehrer J, Liu Y, Zhao SG, Speers C, Morgan TM, Dicker AP, Freedland SJ, Karnes RJ, Weinmann S, Davicioni E, Ross AE, Den RB, Nguyen PL, Feng FY, Lotan TL, Chinnaiyan AM, Schaeffer EM
    2019 Nov 15;25(22):6721-6730. doi: 10.1158/1078-0432.CCR-19-1587. Epub 2019 Sep 12.
    • More

      PURPOSE: The heterogeneity of androgen receptor (AR)-activity (AR-A) is well-characterized in heavily treated metastatic castration-resistant prostate cancer (mCRPC). However, the diversity and clinical implications of AR-A in treatment-naïve primary prostate cancer is largely unknown. We sought to characterize AR-A in localized prostate cancer and understand its molecular and clinical implications.

      EXPERIMENTAL DESIGN: Genome-wide expression profiles from prostatectomy or biopsy samples from 19,470 patients were used, all with independent pathology review. This was comprised of prospective discovery (n = 5,239) and validation (n = 12,728) cohorts, six retrospective institutional cohorts with long-term clinical outcomes data (n = 1,170), and The Cancer Genome Atlas (n = 333).

      RESULTS: A low AR-active subclass was identified, which comprised 9%-11% of each cohort, and was characterized by increased immune signaling, neuroendocrine expression, and decreased DNA repair. These tumors were predominantly ERG and basal subtype. Low AR-active tumors had significantly more rapid development of recurrence or metastatic disease across cohorts, which was maintained on multivariable analysis [HR, 2.61; 95% confidence interval (CI), 1.22-5.60; P = 0.014]. Low AR-active tumors were predicted to be more sensitive to PARP inhibition, platinum chemotherapy, and radiotherapy, and less sensitive to docetaxel and androgen-deprivation therapy. This was validated clinically, in that low AR-active tumors were less sensitive to androgen-deprivation therapy (OR, 0.41; 95% CI, 0.21-0.80; P = 0.008).

      CONCLUSIONS: Leveraging large-scale transcriptomic data allowed the identification of an aggressive subtype of treatment-naïve primary prostate cancer that harbors molecular features more analogous to mCRPC. This suggests that a preexisting subgroup of patients may have tumors that are predisposed to fail multiple current standard-of-care therapies and warrant dedicated therapeutic investigation.

      PMID:31515456 | PMC:PMC6858964 | DOI:10.1158/1078-0432.CCR-19-1587


      View details for PubMedID 31515456
  • DNA-Dependent Protein Kinase Drives Prostate Cancer Progression through Transcriptional Regulation of the Wnt Signaling Pathway Clinical cancer research : an official journal of the American Association for Cancer Research
    Kothari V, Goodwin JF, Zhao SG, Drake JM, Yin Y, Chang SL, Evans JR, Wilder-Romans K, Gabbara K, Dylgjeri E, Chou J, Sun G, Tomlins SA, Mehra R, Hege K, Filvaroff EH, Schaeffer EM, Karnes RJ, Quigley DA, Rathkopf DE, He HH, Speers C, Spratt DE, Gilbert LA, Ashworth A, Chinnaiyan AM, Raj GV, Knudsen KE, Feng FY
    2019 Sep 15;25(18):5608-5622. doi: 10.1158/1078-0432.CCR-18-2387. Epub 2019 Jul 2.
    • More

      PURPOSE: Protein kinases are known to play a prominent role in oncogenic progression across multiple cancer subtypes, yet their role in prostate cancer progression remains underexplored. The purpose of this study was to identify kinases that drive prostate cancer progression.Experimental Design: To discover kinases that drive prostate cancer progression, we investigated the association between gene expression of all known kinases and long-term clinical outcomes in tumor samples from 545 patients with high-risk disease. We evaluated the impact of genetic and pharmacologic inhibition of the most significant kinase associated with metastatic progression in vitro and in vivo.

      RESULTS: DNA-dependent protein kinase (DNAPK) was identified as the most significant kinase associated with metastatic progression in high-risk prostate cancer. Inhibition of DNAPK suppressed the growth of both AR-dependent and AR-independent prostate cancer cells. Gene set enrichment analysis nominated Wnt as the top pathway associated with DNAPK. We found that DNAPK interacts with the Wnt transcription factor LEF1 and is critical for LEF1-mediated transcription.

      CONCLUSIONS: Our data show that DNAPK drives prostate cancer progression through transcriptional regulation of Wnt signaling and is an attractive therapeutic target in aggressive prostate cancer.

      PMID:31266829 | PMC:PMC6744969 | DOI:10.1158/1078-0432.CCR-18-2387


      View details for PubMedID 31266829
  • Conservative management of low-risk prostate cancer among young versus older men in the United States: Trends and outcomes from a novel national database Cancer
    Mahal AR, Butler S, Franco I, Muralidhar V, Larios D, Pike RG, Zhao SG, Sanford NN, Dess RT, Feng FY, D'Amico AV, Spratt DE, Yu JB, Nguyen PL, Rebbeck TR, Mahal BA
    2019 Oct 1;125(19):3338-3346. doi: 10.1002/cncr.32332. Epub 2019 Jun 28.
    • More

      BACKGROUND: Management for men aged ≤55 years with low-risk prostate cancer (LRPC) is debated given quality-of-life implications with definitive treatment versus the potential missed opportunity for cure with conservative management. The objective of this study was to define rates of conservative management for LRPC and associated short-term outcomes in young versus older men in the United States.

      METHODS: The nonpublic Surveillance, Epidemiology, and End Results Prostate with Active Surveillance/Watchful Waiting (AS/WW) Database identified 50,302 men who were diagnosed with LRPC from 2010 through 2015. AS/WW rates in the United States were stratified by age (≤55 vs ≥56 years). Prostate cancer-specific mortality and overall mortality were defined by initial management type (AS/WW vs definitive treatment [referent]) and age.

      RESULTS: AS/WW utilization increased from 8.61% (2010) to 34.56% (2015) among men aged ≤55 years (P for trend <0.001) and from 15.99% to 43.81% among men aged ≥56 years (P for trend <.001). Among patients who had ≤2 positive biopsy cores, AS/WW rates increased from 12.90% to 48.78% for men aged ≤55 years and from 21.85% to 58.01% for men aged ≥56 years. Among patients who had ≥3 positive biopsy cores, AS/WW rates increased from 3.89% to 22.45% for men aged ≤55 years and from 10.05% to 28.49% for men aged ≥56 years (all P for trend <.001). Five-year prostate cancer-specific mortality rates were <0.30% across age and initial management type subgroups.

      CONCLUSIONS: AS/WW rates quadrupled for patients aged ≤55 years from 2010 to 2015, with favorable short-term outcomes. These findings demonstrate the short-term safety and increasing acceptance of AS/WW for both younger and older patients. However, there are still higher absolute rates of AS/WW in older patients (P < .001), suggesting some national ambivalence toward AS/WW in younger patients.

      PMID:31251398 | DOI:10.1002/cncr.32332


      View details for PubMedID 31251398
  • Transcriptomic and Clinical Characterization of Neuropeptide Y Expression in Localized and Metastatic Prostate Cancer: Identification of Novel Prostate Cancer Subtype with Clinical Implications European urology oncology
    Alshalalfa M, Nguyen PL, Beltran H, Chen WS, Davicioni E, Zhao SG, Rebbeck TR, Schaeffer EM, Lotan TL, Feng FY, Mahal BA
    2019 Jul;2(4):405-412. doi: 10.1016/j.euo.2019.05.001. Epub 2019 Jun 2.
    • More

      BACKGROUND: Tumor microenvironment and its interaction with neuroendocrine modulators contribute to prostate carcinogenesis and progression.

      OBJECTIVE: We sought to define the transcriptomic and clinical implications of neuropeptide Y (NPY) expression in prostate cancer progression.

      DESIGN, SETTING, AND PARTICIPANTS: Genome-wide expression profiling of three localized prostate cancer (total n=18818) and five metastatic castrate-resistant prostate cancer (mCRPC; total n=495) cohorts was used to characterize NPY expression. All men underwent radical prostatectomy (RP) for localized prostate cancer.

      OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Patients were grouped into those with low NPY and high NPY based on NPY expression. Associations between these groups and histological, genomic, and clinical outcomes including progression-free survival (PFS) and metastases-free survival (MFS) were examined. Combining ERG-fusion status with NPY expression, four groups were defined (lowNPY/ERG+, lowNPY/ERG-, highNPY/ERG+, and highNPY/ERG-). Cox proportional hazards modeled the time to distant metastasis after RP. Genomic risk scores for metastasis were calculated for prospective samples, based on a 22-gene signature.

      RESULTS AND LIMITATIONS: Across cancers, NPY showed the highest expression in prostate cancer in The Cancer Genome Atlas (TCGA) PAN-Cancer cohort (n=9483, p<0.0001). In 17967 prospective samples, low NPY expression was associated with aggressive grade group 5 disease and a higher genomic risk (p<0.0001). In the retrospective (n=355) and TCGA (n=497) cohorts, low NPY was associated with shorter MFS and PFS, respectively (p=0.001 for both). In mCRPC cohorts, low NPY was associated with neuroendocrine development (p<0.01). NPY was highly correlated to ERG; thus, we defined four groups based on NPY expression and ERG fusion. The lowNPY/ERG+ subtype was associated with the highest genomic risk for metastasis (p<0.0001) and the highest rate of metastasis compared with all other subtypes (hazard ratio [HR]: 2.2 [1.22-4.03], p=0.008), while the highNPY/ERG- subtype was associated with the lowest genomic risk for metastasis (p<0.0001) and the lowest rate of metastasis (HR: 0.53 [0.35-0.81], p=0.003).

      CONCLUSIONS: Low NPY expression is associated with adverse genomic features and clinical correlates and outcomes. The lowNPY/ERG+ subtype was associated with the highest risk of developing metastasis. Prognostic subgrouping and tailored treatments by NPY expression and ERG fusion status warrant further study.

      PATIENT SUMMARY: The low neuropeptide Y prostate cancer subtype appears to be aggressive with a high risk of developing metastasis.

      PMID:31164324 | PMC:PMC7597937 | DOI:10.1016/j.euo.2019.05.001


      View details for PubMedID 31164324
  • Xenograft-based, platform-independent gene signatures to predict response to alkylating chemotherapy, radiation, and combination therapy for glioblastoma Neuro-oncology
    Zhao SG, Yu M, Spratt DE, Chang SL, Feng FY, Kim MM, Speers CW, Carlson BL, Mladek AC, Lawrence TS, Sarkaria JN, Wahl DR
    2019 Sep 6;21(9):1141-1149. doi: 10.1093/neuonc/noz090.
    • More

      BACKGROUND: Predictive molecular biomarkers to select optimal treatment for patients with glioblastoma and other cancers are lacking. New strategies are needed when large randomized trials with correlative molecular data are not feasible.

      METHODS: Gene signatures (GS) were developed from 31 orthotopic glioblastoma patient-derived xenografts (PDXs), treated with standard therapies, to predict benefit from radiotherapy (RT-GS), temozolomide (Chemo-GS), or the combination (ChemoRT-GS). Independent validation was performed in a heterogeneously treated clinical cohort of 502 glioblastoma patients with overall survival as the primary endpoint. Multivariate Cox analysis was used to adjust for confounding variables and evaluate interactions between signatures and treatment.

      RESULTS: PDX models recapitulated the clinical heterogeneity of glioblastoma patients. RT-GS, Chemo-GS, and ChemoRT-GS were correlated with benefit from treatment in the PDX models. In independent clinical validation, higher RT-GS scores were associated with increased survival only in patients receiving RT (P = 0.0031, hazard ratio [HR] = 0.78 [0.66-0.92]), higher Chemo-GS scores were associated with increased survival only in patients receiving chemotherapy (P < 0.0001, HR = 0.66 [0.55-0.8]), and higher ChemoRT-GS scores were associated with increased survival only in patients receiving ChemoRT (P = 0.0001, HR = 0.54 [0.4-0.74]). RT-GS and ChemoRT-GS had significant interactions with treatment on multivariate analysis (P = 0.0009 and 0.02, respectively), indicating that they are bona fide predictive biomarkers.

      CONCLUSIONS: Using a novel PDX-driven methodology, we developed and validated 3 platform-independent molecular signatures that predict benefit from standard of care therapies for glioblastoma. These signatures may be useful to personalize glioblastoma treatment in the clinic and this approach may be a generalizable method to identify predictive biomarkers without resource-intensive randomized trials.

      PMID:31121035 | PMC:PMC6736132 | DOI:10.1093/neuonc/noz090


      View details for PubMedID 31121035
  • Active Surveillance for Low-Risk Prostate Cancer in Black Patients The New England journal of medicine
    Butler S, Muralidhar V, Chavez J, Fullerton Z, Mahal A, Nezolosky M, Vastola M, Zhao SG, D'Amico AV, Dess RT, Feng FY, King MT, Mouw KW, Spratt DE, Trinh Q, Nguyen PL, Rebbeck TR, Mahal BA
    2019 May 23;380(21):2070-2072. doi: 10.1056/NEJMc1900333.
  • Impact of Biochemical Failure After Salvage Radiation Therapy on Prostate Cancer-specific Mortality: Competition Between Age and Time to Biochemical Failure European urology oncology
    Jackson WC, Suresh K, Tumati V, Dess RT, Soni PD, Zhao SG, Zumsteg ZS, Hannan R, Hollenbeck BK, George A, Kaffenberger SD, Salami SS, Hearn WD, Morgan TM, Mehra R, Schipper M, Feng FY, Desai NB, Spratt DE
    2018 Sep;1(4):276-282. doi: 10.1016/j.euo.2018.04.014. Epub 2018 Sep 28.
    • More

      BACKGROUND: Disease progression following salvage radiotherapy (SRT) for prostate cancer (PC) is common, and the time to biochemical recurrence (BCR) is heterogeneous.

      OBJECTIVE: To describe the temporal distribution and clinical impact of BCR following SRT and model outcomes using patient age and time to BCR from SRT.

      DESIGN, SETTING, AND PARTICIPANTS: A retrospective multi-institutional study included 547 consecutive men with lymph node-negative PC receiving SRT from 1985 to 2013. The median follow-up after SRT was 8.4 yr. Intervention All men received SRT with three-dimensional or intensity-modulated RT.

      OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: BCR was defined as a rise in prostate-specific antigen (PSA) ≥0.2ng/ml above the PSA nadir followed by a sequentially equal or higher value. Additional outcomes included distant metastasis (DM), PC-specific mortality (PCSM), and overall mortality (OM). Cox proportional hazards models, a landmark analysis, and comparison of c-indices were used. Cumulative incidence curves were estimated from a Fine and Gray regression model.

      RESULTS AND LIMITATIONS: The estimated 10-yr cumulative incidence of BCR was 60%. Of the 274 men experiencing BCR, 149 (54%) had BCR within 18 mo of SRT. BCR ≤18 mo after SRT was associated with a higher risk of DM (hazard ratio [HR] 7.44, 95% confidence interval [CI] 4.91-11.3; p<0.001), PCSM (HR 12.3, 95% CI 5.95-25.2; p<0.001), and OM (HR 2.85, 95% CI 1.94-4.17; p<0.001). We provide a model to estimate the cumulative incidence of DM and PCSM using age and time to BCR. The retrospective nature of our analysis limits our findings.

      CONCLUSIONS: A strikingly large proportion of men experience early BCR following SRT and are at higher risk of DM and PCSM. Novel predictive biomarkers are needed to identify men harboring micrometastatic disease to avoid potentially futile local therapies or allow for intensification of systemic therapies.

      PATIENT SUMMARY: Many men will develop biochemical recurrence of prostate cancer after salvage radiotherapy. Men with biochemical recurrence within 18 mo of salvage radiotherapy constitute a cohort at higher risk of distant metastasis and prostate cancer-specific mortality.

      PMID:31100248 | DOI:10.1016/j.euo.2018.04.014


      View details for PubMedID 31100248
  • Novel RB1-Loss Transcriptomic Signature Is Associated with Poor Clinical Outcomes across Cancer Types Clinical cancer research : an official journal of the American Association for Cancer Research
    Chen WS, Alshalalfa M, Zhao SG, Liu Y, Mahal BA, Quigley DA, Wei T, Davicioni E, Rebbeck TR, Kantoff PW, Maher CA, Knudsen KE, Small EJ, Nguyen PL, Feng FY
    2019 Jul 15;25(14):4290-4299. doi: 10.1158/1078-0432.CCR-19-0404. Epub 2019 Apr 22.
    • More

      PURPOSE: Rb-pathway disruption is of great clinical interest, as it has been shown to predict outcomes in multiple cancers. We sought to develop a transcriptomic signature for detecting biallelic RB1 loss (RBS) that could be used to assess the clinical implications of RB1 loss on a pan-cancer scale.

      EXPERIMENTAL DESIGN: We utilized data from the Cancer Cell Line Encyclopedia (N = 995) to develop the first pan-cancer transcriptomic signature for predicting biallelic RB1 loss (RBS). Model accuracy was validated using The Cancer Genome Atlas (TCGA) Pan-Cancer dataset (N = 11,007). RBS was then used to assess the clinical relevance of biallelic RB1 loss in TCGA Pan-Cancer and in an additional metastatic castration-resistant prostate cancer (mCRPC) cohort.

      RESULTS: RBS outperformed the leading existing signature for detecting RB1 biallelic loss across all cancer types in TCGA Pan-Cancer (AUC, 0.89 vs. 0.66). High RBS (RB1 biallelic loss) was associated with promoter hypermethylation (P = 0.008) and gene body hypomethylation (P = 0.002), suggesting RBS could detect epigenetic gene silencing. TCGA Pan-Cancer clinical analyses revealed that high RBS was associated with short progression-free (P < 0.00001), overall (P = 0.0004), and disease-specific (P < 0.00001) survival. On multivariable analyses, high RBS was predictive of shorter progression-free survival in TCGA Pan-Cancer (P = 0.03) and of shorter overall survival in mCRPC (P = 0.004) independently of the number of DNA alterations in RB1.

      CONCLUSIONS: Our study provides the first validated tool to assess RB1 biallelic loss across cancer types based on gene expression. RBS can be useful for analyzing datasets with or without DNA-sequencing results to investigate the emerging prognostic and treatment implications of Rb-pathway disruption.See related commentary by Choudhury and Beltran, p. 4199.

      PMID:31010837 | PMC:PMC7883384 | DOI:10.1158/1078-0432.CCR-19-0404


      View details for PubMedID 31010837
  • Genomic Drivers of Poor Prognosis and Enzalutamide Resistance in Metastatic Castration-resistant Prostate Cancer European urology
    Chen WS, Aggarwal R, Zhang L, Zhao SG, Thomas GV, Beer TM, Quigley DA, Foye A, Playdle D, Huang J, Lloyd P, Lu E, Sun D, Guan X, Rettig M, Gleave M, Evans CP, Youngren J, True L, Lara P, Kothari V, Xia Z, Chi KN, Reiter RE, Maher CA, Feng FY, Small EJ, Alumkal JJ, Team CD
    2019 Nov;76(5):562-571. doi: 10.1016/j.eururo.2019.03.020. Epub 2019 Mar 28.
    • More

      BACKGROUND: Metastatic castration-resistant prostate cancer (mCRPC) is the lethal form of the disease. Several recent studies have identified genomic alterations in mCRPC, but the clinical implications of these genomic alterations have not been fully elucidated.

      OBJECTIVE: To use whole-genome sequencing (WGS) to assess the association between key driver gene alterations and overall survival (OS), and to use whole-transcriptome RNA sequencing to identify genomic drivers of enzalutamide resistance.

      DESIGN, SETTING, AND PARTICIPANTS: We performed survival analyses and gene set enrichment analysis (GSEA) on WGS and RNA sequencing results for a cohort of 101 mCRPC patients.

      OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: OS was the clinical endpoint for all univariate and multivariable survival analyses. Candidate drivers of enzalutamide resistance were identified in an unbiased manner, and mutations of the top candidate were further assessed for enrichment among enzalutamide-resistant patients using Fisher's exact test.

      RESULTS AND LIMITATIONS: Harboring two DNA alterations in RB1 was independently predictive of poor OS (median 14.1 vs 42.0mo; p=0.007) for men with mCRPC. GSEA identified the Wnt/β-catenin pathway as the top differentially modulated pathway among enzalutamide-resistant patients. Furthermore, β-catenin mutations were exclusive to enzalutamide-resistant patients (p=0.01) and independently predictive of poor OS (median 13.6 vs 41.7mo; p=0.025).

      CONCLUSIONS: The presence of two RB1 DNA alterations identified in our WGS analysis was independently associated with poor OS among men with mCRPC. The Wnt/β-catenin pathway plays an important role in enzalutamide resistance, with differential pathway expression and enrichment of β-catenin mutations in enzalutamide-resistant patients. Moreover, β-catenin mutations were predictive of poor OS in our cohort.

      PATIENT SUMMARY: We observed a correlation between genomic findings for biopsy samples from metastases from men with metastatic castration-resistant prostate cancer (mCRPC) and clinical outcomes. This work sheds new light on clinically relevant genomic alterations in mCRPC and provides a roadmap for the development of new personalized treatment regimens in mCRPC.

      PMID:30928160 | PMC:PMC6764911 | DOI:10.1016/j.eururo.2019.03.020


      View details for PubMedID 30928160
  • MEK-ERK signaling is a therapeutic target in metastatic castration resistant prostate cancer Prostate cancer and prostatic diseases
    Nickols NG, Nazarian R, Zhao SG, Tan V, Uzunangelov V, Xia Z, Baertsch R, Neeman E, Gao AC, Thomas GV, Howard L, Hoedt MD, Stuart J, Goldstein T, Chi K, Gleave ME, Graff JN, Beer TM, Drake JM, Evans CP, Aggarwal R, Foye A, Feng FY, Small EJ, Aronson WJ, Freedland SJ, Witte ON, Huang J, Alumkal JJ, Reiter RE, Rettig MB
    2019 Dec;22(4):531-538. doi: 10.1038/s41391-019-0134-5. Epub 2019 Feb 25.
    • More

      BACKGROUND: Metastatic castration resistant prostate cancer (mCRPC) is incurable and progression after drugs that target the androgen receptor-signaling axis is inevitable. Thus, there is an urgent need to develop more effective treatments beyond hormonal manipulation. We sought to identify activated kinases in mCRPC as therapeutic targets for existing, approved agents, with the goal of identifying candidate drugs for rapid translation into proof of concept Phase II trials in mCRPC.

      METHODS: To identify evidence of activation of druggable kinases in these patients, we compared mRNA expression from metastatic biopsies of patients with mCRPC (n = 101) to mRNA expression in localized prostate from TCGA and used this analysis to infer differential kinase activity. In addition, we assessed the differential phosphorylation levels for key MAPK pathway kinases between mCRPC and localized prostate cancers.

      RESULTS: Transcriptomic profiling of 101 patients with mCRPC as compared to patients with localized prostate cancer identified evidence of hyperactive ERK1, and whole genome sequencing revealed frequent amplifications of members of the MAPK pathway in 32% of this cohort. Next, we confirmed elevated levels of phosphorylated ERK1/2 in castration resistant prostate cancer as compared to untreated primary prostate cancer. We observed that the presence of detectable phosphorylated ERK1/2 in the primary tumor is associated with biochemical failure after radical prostatectomy independent of clinicopathologic features. ERK1 is the immediate downstream target of MEK1/2, which is druggable with trametinib, an approved therapeutic for melanoma. Trametinib elicited a profound biochemical and clinical response in a patient who had failed multiple prior treatments for mCRPC.

      CONCLUSIONS: We conclude that pharmacologic targeting of the MEK/ERK pathway may be a viable treatment strategy for patients with refractory metastatic prostate cancer. An ongoing Phase II trial tests this hypothesis.

      PMID:30804427 | PMC:PMC6853839 | DOI:10.1038/s41391-019-0134-5


      View details for PubMedID 30804427
  • Prostate Cancer Genomic-risk Differences Between African-American and White Men Across Gleason Scores European urology
    Mahal BA, Alshalalfa M, Spratt DE, Davicioni E, Zhao SG, Feng FY, Rebbeck TR, Nguyen PL, Huang FW
    2019 Jun;75(6):1038-1040. doi: 10.1016/j.eururo.2019.01.010. Epub 2019 Jan 22.
  • Clinical and Genomic Implications of Luminal and Basal Subtypes Across Carcinomas Clinical cancer research : an official journal of the American Association for Cancer Research
    Zhao SG, Chen WS, Das R, Chang SL, Tomlins SA, Chou J, Quigley DA, Dang HX, Barnard TJ, Mahal BA, Gibb EA, Liu Y, Davicioni E, Duska LR, Posadas EM, Jolly S, Spratt DE, Nguyen PL, Maher CA, Small EJ, Feng FY
    2019 Apr 15;25(8):2450-2457. doi: 10.1158/1078-0432.CCR-18-3121. Epub 2018 Dec 20.
    • More

      PURPOSE: Carcinomas originate from epithelial tissues, which have apical (luminal) and basal orientations. The degree of luminal versus basal differentiation in cancer has been shown to be biologically important in some carcinomas and impacts treatment response.

      EXPERIMENTAL DESIGN: Although prior studies have focused on individual cancer types, we used a modified clinical-grade classifier (PAM50) to subtype 8,764 tumors across 22 different carcinomas into luminal A, luminal B, and basal-like tumors.

      RESULTS: We found that all epithelial tumors demonstrated similar gene expression-based luminal/basal subtypes. As expected, basal-like tumors were associated with increased expression of the basal markers KRT5/6 and KRT14, and luminal-like tumors were associated with increased expression of the luminal markers KRT20. Luminal A tumors consistently had improved outcomes compared with basal across many tumor types, with luminal B tumors falling between the two. Basal tumors had the highest rates of TP53 and RB1 mutations and copy number loss. Luminal breast, cervical, ovarian, and endometrial tumors had increased ESR1 expression, and luminal prostate, breast, cervical, and bladder tumors had increased androgen receptor (AR) expression. Furthermore, luminal B tumors had the highest rates of AR and ESR1 mutations and had increased sensitivity in vitro to bicalutamide and tamoxifen. Luminal B tumors were more sensitive to gemcitabine, and basal tumors were more sensitive to docetaxel.

      CONCLUSIONS: This first pan-carcinoma luminal/basal subtyping across epithelial tumors reveals global similarities across carcinomas in the transcriptome, genome, clinical outcomes, and drug sensitivity, emphasizing the biological and translational importance of these luminal versus basal subtypes.

      PMID:30573691 | DOI:10.1158/1078-0432.CCR-18-3121


      View details for PubMedID 30573691
  • PARP-1 regulates DNA repair factor availability EMBO molecular medicine
    Schiewer MJ, Mandigo AC, Gordon N, Huang F, Gaur S, Leeuw Rd, Zhao SG, Evans J, Han S, Parsons T, Birbe R, McCue P, McNair C, Chand SN, Cendon-Florez Y, Gallagher P, McCann JJ, Neupane NP, Shafi AA, Dylgjeri E, Brand LJ, Visakorpi T, Raj GV, Lallas CD, Trabulsi EJ, Gomella LG, Dicker AP, Kelly WK, Leiby BE, Knudsen B, Feng FY, Knudsen KE
    2018 Dec;10(12):e8816. doi: 10.15252/emmm.201708816.
    • More

      PARP-1 holds major functions on chromatin, DNA damage repair and transcriptional regulation, both of which are relevant in the context of cancer. Here, unbiased transcriptional profiling revealed the downstream transcriptional profile of PARP-1 enzymatic activity. Further investigation of the PARP-1-regulated transcriptome and secondary strategies for assessing PARP-1 activity in patient tissues revealed that PARP-1 activity was unexpectedly enriched as a function of disease progression and was associated with poor outcome independent of DNA double-strand breaks, suggesting that enhanced PARP-1 activity may promote aggressive phenotypes. Mechanistic investigation revealed that active PARP-1 served to enhance E2F1 transcription factor activity, and specifically promoted E2F1-mediated induction of DNA repair factors involved in homologous recombination (HR). Conversely, PARP-1 inhibition reduced HR factor availability and thus acted to induce or enhance "BRCA-ness". These observations bring new understanding of PARP-1 function in cancer and have significant ramifications on predicting PARP-1 inhibitor function in the clinical setting.

      PMID:30467127 | PMC:PMC6284389 | DOI:10.15252/emmm.201708816


      View details for PubMedID 30467127
  • The Immune Landscape of Prostate Cancer and Nomination of PD-L2 as a Potential Therapeutic Target Journal of the National Cancer Institute
    Zhao SG, Lehrer J, Chang SL, Das R, Erho N, Liu Y, Sjöström M, Den RB, Freedland SJ, Klein EA, Karnes RJ, Schaeffer EM, Xu M, Speers C, Nguyen PL, Ross AE, Chan JM, Cooperberg MR, Carroll PR, Davicioni E, Fong L, Spratt DE, Feng FY
    2019 Mar 1;111(3):301-310. doi: 10.1093/jnci/djy141.
    • More

      BACKGROUND: Immunotherapy has been less successful in treating prostate cancer than other solid tumors. We sought to better understand the immune landscape in prostate cancer and identify immune-related biomarkers and potential therapeutic targets.

      METHODS: We analyzed gene expression data from 7826 prospectively collected prostatectomy samples (2013-2016), and 1567 retrospective samples with long-term clinical outcomes, for a total of 9393 samples, all profiled on the same commercial clinical platform in a CLIA-certified lab. The primary outcome was distant metastasis-free survival (DMFS). Secondary outcomes included biochemical recurrence-free survival (bRFS), prostate cancer-specific survival (PCSS), and overall survival (OS). All statistical tests were two-sided.

      RESULTS: Unsupervised hierarchical clustering of hallmark pathways demonstrated an immune-related tumor cluster. Increased estimated immune content scores based on immune-specific genes from the literature were associated with worse bRFS (hazard ratio [HR] = 1.26 [95% confidence interval [CI] = 1.12 to 1.42]; P < .001), DMFS (HR = 1.34 [95% CI = 1.13 to 1.58]; P < .001), PCSS (HR = 1.53 [95% CI = 1.21 to 1.92]; P < .001), and OS (HR = 1.27 [95% CI = 1.07 to 1.50]; P = .006). Deconvolution using Cibersort revealed that mast cells, natural killer cells, and dendritic cells conferred improved DMFS, whereas macrophages and T-cells conferred worse DMFS. Interestingly, while PD-L1 was not prognostic, consistent with its low expression in prostate cancer, PD-L2 was expressed at statistically significantly higher levels (P < .001) and was associated with worse bRFS (HR = 1.17 [95% CI = 1.03 to 1.33]; P = .01), DMFS (HR = 1.25 [95% CI = 1.05 to 1.49]; P = .01), and PCSS (HR = 1.45 [95% CI = 1.13 to 1.86]; P = .003). PD-L2 was strongly associated with immune-related pathways on gene set enrichment analysis suggesting that it is playing an important role in immune modulation in clinical prostate cancer samples. Furthermore, PD-L2 was correlated with radiation response pathways, and also predicted response to postoperative radiation therapy (PORT) on multivariable interaction analysis (P = .03).

      CONCLUSION: In the largest study of its kind to date, these results illustrate the complex relationship between the tumor-immune interaction, prognosis, and response to radiotherapy, and nominate PD-L2 as a potential novel therapeutic target in prostate cancer, potentially in combination with radiotherapy.

      PMID:30321406 | DOI:10.1093/jnci/djy141


      View details for PubMedID 30321406
  • Genomic Hallmarks and Structural Variation in Metastatic Prostate Cancer Cell
    Quigley DA, Dang HX, Zhao SG, Lloyd P, Aggarwal R, Alumkal JJ, Foye A, Kothari V, Perry MD, Bailey AM, Playdle D, Barnard TJ, Zhang L, Zhang J, Youngren JF, Cieslik MP, Parolia A, Beer TM, Thomas G, Chi KN, Gleave M, Lack NA, Zoubeidi A, Reiter RE, Rettig MB, Witte O, Ryan CJ, Fong L, Kim W, Friedlander T, Chou J, Li H, Das R, Li H, Moussavi-Baygi R, Goodarzi H, Gilbert LA, Lara PN, Evans CP, Goldstein TC, Stuart JM, Tomlins SA, Spratt DE, Cheetham RK, Cheng DT, Farh K, Gehring JS, Hakenberg J, Liao A, Febbo PG, Shon J, Sickler B, Batzoglou S, Knudsen KE, He HH, Huang J, Wyatt AW, Dehm SM, Ashworth A, Chinnaiyan AM, Maher CA, Small EJ, Feng FY
    2018 Jul 26;174(3):758-769.e9. doi: 10.1016/j.cell.2018.06.039. Epub 2018 Jul 19.
    • More

      While mutations affecting protein-coding regions have been examined across many cancers, structural variants at the genome-wide level are still poorly defined. Through integrative deep whole-genome and -transcriptome analysis of 101 castration-resistant prostate cancer metastases (109X tumor/38X normal coverage), we identified structural variants altering critical regulators of tumorigenesis and progression not detectable by exome approaches. Notably, we observed amplification of an intergenic enhancer region 624 kb upstream of the androgen receptor (AR) in 81% of patients, correlating with increased AR expression. Tandem duplication hotspots also occur near MYC, in lncRNAs associated with post-translational MYC regulation. Classes of structural variations were linked to distinct DNA repair deficiencies, suggesting their etiology, including associations of CDK12 mutation with tandem duplications, TP53 inactivation with inverted rearrangements and chromothripsis, and BRCA2 inactivation with deletions. Together, these observations provide a comprehensive view of how structural variations affect critical regulators in metastatic prostate cancer.

      PMID:30033370 | PMC:PMC6425931 | DOI:10.1016/j.cell.2018.06.039


      View details for PubMedID 30033370
  • The Diverse Genomic Landscape of Clinically Low-risk Prostate Cancer European urology
    Cooperberg MR, Erho N, Chan JM, Feng FY, Fishbane N, Zhao SG, Simko JP, Cowan JE, Lehrer J, Alshalalfa M, Kolisnik T, Chelliserry J, Margrave J, Aranes M, Plessis Md, Buerki C, Tenggara I, Davicioni E, Carroll PR
    2018 Oct;74(4):444-452. doi: 10.1016/j.eururo.2018.05.014. Epub 2018 May 28.
    • More

      BACKGROUND: Among men with clinically low-risk prostate cancer, we have previously documented heterogeneity in terms of clinical characteristics and genomic risk scores.

      OBJECTIVE: To further study the underlying tumor biology of this patient population, by interrogating broader patterns of gene expression among men with clinically low-risk tumors.

      DESIGN, SETTING, AND PARTICIPANTS: Prostate biopsies from 427 patients considered potentially suitable for active surveillance underwent central pathology review and genome-wide expression profiling. These cases were compared with 1290 higher-risk biopsy cases with diverse clinical features from a prospective genomic registry.

      OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Average genomic risk (AGR) was determined from 18 published prognostic signatures, and MSigDB hallmark gene sets were analyzed using bootstrapped clustering methods. These sets were examined in relation to clinical variables and pathological and biochemical outcomes using multivariable regression analysis.

      RESULTS AND LIMITATIONS: A total of 408 (96%) biopsies passed RNA quality control. Based on AGR quartiles defined by the high-risk multicenter cases, the University of California, San Francisco (UCSF) low-risk patients were distributed across the quartiles as 219 (54%), 107 (26%), 61 (15%), and 21 (5%). Unsupervised clustering analysis of the hallmark gene set scores revealed three clusters, which were enriched for the previously described PAM50 luminal A, luminal B, and basal subtypes. AGR, but not the clusters, was associated with both pathological (odds ratio 1.34, 95% confidence interval [CI] 1.14-1.58) and biochemical outcomes (hazard ratio 1.53, 95% CI 1.19-1.93). These results may underestimate within-prostate genomic heterogeneity.

      CONCLUSIONS: Prostate cancers that are homogeneously low risk by traditional characteristics demonstrate substantial diversity at the level of genomic expression. Molecular substratification of low-risk prostate cancer will yield a better understanding of its divergent biology and, in the future may help personalize treatment recommendations.

      PATIENT SUMMARY: We studied the genomic characteristics of tumors from men diagnosed with low-risk prostate cancer. We found three main subtypes of prostate cancer with divergent tumor biology, similar to what has previously been found in women with breast cancer. In addition, we found that genomic risk scores were associated with worse pathology findings and prostate-specific antigen recurrence after surgery. These results suggest even greater genomic diversity among low-risk patients than has previously been documented with more limited signatures.

      PMID:29853306 | PMC:PMC6586429 | DOI:10.1016/j.eururo.2018.05.014


      View details for PubMedID 29853306
  • Development and Validation of a Prostate Cancer Genomic Signature that Predicts Early ADT Treatment Response Following Radical Prostatectomy Clinical cancer research : an official journal of the American Association for Cancer Research
    Karnes RJ, Sharma V, Choeurng V, Ashab HA, Erho N, Alshalalfa M, Trock B, Ross A, Yousefi K, Tsai H, Zhao SG, Tosoian JJ, Haddad Z, Takhar M, Chang SL, Spratt DE, Abdollah F, Jenkins RB, Klein EA, Nguyen PL, Dicker AP, Den RB, Davicioni E, Feng FY, Lotan TL, Schaeffer EM
    2018 Aug 15;24(16):3908-3916. doi: 10.1158/1078-0432.CCR-17-2745. Epub 2018 May 14.
    • More

      Purpose: Currently, no genomic signature exists to distinguish men most likely to progress on adjuvant androgen deprivation therapy (ADT) after radical prostatectomy for high-risk prostate cancer. Here we develop and validate a gene expression signature to predict response to postoperative ADT.Experimental Design: A training set consisting of 284 radical prostatectomy patients was established after 1:1 propensity score matching metastasis between adjuvant-ADT (a-ADT)-treated and no ADT-treated groups. An ADT Response Signature (ADT-RS) was identified from neuroendocrine and AR signaling-related genes. Two independent cohorts were used to form three separate data sets for validation (set I, n = 232; set II, n = 435; set III, n = 612). The primary endpoint of the analysis was postoperative metastasis.Results: Increases in ADT-RS score were associated with a reduction in risk of metastasis only in a-ADT patients. On multivariable analysis, ADT-RS by ADT treatment interaction term remained associated with metastasis in both validation sets (set I: HR = 0.18, Pinteraction = 0.009; set II: HR = 0.25, Pinteraction = 0.019). In a matched validation set III, patients with Low ADT-RS scores had similar 10-year metastasis rates in the a-ADT and no-ADT groups (30.1% vs. 31.0%, P = 0.989). Among High ADT-RS patients, 10-year metastasis rates were significantly lower for a-ADT versus no-ADT patients (9.4% vs. 29.2%, P = 0.021). The marginal ADT-RS by ADT interaction remained significant in the matched dataset (Pinteraction = 0.035).Conclusions: Patients with High ADT-RS benefited from a-ADT. In combination with prognostic risk factors, use of ADT-RS may thus allow for identification of ADT-responsive tumors that may benefit most from early androgen blockade after radical prostatectomy. We discovered a gene signature that when present in primary prostate tumors may be useful to predict patients who may respond to early ADT after surgery. Clin Cancer Res; 24(16); 3908-16. ©2018 AACR.

      PMID:29760221 | PMC:PMC6512950 | DOI:10.1158/1078-0432.CCR-17-2745


      View details for PubMedID 29760221
  • Development and Validation of a Novel Integrated Clinical-Genomic Risk Group Classification for Localized Prostate Cancer Journal of clinical oncology : official journal of the American Society of Clinical Oncology
    Spratt DE, Zhang J, Santiago-Jiménez M, Dess RT, Davis JW, Den RB, Dicker AP, Kane CJ, Pollack A, Stoyanova R, Abdollah F, Ross AE, Cole A, Uchio E, Randall JM, Nguyen H, Zhao SG, Mehra R, Glass AG, Lam LC, Chelliserry J, Plessis Md, Choeurng V, Aranes M, Kolisnik T, Margrave J, Alter J, Jordan J, Buerki C, Yousefi K, Haddad Z, Davicioni E, Trabulsi EJ, Loeb S, Tewari A, Carroll PR, Weinmann S, Schaeffer EM, Klein EA, Karnes RJ, Feng FY, Nguyen PL
    2018 Feb 20;36(6):581-590. doi: 10.1200/JCO.2017.74.2940. Epub 2017 Nov 29.
    • More

      Purpose It is clinically challenging to integrate genomic-classifier results that report a numeric risk of recurrence into treatment recommendations for localized prostate cancer, which are founded in the framework of risk groups. We aimed to develop a novel clinical-genomic risk grouping system that can readily be incorporated into treatment guidelines for localized prostate cancer. Materials and Methods Two multicenter cohorts (n = 991) were used for training and validation of the clinical-genomic risk groups, and two additional cohorts (n = 5,937) were used for reclassification analyses. Competing risks analysis was used to estimate the risk of distant metastasis. Time-dependent c-indices were constructed to compare clinicopathologic risk models with the clinical-genomic risk groups. Results With a median follow-up of 8 years for patients in the training cohort, 10-year distant metastasis rates for National Comprehensive Cancer Network (NCCN) low, favorable-intermediate, unfavorable-intermediate, and high-risk were 7.3%, 9.2%, 38.0%, and 39.5%, respectively. In contrast, the three-tier clinical-genomic risk groups had 10-year distant metastasis rates of 3.5%, 29.4%, and 54.6%, for low-, intermediate-, and high-risk, respectively, which were consistent in the validation cohort (0%, 25.9%, and 55.2%, respectively). C-indices for the clinical-genomic risk grouping system (0.84; 95% CI, 0.61 to 0.93) were improved over NCCN (0.73; 95% CI, 0.60 to 0.86) and Cancer of the Prostate Risk Assessment (0.74; 95% CI, 0.65 to 0.84), and 30% of patients using NCCN low/intermediate/high would be reclassified by the new three-tier system and 67% of patients would be reclassified from NCCN six-tier (very-low- to very-high-risk) by the new six-tier system. Conclusion A commercially available genomic classifier in combination with standard clinicopathologic variables can generate a simple-to-use clinical-genomic risk grouping that more accurately identifies patients at low, intermediate, and high risk for metastasis and can be easily incorporated into current guidelines to better risk-stratify patients.

      PMID:29185869 | PMC:PMC6530900 | DOI:10.1200/JCO.2017.74.2940


      View details for PubMedID 29185869
  • Androgen receptor as a mediator and biomarker of radioresistance in triple-negative breast cancer NPJ breast cancer
    Speers C, Zhao SG, Chandler B, Liu M, Wilder-Romans K, Olsen E, Nyati S, Ritter C, Alluri PG, Kothari V, Hayes DF, Lawrence TS, Spratt DE, Wahl DR, Pierce LJ, Feng FY
    2017 Aug 18;3:29. doi: 10.1038/s41523-017-0038-2. eCollection 2017.
    • More

      Increased rates of locoregional recurrence have been observed in triple-negative breast cancer despite chemotherapy and radiation therapy. Thus, approaches that combine therapies for radiosensitization in triple-negative breast cancer are critically needed. We characterized the radiation therapy response of 21 breast cancer cell lines and paired this radiation response data with high-throughput drug screen data to identify androgen receptor as a top target for radiosensitization. Our radiosensitizer screen nominated bicalutamide as the drug most effective in treating radiation therapy-resistant breast cancer cell lines. We subsequently evaluated the expression of androgen receptor in >2100 human breast tumor samples and 51 breast cancer cell lines and found significant heterogeneity in androgen receptor expression with enrichment at the protein and RNA level in triple-negative breast cancer. There was a strong correlation between androgen receptor RNA and protein expression across all breast cancer subtypes (R2 = 0.72, p < 0.01). In patients with triple-negative breast cancer, expression of androgen receptor above the median was associated with increased risk of locoregional recurrence after radiation therapy (hazard ratio for locoregional recurrence 2.9-3.2)) in two independent data sets, but there was no difference in locoregional recurrence in triple-negative breast cancer patients not treated with radiation therapy when stratified by androgen receptor expression. In multivariable analysis, androgen receptor expression was most significantly associated with worse local recurrence-free survival after radiation therapy (hazard ratio of 3.58) suggesting that androgen receptor expression may be a biomarker of radiation response in triple-negative breast cancer. Inhibition of androgen receptor with MDV3100 (enzalutamide) induced radiation sensitivity (enhancement ratios of 1.22-1.60) in androgen receptor-positive triple-negative breast cancer lines, but did not affect androgen receptor-negative triple-negative breast cancer or estrogen-receptor-positive, androgen receptor-negative breast cancer cell lines. androgen receptor inhibition with MDV3100 significantly radiosensitized triple-negative breast cancer xenografts in mouse models and markedly delayed tumor doubling/tripling time and tumor weight. Radiosensitization was at least partially dependent on impaired dsDNA break repair mediated by DNA protein kinase catalytic subunit. Our results implicate androgen receptor as a mediator of radioresistance in breast cancer and identify androgen receptor inhibition as a potentially effective strategy for the treatment of androgen receptor-positive radioresistant tumors.

      PMID:28840192 | PMC:PMC5562815 | DOI:10.1038/s41523-017-0038-2


      View details for PubMedID 28840192
  • Individual Patient Data Analysis of Randomized Clinical Trials: Impact of Black Race on Castration-resistant Prostate Cancer Outcomes European urology focus
    Spratt DE, Chen Y, Mahal BA, Osborne JR, Zhao SG, Morgan TM, Palapattu G, Feng FY, Nguyen PL
    2016 Dec;2(5):532-539. doi: 10.1016/j.euf.2016.03.010. Epub 2016 Apr 1.
    • More

      BACKGROUND: Population data suggest that black men have a higher risk of dying from prostate cancer (PCa) than other racial ethnicities.

      OBJECTIVE: To examine the impact of black race on progression-free survival (PFS) and overall survival (OS) among men with metastatic castration-resistant PCa (mCRPC) enrolled in randomized controlled trials (RCTs).

      DESIGN, SETTING, AND PARTICIPANTS: A pooled analysis was performed on individual patient data from five modern PCa RCTs available from Project Data Sphere.

      OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Adjusted hazard ratios (HRs) were calculated to compare black and white race regarding PFS and OS. Subgroup analyses of mCRPC trials were performed based on the control arm treatments (mitoxantrone or docetaxel). Relevant covariates were used for adjustment in all analyses.

      RESULTS AND LIMITATIONS: A total of 1613 patients were included; 77 were black (4.7%). No significant differences between black and white men's baseline characteristics were noted regarding age, performance status, or pretreatment prostate-specific antigen. The pooled HRs for black race for OS and PFS were 1.01 (95% confidence interval [CI], 0.73-1.35) and 1.29 (95% CI, 0.95-1.76), respectively. The median OS for black compared with white men was 254 versus 238 d (p=0.92), respectively, with mitoxantrone and 581 versus 546 d (p=0.53), respectively, with docetaxel. The primary limitation was the relatively small number of black men enrolled in mCRPC clinical trials.

      CONCLUSIONS: In the context of RCTs, in which patients receive generally uniform treatment, a significant difference in OS for black men could not be detected in mCRPC. Black men continue to be dramatically underrepresented in RCTs, and efforts are needed to increase minority accrual to these trials.

      PATIENT SUMMARY: We looked at the outcomes of men treated in randomized controlled trials to determine the impact of black race on survival. We found that in the context of modern clinical trials, there does not appear to be a significant difference in survival between black and white races; however, a trend for greater progression in black men was noted.

      PMID:28723519 | DOI:10.1016/j.euf.2016.03.010


      View details for PubMedID 28723519
  • Genome-wide CRISPR screen identifies HNRNPL as a prostate cancer dependency regulating RNA splicing Proceedings of the National Academy of Sciences of the United States of America
    Fei T, Chen Y, Xiao T, Li W, Cato L, Zhang P, Cotter MB, Bowden M, Lis RT, Zhao SG, Wu Q, Feng FY, Loda M, He HH, Liu XS, Brown M
    2017 Jun 27;114(26):E5207-E5215. doi: 10.1073/pnas.1617467114. Epub 2017 Jun 13.
    • More

      Alternative RNA splicing plays an important role in cancer. To determine which factors involved in RNA processing are essential in prostate cancer, we performed a genome-wide CRISPR/Cas9 knockout screen to identify the genes that are required for prostate cancer growth. Functional annotation defined a set of essential spliceosome and RNA binding protein (RBP) genes, including most notably heterogeneous nuclear ribonucleoprotein L (HNRNPL). We defined the HNRNPL-bound RNA landscape by RNA immunoprecipitation coupled with next-generation sequencing and linked these RBP-RNA interactions to changes in RNA processing. HNRNPL directly regulates the alternative splicing of a set of RNAs, including those encoding the androgen receptor, the key lineage-specific prostate cancer oncogene. HNRNPL also regulates circular RNA formation via back splicing. Importantly, both HNRNPL and its RNA targets are aberrantly expressed in human prostate tumors, supporting their clinical relevance. Collectively, our data reveal HNRNPL and its RNA clients as players in prostate cancer growth and potential therapeutic targets.

      PMID:28611215 | PMC:PMC5495225 | DOI:10.1073/pnas.1617467114


      View details for PubMedID 28611215
  • Associations of Luminal and Basal Subtyping of Prostate Cancer With Prognosis and Response to Androgen Deprivation Therapy JAMA oncology
    Zhao SG, Chang SL, Erho N, Yu M, Lehrer J, Alshalalfa M, Speers C, Cooperberg MR, Kim W, Ryan CJ, Den RB, Freedland SJ, Posadas E, Sandler H, Klein EA, Black P, Seiler R, Tomlins SA, Chinnaiyan AM, Jenkins RB, Davicioni E, Ross AE, Schaeffer EM, Nguyen PL, Carroll PR, Karnes RJ, Spratt DE, Feng FY
    2017 Dec 1;3(12):1663-1672. doi: 10.1001/jamaoncol.2017.0751.
    • More

      IMPORTANCE: There is a clear need for a molecular subtyping approach in prostate cancer to identify clinically distinct subgroups that benefit from specific therapies.

      OBJECTIVES: To identify prostate cancer subtypes based on luminal and basal lineage and to determine associations with clinical outcomes and response to treatment.

      DESIGN, SETTING, AND PARTICIPANTS: The PAM50 classifier was used to subtype 1567 retrospectively collected (median follow-up, 10 years) and 2215 prospectively collected prostate cancer samples into luminal- and basal-like subtypes.

      MAIN OUTCOMES AND MEASURES: Metastasis, biochemical recurrence, overall survival, prostate cancer–specific survival, associations with biological pathways, and clinicopathologic variables were the main outcomes.

      RESULTS: Among the 3782 samples, the PAM50 classifier consistently segregated prostate cancer into 3 subtypes in both the retrospective and prospective cohorts: luminal A (retrospective, 538 [34.3%]; prospective, 737 [33.3%]), luminal B (retrospective, 447 [28.5%]; prospective, 723 [32.6%]), and basal (retrospective, 582 [37.1%]; prospective, 755 [34.1%]). Known luminal lineage markers, such as NKX3.1 and KRT18, were enriched in luminal-like cancers, and the basal lineage CD49f signature was enriched in basal-like cancers, demonstrating the connection between these subtypes and established prostate cancer biology. In the retrospective cohort, luminal B prostate cancers exhibited the poorest clinical prognoses on both univariable and multivariable analyses accounting for standard clinicopathologic prognostic factors (10-year biochemical recurrence-free survival [bRFS], 29%; distant metastasis-free survival [DMFS], 53%; prostate cancer-specific survival [PCSS], 78%; overall survival [OS], 69%), followed by basal prostate cancers (10-year bRFS, 39%; DMFS, 73%; PCSS, 86%; OS, 80%) and luminal A prostate cancers (10-year bRFS, 41%; DMFS, 73%; PCSS, 89%; OS, 82%). Although both luminal-like subtypes were associated with increased androgen receptor expression and signaling, only luminal B prostate cancers were significantly associated with postoperative response to androgen deprivation therapy (ADT) in a subset analysis in our retrospective cohorts (n = 315) matching patients based on clinicopathologic variables (luminal B 10-year metastasis: treated, 33% vs untreated, 55%; nonluminal B 10-year metastasis: treated, 37% vs untreated, 21%; P = .006 for interaction).

      CONCLUSIONS AND RELEVANCE: Luminal- and basal-like prostate cancers demonstrate divergent clinical behavior, and patients with luminal B tumors respond better to postoperative ADT than do patients with non–luminal B tumors. These findings contribute novel insight into prostate cancer biology, providing a potential clinical tool to personalize ADT treatment for prostate cancer by predicting which men may benefit from ADT after surgery.

      PMID:28494073 | PMC:PMC5824281 | DOI:10.1001/jamaoncol.2017.0751


      View details for PubMedID 28494073
  • Individual Patient-Level Meta-Analysis of the Performance of the Decipher Genomic Classifier in High-Risk Men After Prostatectomy to Predict Development of Metastatic Disease Journal of clinical oncology : official journal of the American Society of Clinical Oncology
    Spratt DE, Yousefi K, Deheshi S, Ross AE, Den RB, Schaeffer EM, Trock BJ, Zhang J, Glass AG, Dicker AP, Abdollah F, Zhao SG, Lam LC, Plessis Md, Choeurng V, Haddad Z, Buerki C, Davicioni E, Weinmann S, Freedland SJ, Klein EA, Karnes RJ, Feng FY
    2017 Jun 20;35(18):1991-1998. doi: 10.1200/JCO.2016.70.2811. Epub 2017 Mar 30.
    • More

      Purpose To perform the first meta-analysis of the performance of the genomic classifier test, Decipher, in men with prostate cancer postprostatectomy. Methods MEDLINE, EMBASE, and the Decipher genomic resource information database were searched for published reports between 2011 and 2016 of men treated by prostatectomy that assessed the benefit of the Decipher test. Multivariable Cox proportional hazards models fit to individual patient data were performed; meta-analyses were conducted by pooling the study-specific hazard ratios (HRs) using random-effects modeling. Extent of heterogeneity between studies was determined with the I2 test. Results Five studies (975 total patients, and 855 patients with individual patient-level data) were eligible for analysis, with a median follow-up of 8 years. Of the total cohort, 60.9%, 22.6%, and 16.5% of patients were classified by Decipher as low, intermediate, and high risk, respectively. The 10-year cumulative incidence metastases rates were 5.5%, 15.0%, and 26.7% ( P < .001), respectively, for the three risk classifications. Pooling the study-specific Decipher HRs across the five studies resulted in an HR of 1.52 (95% CI, 1.39 to 1.67; I2 = 0%) per 0.1 unit. In multivariable analysis of individual patient data, adjusting for clinicopathologic variables, Decipher remained a statistically significant predictor of metastasis (HR, 1.30; 95% CI, 1.14 to 1.47; P < .001) per 0.1 unit. The C-index for 10-year distant metastasis of the clinical model alone was 0.76; this increased to 0.81 with inclusion of Decipher. Conclusion The genomic classifier test, Decipher, can independently improve prognostication of patients postprostatectomy, as well as within nearly all clinicopathologic, demographic, and treatment subgroups. Future study of how to best incorporate genomic testing in clinical decision-making and subsequent treatment recommendations is warranted.

      PMID:28358655 | PMC:PMC6530581 | DOI:10.1200/JCO.2016.70.2811


      View details for PubMedID 28358655
  • Anatomical patterns of recurrence following biochemical relapse after post-prostatectomy salvage radiation therapy: a multi-institutional study BJU international
    Jackson WC, Desai NB, Abugharib AE, Tumati V, Dess RT, Lee JY, Zhao SG, Soliman M, Folkert M, Laine A, Hannan R, Zumsteg ZS, Sandler H, Hamstra DA, Montgomery JS, Miller DC, Kozminski MA, Hollenbeck BK, Hearn JW, Palapattu G, Tomlins SA, Mehra R, Morgan TM, Feng FY, Spratt DE
    2017 Sep;120(3):351-357. doi: 10.1111/bju.13792. Epub 2017 Feb 26.
    • More

      OBJECTIVES: To characterise the frequency and detailed anatomical sites of failure for patients receiving post-radical prostatectomy (RP) salvage radiation therapy (SRT).

      PATIENTS AND METHODS: A multi-institutional retrospective study was performed on 574 men who underwent SRT between 1986 and 2013. Anatomical recurrence patterns were classified as lymphotrophic (lymph nodes only), osteotrophic (bone only), or multifocal if both were present. Isolated first failure sites were defined as sites of initial clinically detected recurrence that remained isolated for at least 3 months.

      RESULTS: The median follow-up after SRT was 6.8 years. The 8-year rates of local, regional, and distant failure for patients undergoing SRT were 2%, 6%, and 21%, respectively. Of the 22% men (128 of 574) who developed a clinically detectable recurrence, 17%, 50%, and 31% were lymphotrophic, osteotrophic, and multifocal, respectively. The trophic nature of metastases was prognostic for distant metastases-free survival (DMFS) and prostate cancer-specific survival (PCSS); the 10-year rates of DMFS were 18%, 5%, and 7% (P < 0.01), and PCSS were 78%, 68%, and 56% (P < 0.01), for lymphotrophic, osteotrophic, and multifocal failure patterns, respectively.

      CONCLUSIONS: We demonstrate that trophism for metastatic site has significant prognostic impact on PCSS in men treated with SRT. Radiographic local failure is an uncommon event after SRT when compared to historical data of patients treated with surgery monotherapy. However, distant failure remains a challenge in this patient population and warrants further therapeutic investigation.

      PMID:28139024 | DOI:10.1111/bju.13792


      View details for PubMedID 28139024
  • MicroRNA-194 Promotes Prostate Cancer Metastasis by Inhibiting SOCS2 Cancer research
    Das R, Gregory PA, Fernandes RC, Denis I, Wang Q, Townley SL, Zhao SG, Hanson AR, Pickering MA, Armstrong HK, Lokman NA, Ebrahimie E, Davicioni E, Jenkins RB, Karnes RJ, Ross AE, Den RB, Klein EA, Chi KN, Ramshaw HS, Williams ED, Zoubeidi A, Goodall GJ, Feng FY, Butler LM, Tilley WD, Selth LA
    2017 Feb 15;77(4):1021-1034. doi: 10.1158/0008-5472.CAN-16-2529. Epub 2016 Dec 23.
    • More

      Serum levels of miR-194 have been reported to predict prostate cancer recurrence after surgery, but its functional contributions to this disease have not been studied. Herein, it is demonstrated that miR-194 is a driver of prostate cancer metastasis. Prostate tissue levels of miR-194 were associated with disease aggressiveness and poor outcome. Ectopic delivery of miR-194 stimulated migration, invasion, and epithelial-mesenchymal transition in human prostate cancer cell lines, and stable overexpression of miR-194 enhanced metastasis of intravenous and intraprostatic tumor xenografts. Conversely, inhibition of miR-194 activity suppressed the invasive capacity of prostate cancer cell lines in vitro and in vivo Mechanistic investigations identified the ubiquitin ligase suppressor of cytokine signaling 2 (SOCS2) as a direct, biologically relevant target of miR-194 in prostate cancer. Low levels of SOCS2 correlated strongly with disease recurrence and metastasis in clinical specimens. SOCS2 downregulation recapitulated miR-194-driven metastatic phenotypes, whereas overexpression of a nontargetable SOCS2 reduced miR-194-stimulated invasion. Targeting of SOCS2 by miR-194 resulted in derepression of the oncogenic kinases FLT3 and JAK2, leading to enhanced ERK and STAT3 signaling. Pharmacologic inhibition of ERK and JAK/STAT pathways reversed miR-194-driven phenotypes. The GATA2 transcription factor was identified as an upstream regulator of miR-194, consistent with a strong concordance between GATA2 and miR-194 levels in clinical specimens. Overall, these results offer new insights into the molecular mechanisms of metastatic progression in prostate cancer. Cancer Res; 77(4); 1021-34. ©2016 AACR.

      PMID:28011622 | DOI:10.1158/0008-5472.CAN-16-2529


      View details for PubMedID 28011622
  • RB Loss Promotes Prostate Cancer Metastasis Cancer research
    Thangavel C, Boopathi E, Liu Y, Haber A, Ertel A, Bhardwaj A, Addya S, Williams N, Ciment SJ, Cotzia P, Dean JL, Snook A, McNair C, Price M, Hernandez JR, Zhao SG, Birbe R, McCarthy JB, Turley EA, Pienta KJ, Feng FY, Dicker AP, Knudsen KE, Den RB
    2017 Feb 15;77(4):982-995. doi: 10.1158/0008-5472.CAN-16-1589. Epub 2016 Dec 6.
    • More

      RB loss occurs commonly in neoplasia but its contributions to advanced cancer have not been assessed directly. Here we show that RB loss in multiple murine models of cancer produces a prometastatic phenotype. Gene expression analyses showed that regulation of the cell motility receptor RHAMM by the RB/E2F pathway was critical for epithelial-mesenchymal transition, motility, and invasion by cancer cells. Genetic modulation or pharmacologic inhibition of RHAMM activity was sufficient and necessary for metastatic phenotypes induced by RB loss in prostate cancer. Mechanistic studies in this setting established that RHAMM stabilized F-actin polymerization by controlling ROCK signaling. Collectively, our findings show how RB loss drives metastatic capacity and highlight RHAMM as a candidate therapeutic target for treating advanced prostate cancer. Cancer Res; 77(4); 982-95. ©2016 AACR.

      PMID:27923835 | PMC:PMC5700768 | DOI:10.1158/0008-5472.CAN-16-1589


      View details for PubMedID 27923835
  • Glioblastoma Therapy Can Be Augmented by Targeting IDH1-Mediated NADPH Biosynthesis Cancer research
    Wahl DR, Dresser J, Wilder-Romans K, Parsels JD, Zhao SG, Davis M, Zhao L, Kachman M, Wernisch S, Burant CF, Morgan MA, Feng FY, Speers C, Lyssiotis CA, Lawrence TS
    2017 Feb 15;77(4):960-970. doi: 10.1158/0008-5472.CAN-16-2008. Epub 2016 Dec 6.
    • More

      NADPH is a critical reductant needed in cancer cells to fuel the biosynthesis of deoxynucleotides and antioxidants and to sustain stress-survival responses after radiation-induced DNA damage. Thus, one rational strategy to attack cancer cells is to target their heavy reliance on NADPH. Here, we report that the isocitrate dehydrogenase IDH1 is the most strongly upregulated NADPH-producing enzyme in glioblastoma (GBM). IDH1 silencing in GBM cells reduced levels of NADPH, deoxynucleotides, and glutathione and increased their sensitivity to radiation-induced senescence. Rescuing these metabolic restrictions was sufficient to reverse IDH1-mediated radiosensitization. In a murine xenograft model of human GBM, we found that IDH1 silencing significantly improved therapeutic responses to fractionated radiotherapy, when compared with either treatment alone. In summary, our work offers a mechanistic rationale for IDH1 inhibition as a metabolic strategy to improve the response of GBM to radiotherapy. Cancer Res; 77(4); 960-70. ©2016 AACR.

      PMID:27923831 | PMC:PMC5726266 | DOI:10.1158/0008-5472.CAN-16-2008


      View details for PubMedID 27923831
  • Correlation of B7-H3 with androgen receptor, immune pathways and poor outcome in prostate cancer: an expression-based analysis Prostate cancer and prostatic diseases
    Benzon B, Zhao SG, Haffner MC, Takhar M, Erho N, Yousefi K, Hurley P, Bishop JL, Tosoian J, Ghabili K, Alshalalfa M, Glavaris S, Simons BW, Tran P, Davicioni E, Karnes RJ, Boudadi K, Antonarakis ES, Schaeffer EM, Drake CG, Feng F, Ross AE
    2017 Mar;20(1):28-35. doi: 10.1038/pcan.2016.49. Epub 2016 Nov 1.
    • More

      BACKGROUND: B7-H3 (CD276), part of the B7 superfamily of immune checkpoint molecules, has been shown to have an immunomodulatory role. Its regulation, receptor and mechanism of action remain unclear. B7-H3 protein expression correlates with prostate cancer outcomes, and humanized monoclonal antibodies (that is, enoblituzumab) are currently being investigated for therapeutic use. Here we used genomic expression data to examine the relationship between B7-H3 mRNA expression and prostate cancer.

      METHODS: Prostatectomy tissue from 2781 patients were profiled using the Affymetrix HuEx 1.0 ST microarray. Pairwise comparisons were used to identify significant associations between B7-H3 expression and clinicopathologic variables, and survival analyses were used to evaluate the prognostic significance of B7-H3. Pearson's correlation analyses were performed to assess the relationship of B7-H3 expression with molecular subtypes and individual transcripts. Androgen receptor (AR) occupancy at the B7-H3 locus was determined using chromatin immunoprecipitation (ChIP), and androgen-dependent expression changes in B7-H3 was evaluated by quantitative reverse transcription PCR in LNCaP cell lines. Oncomine was queried to evaluate B7-H3 expression in metastatic disease.

      RESULTS: B7-H3 mRNA expression was positively associated with higher Gleason score (P<0.001), tumor stage (P<0.001), and castrate resistant metastatic disease (P<0.0001). High B7-H3 expression correlated with the development of metastasis and prostate cancer specific mortality, but this was not significant on multi-variable analysis. B7-H3 expression correlated with ERG-positive disease (r=0.99) and AR expression (r=0.36). ChIP revealed an AR-binding site upstream of B7-H3, and the presence of androgens decreased B7-H3 expression in LNCaP suggesting potential direct AR regulation. Gene set enrichment analysis demonstrated an association of B7-H3 with androgen signaling as well as immune regulatory pathways.

      CONCLUSIONS: Higher B7-H3 expression correlates with Gleason grade, prostate cancer stage and poor oncologic outcomes in prostatectomy cohorts. B7-H3 expression appears to be related to androgen signaling as well as the immune reactome.

      PMID:27801901 | PMC:PMC6512966 | DOI:10.1038/pcan.2016.49


      View details for PubMedID 27801901
  • Development and validation of a 24-gene predictor of response to postoperative radiotherapy in prostate cancer: a matched, retrospective analysis The Lancet. Oncology
    Zhao SG, Chang SL, Spratt DE, Erho N, Yu M, Ashab HA, Alshalalfa M, Speers C, Tomlins SA, Davicioni E, Dicker AP, Carroll PR, Cooperberg MR, Freedland SJ, Karnes RJ, Ross AE, Schaeffer EM, Den RB, Nguyen PL, Feng FY
    2016 Nov;17(11):1612-1620. doi: 10.1016/S1470-2045(16)30491-0. Epub 2016 Oct 12.
    • More

      BACKGROUND: Postoperative radiotherapy has an important role in the treatment of prostate cancer, but personalised patient selection could improve outcomes and spare unnecessary toxicity. We aimed to develop and validate a gene expression signature to predict which patients would benefit most from postoperative radiotherapy.

      METHODS: Patients were eligible for this matched, retrospective study if they were included in one of five published US studies (cohort, case-cohort, and case-control studies) of patients with prostate adenocarcinoma who had radical prostatectomy (with or without postoperative radiotherapy) and had gene expression analysis of the tumour, with long-term follow-up and complete clinicopathological data. Additional treatment after surgery was at the treating physician's discretion. In each cohort, patients who had postoperative radiotherapy were matched with patients who had not had radiotherapy using Gleason score, prostate-specific antigen concentration, surgical margin status, extracapsular extension, seminal vesicle invasion, lymph node invasion, and androgen deprivation therapy. We constructed a matched training cohort using patients from one study in which we developed a 24-gene Post-Operative Radiation Therapy Outcomes Score (PORTOS). We generated a pooled matched validation cohort using patients from the remaining four studies. The primary endpoint was the development of distant metastasis.

      FINDINGS: In the training cohort (n=196), among patients with a high PORTOS (n=39), those who had radiotherapy had a lower incidence of distant metastasis than did patients who did not have radiotherapy, with a 10-year metastasis rate of 5% (95% CI 0-14) in patients who had radiotherapy (n=20) and 63% (34-80) in patients who did not have radiotherapy (n=19; hazard ratio [HR] 0·12 [95% CI 0·03-0·41], p<0·0001), whereas among patients with a low PORTOS (n=157), those who had postoperative radiotherapy (n=78) had a greater incidence of distant metastasis at 10 years than did their untreated counterparts (n=79; 57% [44-67] vs 31% [20-41]; HR 2·5 [1·6-4·1], p<0·0001), with a significant treatment interaction (pinteraction<0·0001). The finding that PORTOS could predict outcome due to radiotherapy treatment was confirmed in the validation cohort (n=330), which showed that patients who had radiotherapy had a lower incidence of distant metastasis compared with those who did not have radiotherapy, but only in the high PORTOS group (high PORTOS [n=82]: 4% [95% CI 0-10] in the radiotherapy group [n=57] vs 35% [95% CI 7-54] in the no radiotherapy group [n=25] had metastasis at 10 years; HR 0·15 [95% CI 0·04-0·60], p=0·0020; low PORTOS [n=248]: 32% [95% CI 19-43] in the radiotherapy group [n=108] vs 32% [95% CI 22-40] in the no radiotherapy group [n=140]; HR 0·92 [95% CI 0·56-1·51], p=0·76), with a significant interaction (pinteraction=0·016). The conventional prognostic tools Decipher, CAPRA-S, and microarray version of the cell cycle progression signature did not predict response to radiotherapy (pinteraction>0·05 for all).

      INTERPRETATION: Patients with a high PORTOS who had postoperative radiotherapy were less likely to have metastasis at 10 years than those who did not have radiotherapy, suggesting that treatment with postoperative radiotherapy should be considered in this subgroup. PORTOS should be investigated further in additional independent cohorts.

      FUNDING: None.

      PMID:27743920 | DOI:10.1016/S1470-2045(16)30491-0


      View details for PubMedID 27743920
  • Cell cycle-coupled expansion of AR activity promotes cancer progression Oncogene
    McNair C, Urbanucci A, Comstock ES, Augello MA, Goodwin JF, Launchbury R, Zhao SG, Schiewer MJ, Ertel A, Karnes J, Davicioni E, Wang L, Wang Q, Mills IG, Feng FY, Li W, Carroll JS, Knudsen KE
    2017 Mar 23;36(12):1655-1668. doi: 10.1038/onc.2016.334. Epub 2016 Sep 26.
    • More

      The androgen receptor (AR) is required for prostate cancer (PCa) survival and progression, and ablation of AR activity is the first line of therapeutic intervention for disseminated disease. While initially effective, recurrent tumors ultimately arise for which there is no durable cure. Despite the dependence of PCa on AR activity throughout the course of disease, delineation of the AR-dependent transcriptional network that governs disease progression remains elusive, and the function of AR in mitotically active cells is not well understood. Analyzing AR activity as a function of cell cycle revealed an unexpected and highly expanded repertoire of AR-regulated gene networks in actively cycling cells. New AR functions segregated into two major clusters: those that are specific to cycling cells and retained throughout the mitotic cell cycle ('Cell Cycle Common'), versus those that were specifically enriched in a subset of cell cycle phases ('Phase Restricted'). Further analyses identified previously unrecognized AR functions in major pathways associated with clinical PCa progression. Illustrating the impact of these unmasked AR-driven pathways, dihydroceramide desaturase 1 was identified as an AR-regulated gene in mitotically active cells that promoted pro-metastatic phenotypes, and in advanced PCa proved to be highly associated with development of metastases, recurrence after therapeutic intervention and reduced overall survival. Taken together, these findings delineate AR function in mitotically active tumor cells, thus providing critical insight into the molecular basis by which AR promotes development of lethal PCa and nominate new avenues for therapeutic intervention.

      PMID:27669432 | PMC:PMC5364060 | DOI:10.1038/onc.2016.334


      View details for PubMedID 27669432
  • The lncRNA landscape of breast cancer reveals a role for DSCAM-AS1 in breast cancer progression Nature communications
    Niknafs YS, Han S, Ma T, Speers C, Zhang C, Wilder-Romans K, Iyer MK, Pitchiaya S, Malik R, Hosono Y, Prensner JR, Poliakov A, Singhal U, Xiao L, Kregel S, Siebenaler RF, Zhao SG, Uhl M, Gawronski A, Hayes DF, Pierce LJ, Cao X, Collins C, Backofen R, Sahinalp CS, Rae JM, Chinnaiyan AM, Feng FY
    2016 Sep 26;7:12791. doi: 10.1038/ncomms12791.
    • More

      Molecular classification of cancers into subtypes has resulted in an advance in our understanding of tumour biology and treatment response across multiple tumour types. However, to date, cancer profiling has largely focused on protein-coding genes, which comprise <1% of the genome. Here we leverage a compendium of 58,648 long noncoding RNAs (lncRNAs) to subtype 947 breast cancer samples. We show that lncRNA-based profiling categorizes breast tumours by their known molecular subtypes in breast cancer. We identify a cohort of breast cancer-associated and oestrogen-regulated lncRNAs, and investigate the role of the top prioritized oestrogen receptor (ER)-regulated lncRNA, DSCAM-AS1. We demonstrate that DSCAM-AS1 mediates tumour progression and tamoxifen resistance and identify hnRNPL as an interacting protein involved in the mechanism of DSCAM-AS1 action. By highlighting the role of DSCAM-AS1 in breast cancer biology and treatment resistance, this study provides insight into the potential clinical implications of lncRNAs in breast cancer.

      PMID:27666543 | PMC:PMC5052669 | DOI:10.1038/ncomms12791


      View details for PubMedID 27666543
  • Very Early Salvage Radiotherapy Improves Distant Metastasis-Free Survival The Journal of urology
    Abugharib A, Jackson WC, Tumati V, Dess RT, Lee JY, Zhao SG, Soliman M, Zumsteg ZS, Mehra R, Feng FY, Morgan TM, Desai N, Spratt DE
    2017 Mar;197(3 Pt 1):662-668. doi: 10.1016/j.juro.2016.08.106. Epub 2016 Sep 7.
    • More

      PURPOSE: Early salvage radiotherapy following radical prostatectomy for prostate cancer is commonly advocated in place of adjuvant radiotherapy. We aimed to determine the optimal definition of early salvage radiotherapy.

      MATERIALS AND METHODS: We performed a multi-institutional retrospective study of 657 men who underwent salvage radiotherapy between 1986 and 2013. Two comparisons were made to determine the optimal definition of early salvage radiotherapy, including 1) the time from radical prostatectomy to salvage radiotherapy (less than 9, 9 to 21, 22 to 47 or greater than 48 months) and 2) the level of detectable pre-salvage radiotherapy prostate specific antigen (0.01 to 0.2, greater than 0.2 to 0.5 or greater than 0.5 ng/ml). Outcomes included freedom from salvage androgen deprivation therapy, and biochemical relapse-free, distant metastases-free and prostate cancer specific survival.

      RESULTS: Median followup was 9.8 years. Time from radical prostatectomy to salvage radiotherapy did not correlate with 10-year biochemical relapse-free survival rates (R2 = 0.18). Increasing pre-salvage radiotherapy prostate specific antigen strongly correlated with biochemical relapse-free survival (R2 = 0.91). Increasing detectable pre-salvage radiotherapy prostate specific antigen (0.01 to 0.2, greater than 0.2 to 0.5 and greater than 0.5 ng/ml) predicted worse 10-year biochemical relapse-free survival (62%, 44% and 27%), freedom from salvage androgen deprivation therapy (77%, 66% and 49%), distant metastases-free survival (86%, 79% and 66%, each p <0.001) and prostate cancer specific survival (93%, 89% and 80%, respectively, p = 0.001). On multivariable analysis early salvage radiotherapy (prostate specific antigen greater than 0.2 to 0.5 ng/ml) was associated with a twofold increase in biochemical failure, use of salvage androgen deprivation therapy and distant metastases compared to very early salvage radiotherapy (prostate specific antigen 0.01 to 0.2 ng/ml).

      CONCLUSIONS: The duration from radical prostatectomy to salvage radiotherapy is not independently prognostic for outcomes after salvage radiotherapy and it should not be used to define early salvage radiotherapy. Grouping all patients with pre-salvage radiotherapy prostate specific antigen 0.5 ng/ml or less may be inadequate to define early salvage radiotherapy and it has a relevant impact on ongoing and future clinical trials.

      PMID:27614333 | DOI:10.1016/j.juro.2016.08.106


      View details for PubMedID 27614333
  • Multi-institutional Analysis Shows that Low PCAT-14 Expression Associates with Poor Outcomes in Prostate Cancer European urology
    White NM, Zhao SG, Zhang J, Rozycki EB, Dang HX, McFadden SD, Eteleeb AM, Alshalalfa M, Vergara IA, Erho N, Arbeit JM, Karnes RJ, Den RB, Davicioni E, Maher CA
    2017 Feb;71(2):257-266. doi: 10.1016/j.eururo.2016.07.012. Epub 2016 Jul 22.
    • More

      BACKGROUND: Long noncoding RNAs (lncRNAs) are an emerging class of relatively underexplored oncogenic molecules with biological and clinical significance. Current inadequacies for stratifying patients with aggressive disease presents a strong rationale to systematically identify lncRNAs as clinical predictors in localized prostate cancer.

      OBJECTIVE: To identify RNA biomarkers associated with aggressive prostate cancer.

      DESIGN, SETTING, AND PARTICIPANTS: Radical prostatectomy microarray and clinical data was obtained from 910 patients in three published institutional cohorts: Mayo Clinic I (N=545, median follow-up 13.8 yr), Mayo Clinic II (N=235, median follow-up 6.7 yr), and Thomas Jefferson University (N=130, median follow-up 9.6 yr).

      OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: The primary clinical endpoint was distant metastasis-free survival. Secondary endpoints include prostate cancer-specific survival and overall survival. Univariate and multivariate Cox regression were used to evaluate the association of lncRNA expression and these endpoints.

      RESULTS AND LIMITATIONS: An integrative analysis revealed Prostate Cancer Associated Transcript-14 (PCAT-14) as the most prevalent lncRNA that is aberrantly expressed in prostate cancer patients. Down-regulation of PCAT-14 expression significantly associated with Gleason score and a greater probability of metastatic progression, overall survival, and prostate cancer-specific mortality across multiple independent datasets and ethnicities. Low PCAT-14 expression was implicated with genes involved in biological processes promoting aggressive disease. In-vitro analysis confirmed that low PCAT-14 expression increased migration while overexpressing PCAT-14 reduced cellular growth, migration, and invasion.

      CONCLUSIONS: We discovered that androgen-regulated PCAT-14 is overexpressed in prostate cancer, suppresses invasive phenotypes, and lower expression is significantly prognostic for multiple clinical endpoints supporting its significance for predicting metastatic disease that could be used to improve patient management.

      PATIENT SUMMARY: We discovered that aberrant prostate cancer associated transcript-14 expression during prostate cancer progression is prevalent across cancer patients. Prostate cancer associated transcript-14 is also prognostic for metastatic disease and survival highlighting its importance for stratifying patients that could benefit from treatment intensification.

      PMID:27460352 | DOI:10.1016/j.eururo.2016.07.012


      View details for PubMedID 27460352
  • Maternal Embryonic Leucine Zipper Kinase (MELK) as a Novel Mediator and Biomarker of Radioresistance in Human Breast Cancer Clinical cancer research : an official journal of the American Association for Cancer Research
    Speers C, Zhao SG, Kothari V, Santola A, Liu M, Wilder-Romans K, Evans J, Batra N, Bartelink H, Hayes DF, Lawrence TS, Brown PH, Pierce LJ, Feng FY
    2016 Dec 1;22(23):5864-5875. doi: 10.1158/1078-0432.CCR-15-2711. Epub 2016 May 25.
    • More

      PURPOSE: While effective targeted therapies exist for estrogen receptor-positive and HER2-positive breast cancer, no such effective therapies exist for triple-negative breast cancer (TNBC); thus, it is clear that additional targets for radiosensitization and treatment are critically needed.

      EXPERIMENTAL DESIGN: Expression microarrays, qRT-PCR, and Western blotting were used to assess MELK RNA and protein expression levels. Clonogenic survival assays were used to quantitate the radiosensitivity of cell lines at baseline and after MELK inhibition. The effect of MELK knockdown on DNA damage repair kinetics was determined using γH2AX staining. The in vivo effect of MELK knockdown on radiosensitivity was performed using mouse xenograft models. Kaplan-Meier analysis was used to estimate local control and survival information, and a Cox proportional hazards model was constructed to identify potential factors impacting local recurrence-free survival.

      RESULTS: MELK expression is significantly elevated in breast cancer tissues compared with normal tissue as well as in TNBC compared with non-TNBC. MELK RNA and protein expression is significantly correlated with radioresistance in breast cancer cell lines. Inhibition of MELK (genetically and pharmacologically) induces radiation sensitivity in vitro and significantly delayed tumor growth in vivo in multiple models. Kaplan-Meier survival and multivariable analyses identify increasing MELK expression as being the strongest predictor of radioresistance and increased local recurrence in multiple independent datasets.

      CONCLUSIONS: Here, we identify MELK as a potential biomarker of radioresistance and target for radiosensitization in TNBC. Our results support the rationale for developing clinical strategies to inhibit MELK as a novel target in TNBC. Clin Cancer Res; 22(23); 5864-75. ©2016 AACR.

      PMID:27225691 | PMC:PMC8820108 | DOI:10.1158/1078-0432.CCR-15-2711


      View details for PubMedID 27225691
  • Independent validation of the prognostic capacity of the ISUP prostate cancer grade grouping system for radiation treated patients with long-term follow-up Prostate cancer and prostatic diseases
    Spratt DE, Jackson WC, Abugharib A, Tomlins SA, Dess RT, Soni PD, Lee JY, Zhao SG, Cole AI, Zumsteg ZS, Sandler H, Hamstra D, Hearn JW, Palapattu G, Mehra R, Morgan TM, Feng FY
    2016 Sep;19(3):292-7. doi: 10.1038/pcan.2016.18. Epub 2016 May 24.
    • More

      BACKGROUND: There has been a recent proposal to change the grading system of prostate cancer into a five-tier grade grouping system. The prognostic impact of this has been demonstrated in regards only to biochemical recurrence-free survival (bRFS) with short follow-up (3 years).

      METHODS: Between 1990 and 2013, 847 consecutive men were treated with definitive external beam radiation therapy at a single academic center. To validate the new grade grouping system, bRFS, distant metastases-free survival (DMFS) and prostate cancer-specific survival (PCSS) were calculated. Adjusted Kaplan-Meier and multivariable Cox regression analyses were performed to assess the independent impact of the new grade grouping system. Discriminatory analyses were performed to compare the commonly used three-tier Gleason score system (6, 7 and 8-10) to the new system.

      RESULTS: The median follow-up of our cohort was 88 months. The 5-grade groups independently validated differing risks of bRFS (group 1 as reference; adjusted hazard ratio (aHR) 1.35, 2.16, 1.79 and 3.84 for groups 2-5, respectively). Furthermore, a clear stratification was demonstrated for DMFS (aHR 2.03, 3.18, 3.62 and 13.77 for groups 2-5, respectively) and PCSS (aHR 3.00, 5.32, 6.02 and 39.02 for groups 2-5, respectively). The 5-grade group system had improved prognostic discrimination for all end points compared with the commonly used three-tiered system (that is, Gleason score 6, 7 and 8-10).

      CONCLUSIONS: In a large independent radiotherapy cohort with long-term follow-up, we have validated the bRFS benefit of the proposed five-tier grade grouping system. Furthermore, we have demonstrated that the system is highly prognostic for DMFS and PCSS. Grade group 5 had markedly worse outcomes for all end points, and future work is necessary to improve outcomes in these patients.

      PMID:27215611 | DOI:10.1038/pcan.2016.18


      View details for PubMedID 27215611
  • Independent surgical validation of the new prostate cancer grade-grouping system BJU international
    Spratt DE, Cole AI, Palapattu GS, Weizer AZ, Jackson WC, Montgomery JS, Dess RT, Zhao SG, Lee JY, Wu A, Kunju LP, Talmich E, Miller DC, Hollenbeck BK, Tomlins SA, Feng FY, Mehra R, Morgan TM
    2016 Nov;118(5):763-769. doi: 10.1111/bju.13488. Epub 2016 Apr 19.
    • More

      OBJECTIVE: To report the independent prognostic impact of the new prostate cancer grade-grouping system in a large external validation cohort of patients treated with radical prostatectomy (RP).

      PATIENTS AND METHODS: Between 1994 and 2013, 3 694 consecutive men were treated with RP at a single institution. To investigate the performance of and validate the grade-grouping system, biochemical recurrence-free survival (bRFS) rates were assessed using Kaplan-Meier tests, Cox-regression modelling, and discriminatory comparison analyses. Separate analyses were performed based on biopsy and RP grade.

      RESULTS: The median follow-up was 52.7 months. The 5-year actuarial bRFS for biopsy grade groups 1-5 were 94.2%, 89.2%, 73.1%, 63.1%, and 54.7%, respectively (P < 0.001). Similarly, the 5-year actuarial bRFS based on RP grade groups was 96.1%, 93.0%, 74.0%, 64.4%, and 49.9% for grade groups 1-5, respectively (P < 0.001). The adjusted hazard ratios for bRFS relative to biopsy grade group 1 were 1.98, 4.20, 5.57, and 9.32 for groups 2, 3, 4, and 5, respectively (P < 0.001), and for RP grade groups were 2.09, 5.27, 5.86, and 10.42 (P < 0.001). The five-grade-group system had a higher prognostic discrimination compared with the commonly used three-tier system (Gleason score 6 vs 7 vs 8-10).

      CONCLUSIONS: In an independent surgical cohort, we have validated the prognostic benefit of the new prostate cancer grade-grouping system for bRFS, and shown that the benefit is maintained after adjusting for important clinicopathological variables. The greater predictive accuracy of the new system will improve risk stratification in the clinical setting and aid in patient counselling.

      PMID:27009882 | DOI:10.1111/bju.13488


      View details for PubMedID 27009882
  • Patient-Level DNA Damage and Repair Pathway Profiles and Prognosis After Prostatectomy for High-Risk Prostate Cancer JAMA oncology
    Evans JR, Zhao SG, Chang SL, Tomlins SA, Erho N, Sboner A, Schiewer MJ, Spratt DE, Kothari V, Klein EA, Den RB, Dicker AP, Karnes RJ, Yu X, Nguyen PL, Rubin MA, Bono Jd, Knudsen KE, Davicioni E, Feng FY
    2016 Apr;2(4):471-80. doi: 10.1001/jamaoncol.2015.4955.
    • More

      IMPORTANCE: A substantial number of patients diagnosed with high-risk prostate cancer are at risk for metastatic progression after primary treatment. Better biomarkers are needed to identify patients at the highest risk to guide therapy intensification.

      OBJECTIVE: To create a DNA damage and repair (DDR) pathway profiling method for use as a prognostic signature biomarker in high-risk prostate cancer.

      DESIGN, SETTING, AND PARTICIPANTS: A cohort of 1090 patients with high-risk prostate cancer who underwent prostatectomy and were treated at 3 different academic institutions were divided into a training cohort (n = 545) and 3 pooled validation cohorts (n = 232, 130, and 183) assembled for case-control or case-cohort studies. Profiling of 9 DDR pathways using 17 gene sets for GSEA (Gene Set Enrichment Analysis) of high-density microarray gene expression data from formalin-fixed paraffin-embedded prostatectomy samples with median 10.3 years follow-up was performed. Prognostic signature development from DDR pathway profiles was studied, and DDR pathway gene mutation in published cohorts was analyzed.

      MAIN OUTCOMES AND MEASURES: Biochemical recurrence-free, metastasis-free, and overall survival.

      RESULTS: Across the training cohort and pooled validation cohorts, 1090 men were studied; mean (SD) age at diagnosis was 65.3 (6.4) years. We found that there are distinct clusters of DDR pathways within the cohort, and DDR pathway enrichment is only weakly correlated with clinical variables such as age (Spearman ρ [ρ], range, -0.07 to 0.24), Gleason score (ρ, range, 0.03 to 0.20), prostate-specific antigen level (ρ, range, -0.07 to 0.10), while 13 of 17 DDR gene sets are strongly correlated with androgen receptor pathway enrichment (ρ, range, 0.33 to 0.82). In published cohorts, DDR pathway genes are rarely mutated. A DDR pathway profile prognostic signature built in the training cohort was significantly associated with biochemical recurrence-free, metastasis-free, and overall survival in the pooled validation cohorts independent of standard clinicopathological variables. The prognostic performance of the signature for metastasis-free survival appears to be stronger in the younger patients (HR, 1.67; 95% CI, 1.12-2.50) than in the older patients (HR, 0.77; 95% CI, 0.29-2.07) on multivariate Cox analysis.

      CONCLUSIONS AND RELEVANCE: DNA damage and repair pathway profiling revealed patient-level variations and the DDR pathways are rarely affected by mutation. A DDR pathway signature showed strong prognostic performance with the long-term outcomes of metastasis-free and overall survival that may be useful for risk stratification of high-risk prostate cancer patients.

      PMID:26746117 | PMC:PMC5469505 | DOI:10.1001/jamaoncol.2015.4955


      View details for PubMedID 26746117
  • The Landscape of Prognostic Outlier Genes in High-Risk Prostate Cancer Clinical cancer research : an official journal of the American Association for Cancer Research
    Zhao SG, Evans JR, Kothari V, Sun G, Larm A, Mondine V, Schaeffer EM, Ross AE, Klein EA, Den RB, Dicker AP, Karnes RJ, Erho N, Nguyen PL, Davicioni E, Feng FY
    2016 Apr 1;22(7):1777-86. doi: 10.1158/1078-0432.CCR-15-1250. Epub 2015 Dec 2.
    • More

      PURPOSE: There is a clear need to improve risk stratification and to identify novel therapeutic targets in aggressive prostate cancer. The goal of this study was to investigate genes with outlier expression with prognostic association in high-risk prostate cancer patients as potential biomarkers and drug targets.

      EXPERIMENTAL DESIGN: We interrogated microarray gene expression data from prostatectomy samples from 545 high-risk prostate cancer patients with long-term follow-up (mean 13.4 years). Three independent clinical datasets totaling an additional 545 patients were used for validation. Novel prognostic outlier genes were interrogated for impact on oncogenic phenotypes in vitro using siRNA-based knockdown. Association with clinical outcomes and comparison with existing prognostic instruments was assessed with multivariable models using a prognostic outlier score.

      RESULTS: Analysis of the discovery cohort identified 20 prognostic outlier genes. Three top prognostic outlier genes were novel prostate cancer genes; NVL, SMC4, or SQLE knockdown reduced migration and/or invasion and outlier expression was significantly associated with poor prognosis. Increased prognostic outlier score was significantly associated with poor prognosis independent of standard clinicopathologic variables. Finally, the prognostic outlier score prognostic association is independent of, and adds to existing genomic and clinical tools for prognostication in prostate cancer (Decipher, the cell-cycle progression signature, and CAPRA-S).

      CONCLUSIONS: To our knowledge, this study represents the first unbiased high-throughput investigation of prognostic outlier genes in prostate cancer and demonstrates the potential biomarker and therapeutic importance of this previously unstudied class of cancer genes.

      PMID:26631616 | DOI:10.1158/1078-0432.CCR-15-1250


      View details for PubMedID 26631616
  • Maintaining physical activity during head and neck cancer treatment: Results of a pilot controlled trial Head & neck
    Zhao SG, Alexander NB, Djuric Z, Zhou J, Tao Y, Schipper M, Feng FY, Eisbruch A, Worden FP, Strath SJ, Jolly S
    2016 Apr;38 Suppl 1(Suppl 1):E1086-96. doi: 10.1002/hed.24162. Epub 2015 Oct 7.
    • More

      BACKGROUND: Concurrent chemoradiotherapy (concurrent CRT) to treat head and neck cancer is associated with significant reductions of weight, mobility, and quality of life (QOL). An intervention focusing on functional exercise may attenuate these losses.

      METHODS: We allocated patients to a 14-week functional resistance and walking program designed to maintain physical activity during cancer treatment (MPACT group; n = 11), or to usual care (control group; n = 9). Outcomes were assessed at baseline, and 7 and 14 weeks.

      RESULTS: Compared to controls, the MPACT participants had attenuated decline or improvement in several strength, mobility, physical activity, diet, and QOL endpoints. These trends were statistically significant (p < .05) in knee strength, mental health, head and neck QOL, and barriers to exercise.

      CONCLUSION: In this pilot study of patients with head and neck cancer undergoing concurrent CRT, MPACT training was feasible and maintained or improved function and QOL, thereby providing the basis for larger future interventions with longer follow-up. © 2015 Wiley Periodicals, Inc. Head Neck 38: E1086-E1096, 2016.

      PMID:26445898 | PMC:PMC5304917 | DOI:10.1002/hed.24162


      View details for PubMedID 26445898
  • Patient-reported quality of life after stereotactic body radiotherapy (SBRT), intensity modulated radiotherapy (IMRT), and brachytherapy Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology
    Evans JR, Zhao S, Daignault S, Sanda MG, Michalski J, Sandler HM, Kuban DA, Ciezki J, Kaplan ID, Zietman AL, Hembroff L, Feng FY, Suy S, Skolarus TA, McLaughlin PW, Wei JT, Dunn RL, Finkelstein SE, Mantz CA, Collins SP, Hamstra DA, Consortium PS
    2015 Aug;116(2):179-84. doi: 10.1016/j.radonc.2015.07.016. Epub 2015 Aug 11.
    • More

      BACKGROUND AND PURPOSE: Stereotactic body radiotherapy (SBRT) is being used for prostate cancer, but concerns persist about toxicity compared to other radiotherapy options.

      MATERIALS AND METHODS: We conducted a multi-institutional pooled cohort analysis of patient-reported quality of life (QOL) [EPIC-26] before and after intensity-modulated radiotherapy (IMRT), brachytherapy, or SBRT for localized prostate cancer. Data were analyzed by mean domain score, minimal clinically detectable difference (MCD) in domain score, and multivariate analyses to determine factors associated with domain scores at 2-years.

      RESULTS: Data were analyzed from 803 patients at baseline and 645 at 2-years. Mean declines at 2-years across all patients were -1.9, -4.8, -4.9, and -13.3 points for urinary obstructive, urinary incontinence, bowel, and sexual symptom domains, respectively, corresponding to MCD in 29%, 20%, and 28% of patients. On multivariate analysis (vs. IMRT), brachytherapy had worse urinary irritation at 2-years (-6.8 points, p<0.0001) but no differences in other domains (p>0.15). QOL after SBRT was similar for urinary (p>0.5) and sexual domains (p=0.57), but was associated with better bowel score (+6.7 points, p<0.0002).

      CONCLUSIONS: QOL 2-years after brachytherapy, IMRT, or SBRT is very good and largely similar, with small differences in urinary and bowel QOL that are likely minimized by modern techniques.

      PMID:26276528 | DOI:10.1016/j.radonc.2015.07.016


      View details for PubMedID 26276528
  • DNA-PKcs-Mediated Transcriptional Regulation Drives Prostate Cancer Progression and Metastasis Cancer cell
    Goodwin JF, Kothari V, Drake JM, Zhao S, Dylgjeri E, Dean JL, Schiewer MJ, McNair C, Jones JK, Aytes A, Magee MS, Snook AE, Zhu Z, Den RB, Birbe RC, Gomella LG, Graham NA, Vashisht AA, Wohlschlegel JA, Graeber TG, Karnes RJ, Takhar M, Davicioni E, Tomlins SA, Abate-Shen C, Sharifi N, Witte ON, Feng FY, Knudsen KE
    2015 Jul 13;28(1):97-113. doi: 10.1016/j.ccell.2015.06.004.
    • More

      Emerging evidence demonstrates that the DNA repair kinase DNA-PKcs exerts divergent roles in transcriptional regulation of unsolved consequence. Here, in vitro and in vivo interrogation demonstrate that DNA-PKcs functions as a selective modulator of transcriptional networks that induce cell migration, invasion, and metastasis. Accordingly, suppression of DNA-PKcs inhibits tumor metastases. Clinical assessment revealed that DNA-PKcs is significantly elevated in advanced disease and independently predicts for metastases, recurrence, and reduced overall survival. Further investigation demonstrated that DNA-PKcs in advanced tumors is highly activated, independent of DNA damage indicators. Combined, these findings reveal unexpected DNA-PKcs functions, identify DNA-PKcs as a potent driver of tumor progression and metastases, and nominate DNA-PKcs as a therapeutic target for advanced malignancies.

      PMID:26175416 | PMC:PMC4531387 | DOI:10.1016/j.ccell.2015.06.004


      View details for PubMedID 26175416
  • Endothelial CXCR7 regulates breast cancer metastasis Oncogene
    Stacer AC, Fenner J, Cavnar SP, Xiao A, Zhao S, Chang SL, Salomonnson A, Luker KE, Luker GD
    2016 Mar 31;35(13):1716-24. doi: 10.1038/onc.2015.236. Epub 2015 Jun 29.
    • More

      Atypical chemokine receptor CXCR7 (ACKR3) functions as a scavenger receptor for chemokine CXCL12, a molecule that promotes multiple steps in tumor growth and metastasis in breast cancer and multiple other malignancies. Although normal vascular endothelium expresses low levels of CXCR7, marked upregulation of CXCR7 occurs in tumor vasculature in breast cancer and other tumors. To investigate effects of endothelial CXCR7 in breast cancer, we conditionally deleted this receptor from vascular endothelium of adult mice, generating CXCR7(ΔEND/ΔEND) animals. CXCR7(ΔEND/ΔEND) mice appeared phenotypically normal, although these animals exhibited a modest 35±3% increase in plasma CXCL12 as compared with control. Using two different syngeneic, orthotopic tumor implant models of breast cancer, we discovered that CXCR7(ΔEND/ΔEND) mice had significantly greater local recurrence of cancer following resection, elevated numbers of circulating tumor cells and more spontaneous metastases. CXCR7(ΔEND/ΔEND) mice also showed greater experimental metastases following intracardiac injection of cancer cells. These results establish that endothelial CXCR7 limits breast cancer metastasis at multiple steps in the metastatic cascade, advancing understanding of CXCL12 pathways in tumor environments and informing ongoing drug development targeting CXCR7 in cancer.

      PMID:26119946 | PMC:PMC4486335 | DOI:10.1038/onc.2015.236


      View details for PubMedID 26119946
  • High-throughput transcriptomic analysis nominates proteasomal genes as age-specific biomarkers and therapeutic targets in prostate cancer Prostate cancer and prostatic diseases
    Zhao SG, Jackson WC, Kothari V, Schipper MJ, Erho N, Evans JR, Speers C, Hamstra DA, Niknafs YS, Nguyen PL, Schaeffer EM, Ross AE, Den RB, Klein EA, Jenkins RB, Davicioni E, Feng FY
    2015 Sep;18(3):229-36. doi: 10.1038/pcan.2015.22. Epub 2015 May 19.
    • More

      BACKGROUND: Although prostate cancer (PCa) is hypothesized to differ in nature between younger versus older patients, the underlying molecular distinctions are poorly understood. We hypothesized that high-throughput transcriptomic analysis would elucidate biological differences in PCas arising in younger versus older men, and would nominate potential age-specific biomarkers and therapeutic targets.

      METHODS: The high-density Affymetrix GeneChip platform, encompassing >1 million genomic loci, was utilized to assess gene expression in 1090 radical prostatectomy samples from patients with long-term follow-up. We identified genes associated with metastatic progression by 10 years post-treatment in younger (age<65) versus older (age⩾65) patients, and ranked these genes by their prognostic value. We performed Gene Set Enrichment Analysis (GSEA) to nominate biological concepts that demonstrated age-specific effects, and validated a target by treating with a clinically available drug in three PCa cell lines derived from younger men.

      RESULTS: Over 80% of the top 1000 prognostic genes in younger and older men were specific to that age group. GSEA nominated the proteasome pathway as the most differentially prognostic in younger versus older patients. High expression of proteasomal genes conferred worse prognosis in younger but not older men on univariate and multivariate analysis. Bortezomib, a Food and Drug Administration approved proteasome inhibitor, decreased proliferation in three PCa cell lines derived from younger patients.

      CONCLUSIONS: Our data show significant global differences in prognostic genes between older versus younger men. We nominate proteasomeal gene expression as an age-specific biomarker and potential therapeutic target specifically in younger men. Limitations of our study include clinical differences between cohorts, and increased comorbidities and lower survival in older patients. These intriguing findings suggest that current models of PCa biology do not adequately represent genetic heterogeneity of PCa related to age, and future clinical trials would benefit from stratification based on age.

      PMID:25986914 | PMC:PMC4579590 | DOI:10.1038/pcan.2015.22


      View details for PubMedID 25986914
  • Development and validation of a novel platform-independent metastasis signature in human breast cancer PloS one
    Zhao SG, Shilkrut M, Speers C, Liu M, Wilder-Romans K, Lawrence TS, Pierce LJ, Feng FY
    2015 May 14;10(5):e0126631. doi: 10.1371/journal.pone.0126631. eCollection 2015.
    • More

      PURPOSE: The molecular drivers of metastasis in breast cancer are not well understood. Therefore, we sought to identify the biological processes underlying distant progression and define a prognostic signature for metastatic potential in breast cancer.

      EXPERIMENTAL DESIGN: In vivo screening for metastases was performed using Chick Chorioallantoic Membrane assays in 21 preclinical breast cancer models. Expressed genes associated with metastatic potential were identified using high-throughput analysis. Correlations with biological function were determined using the Database for Annotation, Visualization and Integrated Discovery.

      RESULTS: We identified a broad range of metastatic potential that was independent of intrinsic breast cancer subtypes. 146 genes were significantly associated with metastasis progression and were linked to cancer-related biological functions, including cell migration/adhesion, Jak-STAT, TGF-beta, and Wnt signaling. These genes were used to develop a platform-independent gene expression signature (M-Sig), which was trained and subsequently validated on 5 independent cohorts totaling nearly 1800 breast cancer patients with all p-values < 0.005 and hazard ratios ranging from approximately 2.5 to 3. On multivariate analysis accounting for standard clinicopathologic prognostic variables, M-Sig remained the strongest prognostic factor for metastatic progression, with p-values < 0.001 and hazard ratios > 2 in three different cohorts.

      CONCLUSION: M-Sig is strongly prognostic for metastatic progression, and may provide clinical utility in combination with treatment prediction tools to better guide patient care. In addition, the platform-independent nature of the signature makes it an excellent research tool as it can be directly applied onto existing, and future, datasets.

      PMID:25974184 | PMC:PMC4431866 | DOI:10.1371/journal.pone.0126631


      View details for PubMedID 25974184
  • Characterization of 1577 primary prostate cancers reveals novel biological and clinicopathologic insights into molecular subtypes European urology
    Tomlins SA, Alshalalfa M, Davicioni E, Erho N, Yousefi K, Zhao S, Haddad Z, Den RB, Dicker AP, Trock BJ, DeMarzo AM, Ross AE, Schaeffer EM, Klein EA, Magi-Galluzzi C, Karnes RJ, Jenkins RB, Feng FY
    2015 Oct;68(4):555-67. doi: 10.1016/j.eururo.2015.04.033. Epub 2015 May 8.
    • More

      BACKGROUND: Prostate cancer (PCa) molecular subtypes have been defined by essentially mutually exclusive events, including ETS gene fusions (most commonly involving ERG) and SPINK1 overexpression. Clinical assessment may aid in disease stratification, complementing available prognostic tests.

      OBJECTIVE: To determine the analytical validity and clinicopatholgic associations of microarray-based molecular subtyping.

      DESIGN, SETTING, AND PARTICIPANTS: We analyzed Affymetrix GeneChip expression profiles for 1577 patients from eight radical prostatectomy cohorts, including 1351 cases assessed using the Decipher prognostic assay (GenomeDx Biosciences, San Diego, CA, USA) performed in a laboratory with Clinical Laboratory Improvements Amendment certification. A microarray-based (m-) random forest ERG classification model was trained and validated. Outlier expression analysis was used to predict other mutually exclusive non-ERG ETS gene rearrangements (ETS(+)) or SPINK1 overexpression (SPINK1(+)).

      OUTCOME MEASUREMENTS: Associations with clinical features and outcomes by multivariate logistic regression analysis and receiver operating curves.

      RESULTS AND LIMITATIONS: The m-ERG classifier showed 95% accuracy in an independent validation subset (155 samples). Across cohorts, 45% of PCas were classified as m-ERG(+), 9% as m-ETS(+), 8% as m-SPINK1(+), and 38% as triple negative (m-ERG(-)/m-ETS(-)/m-SPINK1(-)). Gene expression profiling supports three underlying molecularly defined groups: m-ERG(+), m-ETS(+), and m-SPINK1(+)/triple negative. On multivariate analysis, m-ERG(+) tumors were associated with lower preoperative serum prostate-specific antigen and Gleason scores, but greater extraprostatic extension (p<0.001). m-ETS(+) tumors were associated with seminal vesicle invasion (p=0.01), while m-SPINK1(+)/triple negative tumors had higher Gleason scores and were more frequent in Black/African American patients (p<0.001). Clinical outcomes were not significantly different among subtypes.

      CONCLUSIONS: A clinically available prognostic test (Decipher) can also assess PCa molecular subtypes, obviating the need for additional testing. Clinicopathologic differences were found among subtypes based on global expression patterns.

      PATIENT SUMMARY: Molecular subtyping of prostate cancer can be achieved using extra data generated from a clinical-grade, genome-wide expression-profiling prognostic assay (Decipher). Transcriptomic and clinical analysis support three distinct molecular subtypes: (1) m-ERG(+), (2) m-ETS(+), and (3) m-SPINK1(+)/triple negative (m-ERG(-)/m-ETS(-)/m-SPINK1(-)). Incorporation of subtyping into a clinically available assay may facilitate additional applications beyond routine prognosis.

      PMID:25964175 | PMC:PMC4562381 | DOI:10.1016/j.eururo.2015.04.033


      View details for PubMedID 25964175
  • Development and Validation of a Novel Radiosensitivity Signature in Human Breast Cancer Clinical cancer research : an official journal of the American Association for Cancer Research
    Speers C, Zhao S, Liu M, Bartelink H, Pierce LJ, Feng FY
    2015 Aug 15;21(16):3667-77. doi: 10.1158/1078-0432.CCR-14-2898. Epub 2015 Apr 22.
    • More

      PURPOSE: An unmet clinical need in breast cancer management is the accurate identification of patients who will benefit from adjuvant radiotherapy. We hypothesized that integration of postradiation clonogenic survival data with gene expression data across breast cancer cell (BCC) lines would generate a radiation sensitivity signature (RSS) and identify patients with tumors refractive to conventional therapy.

      EXPERIMENTAL DESIGN: Using clonogenic survival assays, we identified the surviving fraction (SF-2Gy) after radiation across a range of BCC lines. Intrinsic radiosensitivity was correlated to gene expression using Spearman correlation. Functional analysis was performed in vitro, and enriched biologic concepts were identified. The RSS was generated using a Random Forest model and was refined, cross-validated, and independently validated in additional breast cancer datasets.

      RESULTS: Clonogenic survival identifies a range of radiosensitivity in human BCC lines (SF-2Gy 77%-17%) with no significant correlation to the intrinsic breast cancer subtypes. One hundred forty-seven genes were correlated with radiosensitivity. Functional analysis of RSS genes identifies previously unreported radioresistance-associated genes. RSS was trained, cross-validated, and further refined to 51 genes that were enriched for concepts involving cell-cycle arrest and DNA damage response. RSS was validated in an independent dataset and was the most significant factor in predicting local recurrence on multivariate analysis, outperfoming all clinically used clinicopathologic features.

      CONCLUSIONS: We derive a human breast cancer-specific RSS with biologic relevance and validate this signature for prediction of locoregional recurrence. By identifying patients with tumors refractory to standard radiation this signature has the potential to allow for personalization of radiotherapy.

      PMID:25904749 | DOI:10.1158/1078-0432.CCR-14-2898


      View details for PubMedID 25904749
  • The landscape of long noncoding RNAs in the human transcriptome Nature genetics
    Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, Barrette TR, Prensner JR, Evans JR, Zhao S, Poliakov A, Cao X, Dhanasekaran SM, Wu Y, Robinson DR, Beer DG, Feng FY, Iyer HK, Chinnaiyan AM
    2015 Mar;47(3):199-208. doi: 10.1038/ng.3192. Epub 2015 Jan 19.
    • More

      Long noncoding RNAs (lncRNAs) are emerging as important regulators of tissue physiology and disease processes including cancer. To delineate genome-wide lncRNA expression, we curated 7,256 RNA sequencing (RNA-seq) libraries from tumors, normal tissues and cell lines comprising over 43 Tb of sequence from 25 independent studies. We applied ab initio assembly methodology to this data set, yielding a consensus human transcriptome of 91,013 expressed genes. Over 68% (58,648) of genes were classified as lncRNAs, of which 79% were previously unannotated. About 1% (597) of the lncRNAs harbored ultraconserved elements, and 7% (3,900) overlapped disease-associated SNPs. To prioritize lineage-specific, disease-associated lncRNA expression, we employed non-parametric differential expression testing and nominated 7,942 lineage- or cancer-associated lncRNA genes. The lncRNA landscape characterized here may shed light on normal biology and cancer pathogenesis and may be valuable for future biomarker development.

      PMID:25599403 | PMC:PMC4417758 | DOI:10.1038/ng.3192


      View details for PubMedID 25599403
  • RNA biomarkers associated with metastatic progression in prostate cancer: a multi-institutional high-throughput analysis of SChLAP1 The Lancet. Oncology
    Prensner JR, Zhao S, Erho N, Schipper M, Iyer MK, Dhanasekaran SM, Magi-Galluzzi C, Mehra R, Sahu A, Siddiqui J, Davicioni E, Den RB, Dicker AP, Karnes RJ, Wei JT, Klein EA, Jenkins RB, Chinnaiyan AM, Feng FY
    2014 Dec;15(13):1469-1480. doi: 10.1016/S1470-2045(14)71113-1. Epub 2014 Nov 17.
    • More

      BACKGROUND: Improved clinical predictors for disease progression are needed for localised prostate cancer, since only a subset of patients develop recurrent or refractory disease after first-line treatment. Therefore, we undertook an unbiased analysis to identify RNA biomarkers associated with metastatic progression after prostatectomy.

      METHODS: Prostate cancer samples from patients treated with radical prostatectomy at three academic institutions were analysed for gene expression by a high-density Affymetrix GeneChip platform, encompassing more than 1 million genomic loci. In a discovery cohort, all protein-coding genes and known long non-coding RNAs were ranked by fold change in expression between tumours that subsequently metastasised versus those that did not. The top ranked gene was then validated for its prognostic value for metastatic progression in three additional independent cohorts. 95% of the gene expression assays were done in a Clinical Laboratory Improvements Amendments certified laboratory facility. All genes were assessed for their ability to predict metastatic progression by receiver-operating-curve area-under-the-curve analyses. Multivariate analyses were done for the primary endpoint of metastatic progression, with variables including Gleason score, preoperative prostate-specific antigen concentration, seminal vesicle invasion, surgical margin status, extracapsular extension, lymph node invasion, and expression of the highest ranked gene.

      FINDINGS: 1008 patients were included in the study: 545 in the discovery cohort and 463 in the validation cohorts. The long non-coding RNA SChLAP1 was identified as the highest-ranked overexpressed gene in cancers with metastatic progression. Validation in three independent cohorts confirmed the prognostic value of SChLAP1 for metastatic progression. On multivariate modelling, SChLAP1 expression (high vs low) independently predicted metastasis within 10 years (odds ratio [OR] 2·45, 95% CI 1·70-3·53; p<0·0001). The only other variable that independently predicted metastasis within 10 years was Gleason score (8-10 vs 5-7; OR 2·14, 95% CI 1·77-2·58; p<0·0001).

      INTERPRETATION: We identified and validated high SChLAP1 expression as significantly prognostic for metastatic disease progression of prostate cancer. Our findings suggest that further development of SChLAP1 as a potential biomarker, for treatment intensification in aggressive prostate cancer, warrants future study.

      FUNDING: Prostate Cancer Foundation, National Institutes of Health, Department of Defense, Early Detection Research Network, Doris Duke Charitable Foundation, and Howard Hughes Medical Institute.

      PMID:25456366 | PMC:PMC4559342 | DOI:10.1016/S1470-2045(14)71113-1


      View details for PubMedID 25456366
  • Etanercept plus topical corticosteroids as initial therapy for grade one acute graft-versus-host disease after allogeneic hematopoietic cell transplantation Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation
    Gatza E, Braun T, Levine JE, Ferrara LM, Zhao S, Wang T, Chang L, Harris A, Pawarode A, Kitko C, Magenau JM, Yanik GA, Couriel DR, Goldstein S, Connelly J, Reddy P, Paczesny S, Choi SW
    2014 Sep;20(9):1426-34. doi: 10.1016/j.bbmt.2014.05.023. Epub 2014 Jun 2.
    • More

      Clinical diagnosis of grade 1 acute graft-versus-host disease (GVHD) marks the beginning of a potentially progressive and fatal course of GVHD after hematopoietic stem cell transplantation (HSCT). However, interventional studies to treat early GVHD are lacking. We conducted a single-arm prospective phase II trial to test the hypothesis that treatment of newly diagnosed grade 1 acute GVHD with etanercept and topical corticosteroids would reduce progression to grade 2 to 4 within 28 days. Study patients (n = 34) had a median age of 51 years (range, 10 to 67 years) and had undergone unrelated (n = 22) or related (n = 12) donor HSCT. Study patients were treated with etanercept (.4 mg/kg, maximum 25 mg/dose) twice weekly for 4 to 8 weeks. Ten of 34 patients (29%) progressed to grade 2 to 4 acute GVHD within 28 days. The cumulative incidence of grade 2 to 4 and grade 3 to 4 acute GVHD at 1 year was 41% and 3%, respectively. Nonrelapse mortality was 19% and overall survival was 63% at 2 years. Among a contemporaneous control cohort of patients who were diagnosed with grade 1 acute GVHD and treated with topical corticosteroids but not etanercept during the study period, 12 of 28 patients (43%) progressed to grade 2 to 4 GVHD within 28 days, with a 1-year incidence of grade 2 to 4 GVHD and grade 3 to 4 GVHD of 61% (41% versus 61%, P = .08) and 18% (3% versus 18%, P = .05), respectively. Patients treated with etanercept also experienced less increase in GVHD plasma biomarkers suppression of tumorigenicity 2 (P = .06) and regenerating islet-derived 3-alpha (P = .01) 28 days after grade 1 acute GVHD diagnosis compared with contemporaneous control patients. This study was terminated early because of poor accrual. Future prospective studies are needed to identify patients with grade 1 acute GVHD at risk of swift progression to more severe GVHD and to establish consensus for the treatment of grade 1 acute GVHD. This trial is registered with ClinicalTrials.gov, number NCT00726375.

      PMID:24892263 | PMC:PMC4145722 | DOI:10.1016/j.bbmt.2014.05.023


      View details for PubMedID 24892263
  • Engraftment syndrome after allogeneic hematopoietic cell transplantation predicts poor outcomes Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation
    Chang L, Frame D, Braun T, Gatza E, Hanauer DA, Zhao S, Magenau JM, Schultz K, Tokala H, Ferrara LM, Levine JE, Reddy P, Paczesny S, Choi SW
    2014 Sep;20(9):1407-17. doi: 10.1016/j.bbmt.2014.05.022. Epub 2014 Jun 2.
    • More

      Engraftment syndrome (ES), characterized by fever, rash, pulmonary edema, weight gain, liver and renal dysfunction, and/or encephalopathy, occurs at the time of neutrophil recovery after hematopoietic cell transplantation (HCT). In this study, we evaluated the incidence, clinical features, risk factors, and outcomes of ES in children and adults undergoing first-time allogeneic HCT. Among 927 patients, 119 (13%) developed ES at a median of 10 days (interquartile range 9 to 12) after HCT. ES patients experienced significantly higher cumulative incidence of grade 2 to 4 acute GVHD at day 100 (75% versus 34%, P < .001) and higher nonrelapse mortality at 2 years (38% versus 19%, P < .001) compared with non-ES patients, resulting in lower overall survival at 2 years (38% versus 54%, P < .001). There was no significant difference in relapse at 2 years (26% versus 31%, P = .772). Suppression of tumorigenicity 2, interleukin 2 receptor alpha, and tumor necrosis factor receptor 1 plasma biomarker levels were significantly elevated in ES patients. Our results illustrate the clinical significance and prognostic impact of ES on allogeneic HCT outcomes. Despite early recognition of the syndrome and prompt institution of corticosteroid therapy, outcomes in ES patients were uniformly poor. This study suggests the need for a prospective approach of collecting clinical features combined with correlative laboratory analyses to better characterize ES.

      PMID:24892262 | PMC:PMC4142041 | DOI:10.1016/j.bbmt.2014.05.022


      View details for PubMedID 24892262
  • A Comprehensive Analysis of CXCL12 Isoforms in Breast Cancer<sup>1,2</sup> Translational oncology
    Zhao S, Chang SL, Linderman JJ, Feng FY, Luker GD
    2014 May 13;7(3):429-38. doi: 10.1016/j.tranon.2014.04.001. Online ahead of print.
    • More

      CXCL12-CXCR4-CXCR7 signaling promotes tumor growth and metastasis in breast cancer. Alternative splicing of CXCL12 produces isoforms with distinct structural and biochemical properties, but little is known about isoform-specific differences in breast cancer subtypes and patient outcomes. We investigated global expression profiles of the six CXCL12 isoforms, CXCR4, and CXCR7 in The Cancer Genome Atlas breast cancer cohort using next-generation RNA sequencing in 948 breast cancer and benign samples and seven breast cancer cell lines. We compared expression levels with several clinical parameters, as well as metastasis, recurrence, and overall survival (OS). CXCL12-α, -β, and -γ are highly co-expressed, with low expression correlating with more aggressive subtypes, higher stage disease, and worse clinical outcomes. CXCL12-δ did not correlate with other isoforms but was prognostic for OS and showed the same trend for metastasis and recurrence-free survival. Effects of CXCL12-δ remained independently prognostic when taking into account expression of CXCL12,CXCR4, and CXCR7. These results were also reflected when comparing CXCL12-α, -β, and -γ in breast cancer cell lines. We summarized expression of all CXCL12 isoforms in an important chemokine signaling pathway in breast cancer in a large clinical cohort and common breast cancer cell lines, establishing differences among isoforms in multiple clinical, pathologic, and molecular subgroups. We identified for the first time the clinical importance of a previously unstudied isoform, CXCL12-δ.

      PMID:24836649 | PMC:PMC4145355 | DOI:10.1016/j.tranon.2014.04.001


      View details for PubMedID 24836649
  • Central nervous system endoplasmic reticulum stress in a murine model of type 2 diabetes Diabetologia
    Sims-Robinson C, Zhao S, Hur J, Feldman EL
    2012 Aug;55(8):2276-84. doi: 10.1007/s00125-012-2573-6. Epub 2012 May 14.
    • More

      AIMS/HYPOTHESIS: Type 2 diabetes is associated with complications in the central nervous system (CNS), including learning and memory, and an increased risk for neurodegenerative diseases. The mechanism underlying this association is not understood. The aim of this study was to gain greater insight into the possible mechanisms of diabetes-induced cognitive decline.

      METHODS: We used microarray technology to identify and examine changes in gene expression in the hippocampus of a murine model of type 2 diabetes, the db/db mouse. Bioinformatics approaches were then used to investigate the biological significance of these genes. To validate the biological significance we evaluated mRNA and protein levels.

      RESULTS: At 8 and 24 weeks, 256 and 822 genes, respectively, were differentially expressed in the db/db mice. The most significantly enriched biological functions were related to mitochondria, heat shock proteins, or the endoplasmic reticulum (ER), the majority of which were downregulated. The ER-enriched cluster was one of the clusters that contained the highest number of differentially expressed genes. Several of the downregulated genes that were differentially expressed at 24 but not at 8 weeks are directly involved in the unfolded protein response (UPR) pathway and include two heat shock proteins (encoded by Hspa5 and Hsp90b1), a transcriptional factor (x-box binding protein 1, encoded by Xbp1), and an apoptotic mediator (DNA-damage inducible transcript 3, encoded by Ddit3).

      CONCLUSIONS/INTERPRETATION: The changes that we observed in the UPR pathway due to ER stress may play a role in the pathogenesis of CNS complications in diabetes. The results of this study are a foundation for the development of pharmacological targets to reduce ER stress in diabetic hippocampi.

      PMID:22581041 | PMC:PMC3391332 | DOI:10.1007/s00125-012-2573-6


      View details for PubMedID 22581041
  • Epidemiology and risk factors for thromboembolic complications of childhood nephrotic syndrome: a Midwest Pediatric Nephrology Consortium (MWPNC) study The Journal of pediatrics
    Kerlin BA, Blatt NB, Fuh B, Zhao S, Lehman A, Blanchong C, Mahan JD, Smoyer WE
    2009 Jul;155(1):105-10, 110.e1. doi: 10.1016/j.jpeds.2009.01.070. Epub 2009 Apr 24.
    • More

      OBJECTIVES: To identify clinical variables predictive of the risk of thromboembolism (TE), and to confirm the incidence of TE in primary and secondary childhood nephrotic syndrome (NS).

      STUDY DESIGN: A comprehensive chart review identified 326 children with NS from any cause evaluated between 1999 and 2006. These patients had a total of 1472.8 patient-years of follow-up. Comparison statistics, survival analysis, and logistic regression were used to define TE epidemiology and clinical risk factors.

      RESULTS: We found that 9.2% of our cohort had experienced at least 1 TE. The overall incidence was 20.4 patients with TEs/1000 patient-years. The median time to the first TE was 70.5 days after diagnosis of NS. Deep venous thrombosis was the most common TE (76%) and was frequently associated with the use of a central venous catheter (45%). Significant independent predictors of TE included age > or = 12 years at onset of NS (P < .0001), severity of proteinuria (P < .0001), and history of TE preceding diagnosis of NS (P < .0001). Life- or limb-threatening TEs represented 23.7% of the events.

      CONCLUSIONS: Children with NS should be carefully followed for TE, particularly those who are age 12 years or older, have severe proteinuria, or have a previous history of TE.

      PMID:19394032 | PMC:PMC3685482 | DOI:10.1016/j.jpeds.2009.01.070


      View details for PubMedID 19394032
  • Significance of hemolysis on extracorporeal life support after cardiac surgery in children Pediatric nephrology (Berlin, Germany)
    Gbadegesin R, Zhao S, Charpie J, Brophy PD, Smoyer WE, Lin J
    2009 Mar;24(3):589-95. doi: 10.1007/s00467-008-1047-z. Epub 2008 Nov 12.
    • More

      Hemolysis is common during extracorporeal life support (ECLS). Elevated levels of circulating plasma free hemoglobin (FHb) has been linked to the development of hemoglobinuria nephropathy. Its clinical significance in patients receiving ECLS remains unknown. Medical records of 104 children <3 years old who required ECLS after repair of congenital heart disease were reviewed. Forty-two patients required continuous renal replacement therapy (CRRT) during ECLS (CRRT group), and 62 patients did not (no-CRRT group). For all patients, FHb level and the degree of fluid overload at the end of ECLS predicted the mortality rate during ECLS. Compared with the no-CRRT group, the CRRT group had a higher mortality rate during ECLS, a higher peak FHb level during ECLS, a higher FHb level at the end of ECLS, and more days of ECLS. In the CRRT group, the FHb level at the end of ECLS predicted death during ECLS. In the no-CRRT group, the peak FHb level was associated with a worse renal function. In conclusion, elevated FHb levels were associated with renal dysfunction and death during ECLS in children undergoing cardiac surgery. Further studies are needed to elucidate the cause-effect relationship in our findings.

      PMID:19002722 | DOI:10.1007/s00467-008-1047-z


      View details for PubMedID 19002722

Contact Information

Shuang (George) Zhao, MD


Email