171 results on '"Adam G. Sowalsky"'
Search Results
52. Data from Sequential Prostate Magnetic Resonance Imaging in Newly Diagnosed High-risk Prostate Cancer Treated with Neoadjuvant Enzalutamide is Predictive of Therapeutic Response
- Author
-
William L. Dahut, Peter A. Pinto, Baris Turkbey, Adam G. Sowalsky, James L. Gulley, Peter L. Choyke, Huihui Ye, Rosina T. Lis, Monique N. Williams, Amy Hankin, Anna Couvillon, Marijo Bilusic, Guinevere Chun, Nicolas T. Terrigino, John R. Bright, Nicole V. Carrabba, Lisa M. Cordes, David J. VanderWeele, Stephanie A. Harmon, Maria J. Merino, Joanna H. Shih, Ravi A. Madan, Scott Wilkinson, Stephanie M. Walker, and Fatima Karzai
- Abstract
Purpose:For high-risk prostate cancer, standard treatment options include radical prostatectomy (RP) or radiotherapy plus androgen deprivation therapy (ADT). Despite definitive therapy, many patients will have disease recurrence. Imaging has the potential to better define characteristics of response and resistance. In this study, we evaluated prostate multiparametric MRI (mpMRI) before and after neoadjuvant enzalutamide plus ADT.Patients and Methods:Men with localized intermediate- or high-risk prostate cancer underwent a baseline mpMRI and mpMRI-targeted biopsy followed by a second mpMRI after 6 months of enzalutamide and ADT prior to RP. Specimens were sectioned in the same plane as mpMRI using patient-specific 3D-printed molds to permit mpMRI-targeted biopsies to be compared with the same lesion from the RP. Specimens were analyzed for imaging and histologic correlates of response.Results:Of 39 patients enrolled, 36 completed imaging and RP. Most patients (92%) had high-risk disease. Fifty-eight lesions were detected on baseline mpMRI, of which 40 (69%) remained measurable at 6-month follow-up imaging. Fifty-five of 59 lesions (93%) demonstrated >50% volume reduction on posttreatment mpMRI. Three of 59 lesions (5%) demonstrated growth in size at follow-up imaging, with two lesions increasing more than 3-fold in volume. On whole-mount pathology, 15 patients demonstrated minimal residual disease (MRD) of Conclusions:Low relative lesion volume at baseline mpMRI was predictive of pathologic response. A subset of patients had limited response. Selection of patients based on these metrics may improve outcomes in high-risk disease.
- Published
- 2023
- Full Text
- View/download PDF
53. Supplementary Figures from A PDX/Organoid Biobank of Advanced Prostate Cancers Captures Genomic and Phenotypic Heterogeneity for Disease Modeling and Therapeutic Screening
- Author
-
Kathleen Kelly, Eva Corey, William L. Dahut, Fatima H. Karzai, Aian Neil Alilin, JuanJuan Yin, Kerry M. McGowen, Qi Yang, Keith H. Jansson, Supreet Agarwal, Adam G. Sowalsky, Holly M. Nguyen, Crystal Tran, Caitlin M. Tice, and Michael L. Beshiri
- Abstract
Supplementary Figures
- Published
- 2023
- Full Text
- View/download PDF
54. Supplementary Tables from A PDX/Organoid Biobank of Advanced Prostate Cancers Captures Genomic and Phenotypic Heterogeneity for Disease Modeling and Therapeutic Screening
- Author
-
Kathleen Kelly, Eva Corey, William L. Dahut, Fatima H. Karzai, Aian Neil Alilin, JuanJuan Yin, Kerry M. McGowen, Qi Yang, Keith H. Jansson, Supreet Agarwal, Adam G. Sowalsky, Holly M. Nguyen, Crystal Tran, Caitlin M. Tice, and Michael L. Beshiri
- Abstract
Supplementary Tables
- Published
- 2023
- Full Text
- View/download PDF
55. Data from A Subset of Localized Prostate Cancer Displays an Immunogenic Phenotype Associated with Losses of Key Tumor Suppressor Genes
- Author
-
David J. Einstein, Steven P. Balk, Adam G. Sowalsky, Haydn Kissick, Sabina Signoretti, Huihui Ye, Philip J. Saylor, David B. Sykes, Olga Voznesensky, Anson T. Ku, Ross Lake, Scott Wilkinson, Luke del Balzo, Taghreed Hirz, Caroline S. Jansen, Miriam Ficial, and Carla Calagua
- Abstract
Purpose:A subset of primary prostate cancer expresses programmed death-ligand 1 (PD-L1), but whether they have a unique tumor immune microenvironment or genomic features is unclear.Experimental Design:We selected PD-L1–positive high-grade and/or high-risk primary prostate cancer, characterized tumor-infiltrating lymphocytes with multiplex immunofluorescence, and identified genomic alterations in immunogenic and nonimmunogenic tumor foci.Results:One quarter of aggressive localized prostate cancer cases (29/115) had tumor PD-L1 expression more than 5%. This correlated with increased density of CD8+ T cells, a large fraction coexpressing PD-1, versus absent PD-1 expression on sparse CD8 T cells in unselected cases. Most CD8+PD-1+ cells did not express terminal exhaustion markers (TIM3 or LAG3), while a subset expressed TCF1. Consistent with these CD8+PD-1+TCF1+ cells being progenitors, they were found in antigen-presenting cell niches in close proximity to MHC-II+ cells. CD8 T-cell density in immunogenic prostate cancer and renal cell carcinoma (RCC) was nearly identical. Shallow RB1 and BRCA2 losses, and deep deletions of CHD1, were prevalent, the latter being strongly associated with a dendritic cell gene set in The Cancer Genome Atlas. Tumor mutation burden was variable; neither high microsatellite instability nor CDK12 alterations were present.Conclusions:A subset of localized prostate cancer is immunogenic, manifested by PD-L1 expression and CD8+ T-cell content comparable with RCC. The CD8+ T cells include effector cells and exhausted progenitor cells, which may be expanded by immune checkpoint inhibitors (ICI). Genomic losses of RB1, BRCA2, and CHD1 may be drivers of this phenotype. These findings indicate that immunotherapies may be effective in biomarker-selected subpopulations of patients with localized prostate cancer.
- Published
- 2023
- Full Text
- View/download PDF
56. Data from ErbB2 Signaling Increases Androgen Receptor Expression in Abiraterone-Resistant Prostate Cancer
- Author
-
Changmeng Cai, Steven P. Balk, Mary-Ellen Taplin, Peter S. Nelson, Elahe A. Mostaghel, Ziyang Yu, Wanting Han, Olga Voznesensky, Adam G. Sowalsky, Akash Patnaik, Sen Chen, Ankur Sharma, Hongyun Wang, Sean Gerrin, Huihui Ye, and Shuai Gao
- Abstract
Purpose: ErbB2 signaling appears to be increased and may enhance androgen receptor (AR) activity in a subset of patients with castration-resistant prostate cancer (CRPC), but agents targeting ErbB2 have not been effective. This study was undertaken to assess ErbB2 activity in abiraterone-resistant prostate cancer and to determine whether it may contribute to AR signaling in these tumors.Experimental Design: AR activity and ErbB2 signaling were examined in the radical prostatectomy specimens from a neoadjuvant clinical trial of leuprolide plus abiraterone and in the specimens from abiraterone-resistant CRPC xenograft models. The effect of ErbB2 signaling on AR activity was determined in two CRPC cell lines. Moreover, the effect of combination treatment with abiraterone and an ErbB2 inhibitor was assessed in a CRPC xenograft model.Results: We found that ErbB2 signaling was elevated in residual tumor following abiraterone treatment in a subset of patients and was associated with higher nuclear AR expression. In xenograft models, we similarly demonstrated that ErbB2 signaling was increased and associated with AR reactivation in abiraterone-resistant tumors. Mechanistically, we show that ErbB2 signaling and subsequent activation of the PI3K/AKT signaling stabilizes AR protein. Furthermore, concomitantly treating CRPC cells with abiraterone and an ErbB2 inhibitor, lapatinib, blocked AR reactivation and suppressed tumor progression.Conclusions: ErbB2 signaling is elevated in a subset of patients with abiraterone-resistant prostate cancer and stabilizes AR protein. Combination therapy with abiraterone and ErbB2 antagonists may be effective for treating the subset of CRPC with elevated ErbB2 activity. Clin Cancer Res; 22(14); 3672–82. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
57. Data from A PDX/Organoid Biobank of Advanced Prostate Cancers Captures Genomic and Phenotypic Heterogeneity for Disease Modeling and Therapeutic Screening
- Author
-
Kathleen Kelly, Eva Corey, William L. Dahut, Fatima H. Karzai, Aian Neil Alilin, JuanJuan Yin, Kerry M. McGowen, Qi Yang, Keith H. Jansson, Supreet Agarwal, Adam G. Sowalsky, Holly M. Nguyen, Crystal Tran, Caitlin M. Tice, and Michael L. Beshiri
- Abstract
Purpose: Prostate cancer translational research has been hampered by the lack of comprehensive and tractable models that represent the genomic landscape of clinical disease. Metastatic castrate-resistant prostate cancer (mCRPC) patient-derived xenografts (PDXs) recapitulate the genetic and phenotypic diversity of the disease. We sought to establish a representative, preclinical platform of PDX-derived organoids that is experimentally facile for high-throughput and mechanistic analysis.Experimental Design: Using 20 models from the LuCaP mCRPC PDX cohort, including adenocarcinoma and neuroendocrine lineages, we systematically tested >20 modifications to prostate organoid conditions. Organoids were evaluated for genomic and phenotypic stability and continued reliance on the AR signaling pathway. The utility of the platform as a genotype-dependent model of drug sensitivity was tested with olaparib and carboplatin.Results: All PDX models proliferated as organoids in culture. Greater than 50% could be continuously cultured long-term in modified conditions; however, none of the PDXs could be established long-term as organoids under previously reported conditions. In addition, the modified conditions improved the establishment of patient biopsies over current methods. The genomic heterogeneity of the PDXs was conserved in organoids. Lineage markers and transcriptomes were maintained between PDXs and organoids. Dependence on AR signaling was preserved in adenocarcinoma organoids, replicating a dominant characteristic of CRPC. Finally, we observed maximum cytotoxicity to the PARP inhibitor olaparib in BRCA2−/− organoids, similar to responses observed in patients.Conclusions: The LuCaP PDX/organoid models provide an expansive, genetically characterized platform to investigate the mechanisms of pathogenesis as well as therapeutic responses and their molecular correlates in mCRPC. Clin Cancer Res; 24(17); 4332–45. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
58. Supplementary Data from Sequential Prostate Magnetic Resonance Imaging in Newly Diagnosed High-risk Prostate Cancer Treated with Neoadjuvant Enzalutamide is Predictive of Therapeutic Response
- Author
-
William L. Dahut, Peter A. Pinto, Baris Turkbey, Adam G. Sowalsky, James L. Gulley, Peter L. Choyke, Huihui Ye, Rosina T. Lis, Monique N. Williams, Amy Hankin, Anna Couvillon, Marijo Bilusic, Guinevere Chun, Nicolas T. Terrigino, John R. Bright, Nicole V. Carrabba, Lisa M. Cordes, David J. VanderWeele, Stephanie A. Harmon, Maria J. Merino, Joanna H. Shih, Ravi A. Madan, Scott Wilkinson, Stephanie M. Walker, and Fatima Karzai
- Abstract
Contains supplementary methods and 3 supplementary tables.
- Published
- 2023
- Full Text
- View/download PDF
59. Data from Neoadjuvant-Intensive Androgen Deprivation Therapy Selects for Prostate Tumor Foci with Diverse Subclonal Oncogenic Alterations
- Author
-
Steven P. Balk, Mary-Ellen Taplin, Peter S. Nelson, Elahe A. Mostaghel, Bruce Montgomery, Glenn J. Bubley, Zhenwei Zhang, Olga S. Voznesensky, Rachel J. Schaefer, Joshua W. Russo, Fen Ma, Carla Calagua, Laleh Montaser-Kouhsari, Rosina T. Lis, Massimo Loda, Eliezer M. Van Allen, Manoj Bhasin, Huihui Ye, and Adam G. Sowalsky
- Abstract
Primary prostate cancer can have extensive microheterogeneity, but its contribution to the later emergence of metastatic castration-resistant prostate cancer (mCRPC) remains unclear. In this study, we microdissected residual prostate cancer foci in radical prostatectomies from 18 men treated with neoadjuvant-intensive androgen deprivation therapy (leuprolide, abiraterone acetate, and prednisone) and analyzed them for resistance mechanisms. Transcriptome profiling showed reduced but persistent androgen receptor (AR) activity in residual tumors, with no increase in neuroendocrine differentiation. Proliferation correlated negatively with AR activity but positively with decreased RB1 expression, and whole-exome sequencing (WES) further showed enrichment for RB1 genomic loss. In 15 cases where 2 or 3 tumor foci were microdissected, WES confirmed a common clonal origin but identified multiple oncogenic alterations unique to each focus. These findings show that subclones with oncogenic alterations found in mCRPC are present in primary prostate cancer and are selected for by neoadjuvant-intense androgen deprivation therapy. In particular, this study indicates that subclonal RB1 loss may be more common than previously appreciated in intermediate- to high-risk primary prostate cancer and may be an early event, independent of neuroendocrine differentiation, in the development of mCRPC. Comprehensive molecular analyses of primary prostate cancer may detect aggressive subclones and possibly inform adjuvant strategies to prevent recurrence.Significance: Neoadjuvant androgen deprivation therapy for prostate cancer selects for tumor foci with subclonal genomic alterations, which may comprise the origin of metastatic castration-resistant prostate cancer. Cancer Res; 78(16); 4716–30. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
60. Supplementary Figures 1-5 from RalA Function in Dermal Fibroblasts Is Required for the Progression of Squamous Cell Carcinoma of the Skin
- Author
-
Larry A. Feig, Jonathan A. Garlick, Yulia Shamis, Addy Alt-Holland, and Adam G. Sowalsky
- Abstract
Supplementary Figures 1-5 from RalA Function in Dermal Fibroblasts Is Required for the Progression of Squamous Cell Carcinoma of the Skin
- Published
- 2023
- Full Text
- View/download PDF
61. Data from RalA Function in Dermal Fibroblasts Is Required for the Progression of Squamous Cell Carcinoma of the Skin
- Author
-
Larry A. Feig, Jonathan A. Garlick, Yulia Shamis, Addy Alt-Holland, and Adam G. Sowalsky
- Abstract
A large body of evidence has shown that stromal cells play a significant role in determining the fate of neighboring tumor cells through the secretion of various cytokines. How cytokine secretion by stromal cells is regulated in this context is poorly understood. In this study, we used a bioengineered human tissue model of skin squamous cell carcinoma progression to reveal that RalA function in dermal fibroblasts is required for tumor progression of neighboring neoplastic keratinocytes. This conclusion is based on the observations that suppression of RalA expression in dermal fibroblasts blocked tumorigenic keratinocytes from invading into the dermal compartment of engineered tissues and suppressed more advanced tumor progression after these tissues were transplanted onto the dorsum of mice. RalA executes this tumor-promoting function of dermal fibroblasts, at least in part, by mediating hepatocyte growth factor (HGF) secretion through its effector proteins, the Sec5 and Exo84 subunits of the exocyst complex. These findings reveal a new level of HGF regulation and highlight the RalA signaling cascade in dermal fibroblasts as a potential anticancer target. Cancer Res; 71(3); 758–67. ©2010 AACR.
- Published
- 2023
- Full Text
- View/download PDF
62. Supplementary Figures 1-3 and Tables 1-5 from Clonal Progression of Prostate Cancers from Gleason Grade 3 to Grade 4
- Author
-
Steven P. Balk, Glenn J. Bubley, Huihui Ye, and Adam G. Sowalsky
- Abstract
PDF file-801KB, Supplementary Figure 1: H&E, PIN-4, and ERG stains on consecutive sections of cancer tissue from Patient 1, 3, and 4 that were used for laser capture microdissection and breakpoint mapping. Supplementary Figure 2: Breakpoint sequence confirmation using Sanger sequencing. Supplementary Figure 3: H&E, PIN-4, and ERG stains on representative sections of tissue showing the presence of ERG-positive and ERG-negative PIN lesions adjacent to ERG-positive cancer tissue. Supplementary Table 1: Summary of TMPRSS2:ERG expression and clinicopathologic characteristics of 52 cases with adjacent G3 and G4 tumor foci stained with anti-ERG. Supplementary Table 2: Percentage of TMPRSS2:ERG Illumina reads for each sample. Supplementary Table 3: The location of the TMPRSS2 and ERG breakpoints for each patient, the mechanism of ERG fusion inferred by SNP genotyping of the interstitial region (either a deletion or balanced translocation) and the type of double-stranded break and end joining indicated by each fusion. Supplmenetary Table 4: Percentage of PTEN Illumina reads for each sample. Supplementary Table 5: SNPs present in the PTEN locus for Patient 3 in G3 (left) and G4 (right) DNA, as determined by Illumina sequencing.
- Published
- 2023
- Full Text
- View/download PDF
63. Data from Clonal Progression of Prostate Cancers from Gleason Grade 3 to Grade 4
- Author
-
Steven P. Balk, Glenn J. Bubley, Huihui Ye, and Adam G. Sowalsky
- Abstract
Low-grade prostate cancers (Gleason pattern 3, G3) detected on needle biopsies are generally viewed as indolent and suitable for conservative management with only interval repeat biopsies to monitor by watchful waiting. Higher grade tumors eventually emerge in 20% to 30% of these cases, but this process may only reflect incomplete sampling on the initial biopsy, such that it remains unknown whether G3 tumors generally progress to higher grades. In this study, we examined a series of adjacent G3 and Gleason pattern 4 (G4) tumors in radical prostatectomy specimens and found that all were concordant for the TMPRSS2:ERG gene fusion. Using hybrid-capture and deep sequencing in four fusion-positive cases, we found that adjacent laser-capture microdissected G3 and G4 tumors had identical TMPRSS2:ERG fusion breakpoints, confirming their clonal origin. Two of these G3 tumors had deletion of a single PTEN gene that was also deleted in the adjacent G4, while the G4 tumors in two cases had additional PTEN losses. These findings establish that a subset of G3 tumors progress to G4 or emerge from a common precursor. Further, they show that G3 tumors that progress to G4 may have molecular features distinguishing them from G3 tumors that do not progress. Thus, determining the spectrum of these genetic or epigenetic features may allow for the identification of G3 tumors on needle biopsies that are truly indolent versus those that have the potential to progress or that may already be associated with a G4 tumor that was not sampled at the initial biopsy, therefore, requiring more aggressive surveillance or intervention. Cancer Res; 73(3); 1050–5. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
64. Supplementary Methods and Materials from RalA Function in Dermal Fibroblasts Is Required for the Progression of Squamous Cell Carcinoma of the Skin
- Author
-
Larry A. Feig, Jonathan A. Garlick, Yulia Shamis, Addy Alt-Holland, and Adam G. Sowalsky
- Abstract
Supplementary Methods and Materials from RalA Function in Dermal Fibroblasts Is Required for the Progression of Squamous Cell Carcinoma of the Skin
- Published
- 2023
- Full Text
- View/download PDF
65. Supplementary Methods and Materials, Figure Legends, and References from Clonal Progression of Prostate Cancers from Gleason Grade 3 to Grade 4
- Author
-
Steven P. Balk, Glenn J. Bubley, Huihui Ye, and Adam G. Sowalsky
- Abstract
PDF file-124KB, The Supplementary Methods and Materials section contains the procedures for DNA/RNA extraction, Real-Time PCR, Library Preparation and Deep Sequencing, Data Alignment, Breakpoint Sequencing, and Determining PTEN Status.
- Published
- 2023
- Full Text
- View/download PDF
66. Supplementary Figure Legends 1-5 from RalA Function in Dermal Fibroblasts Is Required for the Progression of Squamous Cell Carcinoma of the Skin
- Author
-
Larry A. Feig, Jonathan A. Garlick, Yulia Shamis, Addy Alt-Holland, and Adam G. Sowalsky
- Abstract
Supplementary Figure Legends 1-5 from RalA Function in Dermal Fibroblasts Is Required for the Progression of Squamous Cell Carcinoma of the Skin
- Published
- 2023
- Full Text
- View/download PDF
67. AR Inhibition Increases MHC Class I Expression and Improves Immune Response in Prostate Cancer
- Author
-
Lisa N. Chesner, Julie N. Graff, Fanny Polesso, Alexis Smith, Arian Lundberg, Rajdeep Das, Tanushree Shenoy, Zheng Xia, Ya-Mei Hu, Martin Sjöström, Simon Linder, William S. Chen, Adam Foye, Haolong Li, Lisa Kim, Megha Bhalla, Thomas O’loughlin, Duygu Kuzuoglu-Ozturk, Tony Hua, Scott Wilkinson, Shana Y. Trostel, Andries M. Bergman, Davide Ruggero, Charles G. Drake, Adam G. Sowalsky, Lawrence Fong, Matthew Cooperberg, Alan Ashworth, Wilbert Zwart, David A. Quigley, Luke A. Gilbert, Amy E. Moran, and Felix Feng
- Published
- 2023
- Full Text
- View/download PDF
68. Defining cellular population dynamics at single-cell resolution during prostate cancer progression
- Author
-
Alexandre A Germanos, Sonali Arora, Ye Zheng, Erica T Goddard, Ilsa M Coleman, Anson T Ku, Scott Wilkinson, Hanbing Song, Nicholas J Brady, Robert A Amezquita, Michael Zager, Annalysa Long, Yu Chi Yang, Jason H Bielas, Raphael Gottardo, David S Rickman, Franklin W Huang, Cyrus M Ghajar, Peter S Nelson, Adam G Sowalsky, Manu Setty, and Andrew C Hsieh
- Subjects
Male ,Urologic Diseases ,PTEN ,Aging ,Mouse ,Population Dynamics ,immune microenvironment ,mRNA Translation ,General Biochemistry, Genetics and Molecular Biology ,Single cell RNAseq ,Androgen ,Mice ,computational biology ,Receptors ,Tumor Microenvironment ,Genetics ,Animals ,Humans ,2.1 Biological and endogenous factors ,Aetiology ,cancer biology ,Cancer ,General Immunology and Microbiology ,General Neuroscience ,Prostate Cancer ,Prostate ,Prostatic Neoplasms ,systems biology ,General Medicine ,epithelial cells ,Receptors, Androgen ,Disease Progression ,Androgens ,Biochemistry and Cell Biology ,Orchiectomy - Abstract
Advanced prostate malignancies are a leading cause of cancer-related deaths in men, in large part due to our incomplete understanding of cellular drivers of disease progression. We investigate prostate cancer cell dynamics at single-cell resolution from disease onset to the development of androgen independence in an in vivo murine model. We observe an expansion of a castration-resistant intermediate luminal cell type that correlates with treatment resistance and poor prognosis in human patients. Moreover, transformed epithelial cells and associated fibroblasts create a microenvironment conducive to pro-tumorigenic immune infiltration, which is partially androgen responsive. Androgen-independent prostate cancer leads to significant diversification of intermediate luminal cell populations characterized by a range of androgen signaling activity, which is inversely correlated with proliferation and mRNA translation. Accordingly, distinct epithelial populations are exquisitely sensitive to translation inhibition, which leads to epithelial cell death, loss of pro-tumorigenic signaling, and decreased tumor heterogeneity. Our findings reveal a complex tumor environment largely dominated by castration-resistant luminal cells and immunosuppressive infiltrates.
- Published
- 2022
69. Author response: Defining cellular population dynamics at single-cell resolution during prostate cancer progression
- Author
-
Alexandre A Germanos, Sonali Arora, Ye Zheng, Erica T Goddard, Ilsa M Coleman, Anson T Ku, Scott Wilkinson, Hanbing Song, Nicholas J Brady, Robert A Amezquita, Michael Zager, Annalysa Long, Yu Chi Yang, Jason H Bielas, Raphael Gottardo, David S Rickman, Franklin W Huang, Cyrus M Ghajar, Peter S Nelson, Adam G Sowalsky, Manu Setty, and Andrew C Hsieh
- Published
- 2022
- Full Text
- View/download PDF
70. Loss of tristetraprolin activates NF-κB induced phenotypic plasticity and primes transition to lethal prostate cancer
- Author
-
Katherine L. Morel, Anis A. Hamid, Beatriz G. Falcón, Jagpreet S. Nanda, Simon Linder, Andries M. Bergman, Henk van der Poel, Ingrid Hofland, Elise M. Bekers, Shana Trostel, Scott Wilkinson, Anson T. Ku, Deborah L. Burkhart, Minhyung Kim, Jina Kim, Jasmine T. Plummer, Sungyong You, Adam G. Sowalsky, Wilbert Zwart, Christopher J. Sweeney, and Leigh Ellis
- Abstract
Phenotypic plasticity is a hallmark of cancer and increasingly realized as a mechanism of resistance in androgen indifferent prostate tumors. It is critical to identify mechanisms and actionable targets driving phenotypic plasticity. Here, we report that loss of tristetraprolin (TTP, geneZFP36), an RNA binding protein that regulates mRNA stability increases NF-κB activation and is associated with higher rates of aggressive disease and early recurrence in primary prostate cancer (PCa). We examined the clinical and biological impact ofZFP36loss combined withPTENloss, a known driver of PCa. Combined loss ofPTENandZFP36expression was associated with increased risk of recurrence in multiple independent primary PCa cohorts, and significantly reduced overall survival and time to progression following castration in genetically engineered mouse models.ZFP36loss alters the cell state that is driven byPTENloss, demonstrated by positive enrichment of gene sets including EMT, inflammation, TNFα/NF-κB, IL6-JAK/STAT3.ZFP36loss also induces enrichment of multiple gene sets involved in cell migration, chemotaxis, and proliferation. Use of the NF-κB inhibitor dimethylaminoparthenolide induced significant therapeutic responses in tumors withPTENandZFP36co-loss and reversed castration resistance. This work identifies a novel molecular mechanism driving phenotypic plasticity and castration resistance through loss ofZFP36expression, that can be reversed by inhibition of NF-κB activity.
- Published
- 2022
- Full Text
- View/download PDF
71. EZH2 inhibition activates a dsRNA–STING–interferon stress axis that potentiates response to PD-1 checkpoint blockade in prostate cancer
- Author
-
Anis A. Hamid, Sukanya Panja, Carla Calagua, Housheng Hansen He, Israel Cañadas, Leigh Ellis, Nichelle C. Whitlock, Brian Olson, Steven P. Balk, Phillip M. Galbo, Geoffrey I. Shapiro, Stephanie K. Dougan, Mark Pomerantz, Nadia Boufaied, Deborah L. Burkhart, Anjali V. Sheahan, Sylvan C. Baca, David J. Einstein, Antonina Mitrofanova, Shana Y. Trostel, Constantia Pantelidou, Max Heckler, Yin Liu, Atish D. Choudhury, David P. Labbé, Adam G. Sowalsky, Massimo Loda, Kevin Roehle, Christopher Sweeney, Katherine L. Morel, Scott Wilkinson, Matthew L. Freedman, Xintao Qiu, Huihui Ye, Adam S. Kibel, David A. Barbie, and Henry W. Long
- Subjects
Male ,Cancer Research ,Chemokine ,Programmed Cell Death 1 Receptor ,Antigen presentation ,macromolecular substances ,CD8-Positive T-Lymphocytes ,Article ,Prostate cancer ,Interferon ,Prostate ,Humans ,Medicine ,Enhancer of Zeste Homolog 2 Protein ,RNA, Double-Stranded ,biology ,business.industry ,EZH2 ,Prostatic Neoplasms ,medicine.disease ,Sting ,medicine.anatomical_structure ,Oncology ,Cancer research ,biology.protein ,Interferons ,business ,CD8 ,medicine.drug - Abstract
Prostate cancers are considered to be immunologically ‘cold’ tumors given the very few patients who respond to checkpoint inhibitor (CPI) therapy. Recently, enrichment of interferon-stimulated genes (ISGs) predicted a favorable response to CPI across various disease sites. The enhancer of zeste homolog-2 (EZH2) is overexpressed in prostate cancer and known to negatively regulate ISGs. In the present study, we demonstrate that EZH2 inhibition in prostate cancer models activates a double-stranded RNA–STING–ISG stress response upregulating genes involved in antigen presentation, Th1 chemokine signaling and interferon response, including programmed cell death protein 1 (PD-L1) that is dependent on STING activation. EZH2 inhibition substantially increased intratumoral trafficking of activated CD8(+) T cells and increased M1 tumor-associated macrophages, overall reversing resistance to PD-1 CPI. Our study identifies EZH2 as a potent inhibitor of antitumor immunity and responsiveness to CPI. These data suggest EZH2 inhibition as a therapeutic direction to enhance prostate cancer response to PD-1 CPI.
- Published
- 2021
- Full Text
- View/download PDF
72. Sequential Prostate Magnetic Resonance Imaging in Newly Diagnosed High-risk Prostate Cancer Treated with Neoadjuvant Enzalutamide is Predictive of Therapeutic Response
- Author
-
Baris Turkbey, Peter A. Pinto, Joanna H. Shih, Maria Merino, Peter L. Choyke, Ravi A. Madan, John R. Bright, Stephanie Harmon, Guinevere Chun, Fatima Karzai, Huihui Ye, Amy Hankin, Scott Wilkinson, Anna Couvillon, Marijo Bilusic, Lisa M. Cordes, Nicolas T. Terrigino, David J. VanderWeele, Rosina T. Lis, William L. Dahut, Nicole V. Carrabba, Stephanie M. Walker, Monique N. Williams, James L. Gulley, and Adam G. Sowalsky
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,030232 urology & nephrology ,Article ,Androgen deprivation therapy ,03 medical and health sciences ,Prostate cancer ,chemistry.chemical_compound ,0302 clinical medicine ,Risk Factors ,Prostate ,Antineoplastic Combined Chemotherapy Protocols ,Nitriles ,Outcome Assessment, Health Care ,Phenylthiohydantoin ,medicine ,Humans ,Enzalutamide ,Multiparametric Magnetic Resonance Imaging ,Fatigue ,Aged ,medicine.diagnostic_test ,business.industry ,Prostatectomy ,Prostatic Neoplasms ,Androgen Antagonists ,Magnetic resonance imaging ,Middle Aged ,medicine.disease ,Minimal residual disease ,Neoadjuvant Therapy ,Tumor Burden ,Radiation therapy ,medicine.anatomical_structure ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Benzamides ,Hot Flashes ,Radiology ,business - Abstract
Purpose: For high-risk prostate cancer, standard treatment options include radical prostatectomy (RP) or radiotherapy plus androgen deprivation therapy (ADT). Despite definitive therapy, many patients will have disease recurrence. Imaging has the potential to better define characteristics of response and resistance. In this study, we evaluated prostate multiparametric MRI (mpMRI) before and after neoadjuvant enzalutamide plus ADT. Patients and Methods: Men with localized intermediate- or high-risk prostate cancer underwent a baseline mpMRI and mpMRI-targeted biopsy followed by a second mpMRI after 6 months of enzalutamide and ADT prior to RP. Specimens were sectioned in the same plane as mpMRI using patient-specific 3D-printed molds to permit mpMRI-targeted biopsies to be compared with the same lesion from the RP. Specimens were analyzed for imaging and histologic correlates of response. Results: Of 39 patients enrolled, 36 completed imaging and RP. Most patients (92%) had high-risk disease. Fifty-eight lesions were detected on baseline mpMRI, of which 40 (69%) remained measurable at 6-month follow-up imaging. Fifty-five of 59 lesions (93%) demonstrated >50% volume reduction on posttreatment mpMRI. Three of 59 lesions (5%) demonstrated growth in size at follow-up imaging, with two lesions increasing more than 3-fold in volume. On whole-mount pathology, 15 patients demonstrated minimal residual disease (MRD) of Conclusions: Low relative lesion volume at baseline mpMRI was predictive of pathologic response. A subset of patients had limited response. Selection of patients based on these metrics may improve outcomes in high-risk disease.
- Published
- 2021
- Full Text
- View/download PDF
73. Re: Heng Li, Yucong Zhang, Dong Li, et al. Androgen Receptor Splice Variant 7 Predicts Shorter Response in Patients with Metastatic Hormone-sensitive Prostate Cancer Receiving Androgen Deprivation Therapy. Eur Urol 2021;79:879–86 AR-V7 is Rare in Hormone-sensitive Prostate Cancer
- Author
-
Adam G. Sowalsky, Stephen R. Plymate, Michael C. Haffner, Johann S. de Bono, and Adam Sharp
- Subjects
Male ,Prostatic Neoplasms, Castration-Resistant ,Receptors, Androgen ,Urology ,Phenylthiohydantoin ,Androgens ,Humans ,Protein Isoforms ,Androgen Antagonists - Published
- 2022
- Full Text
- View/download PDF
74. Clinical outcome following checkpoint therapy in renal cell carcinoma is associated with a burst of activated CD8 T cells in blood
- Author
-
Jennifer Wilkinson Carlisle, Caroline S Jansen, Maria Andrea Cardenas, Ewelina Sobierajska, Adriana Moon Reyes, Rachel Greenwald, Luke Del Balzo, Nataliya Prokhnevska, Omer Kucuk, Bradley C Carthon, Patrick Connor Mullane, Adeboye Osunkoya, Deborah Baumgarten, Fares Hosseinzadeh, Scott Wilkinson, Ross Lake, Adam G Sowalsky, Yuan Liu, Viraj A Master, Mehmet A Bilen, and Haydn Kissick
- Subjects
Pharmacology ,Cancer Research ,Oncology ,Immunology ,Receptors, Antigen, T-Cell ,Molecular Medicine ,Immunology and Allergy ,Humans ,HLA-DR Antigens ,CD8-Positive T-Lymphocytes ,Carcinoma, Renal Cell ,Immune Checkpoint Inhibitors ,Kidney Neoplasms - Abstract
PurposeCheckpoint therapy is now the cornerstone of treatment for patients with renal cell carcinoma (RCC) with advanced disease, but biomarkers are lacking to predict which patients will benefit. This study proposes potential immunological biomarkers that could developed for predicting therapeutic response in patients with RCC.MethodsUsing flow cytometry, RNA sequencing, and T-cell receptor (TCR) sequencing, we investigated changes in T cells in the peripheral blood of patients with advanced RCC after receiving immunotherapy. We used immunofluorescence (IF) imaging and flow cytometry to investigate how intratumoral T cells in patients’ tumors (resected months/years prior to receiving checkpoint therapy) predicted patient outcomes after immunotherapy.ResultsWe found that a small proportion of CD4 and CD8 T cells in the blood activate following checkpoint therapy, expressing the proliferation marker Ki67 and activation markers HLA-DR and CD38. Patients who had the highest increase in these HLA-DR +CD38+CD8 T cells after treatment had the best antitumor immune response and experienced clinical benefit. Using RNA sequencing, we found that while these cells expanded in most patients, their phenotype did not drastically change during treatment. However, when we analyzed the TCR repertoire of these HLA-DR +CD38+CD8+T cells, we found that only patients who clinically benefitted had a burst of new clonotypes enter this pool of activated cells. Finally, we found that abundant T cells in the untreated tumors predicted clinical benefit to checkpoint therapy on disease progression.ConclusionsTogether, these data suggest that having a strong pre-existing immune response and immediate peripheral T-cell activation after checkpoint therapy is a predictor of clinical benefit in patients with RCC.
- Published
- 2022
75. Prediction of cancer treatment response from histopathology images through imputed transcriptomics
- Author
-
Danh-Tai Hoang, Gal Dinstag, Leandro C. Hermida, Doreen S. Ben-Zvi, Efrat Elis, Katherine Caley, Stephen-John Sammut, Sanju Sinha, Neelam Sinha, Christopher H. Dampier, Chani Stossel, Tejas Patil, Arun Rajan, Wiem Lassoued, Julius Strauss, Shania Bailey, Clint Allen, Jason Redman, Tuvik Beker, Peng Jiang, Talia Golan, Scott Wilkinson, Adam G. Sowalsky, Sharon R. Pine, Carlos Caldas, James L. Gulley, Kenneth Aldape, Ranit Aharonov, Eric A. Stone, and Eytan Ruppin
- Abstract
Advances in artificial intelligence have paved the way for leveraging hematoxylin and eosin (H&E)-stained tumor slides for precision oncology. We present ENLIGHT-DeepPT, an approach for predicting response to multiple targeted and immunotherapies from H&E-slides. In difference from existing approaches that aim to predict treatment response directly from the slides, ENLIGHT-DeepPT is an indirect two-step approach consisting of (1) DeepPT, a new deep-learning framework that predicts genome-wide tumor mRNA expression from slides, and (2) ENLIGHT, which predicts response based on the DeepPT inferred expression values. DeepPT successfully predicts transcriptomics in all 16 TCGA cohorts tested and generalizes well to two independent datasets. Importantly, ENLIGHT-DeepPT successfully predicts true responders in five independent patients’ cohorts involving four different treatments spanning six cancer types with an overall odds ratio of 2.44, increasing the baseline response rate by 43.47% among predicted responders, without the need for any treatment data for training. Furthermore, its prediction accuracy on these datasets is comparable to a supervised approach predicting the response directly from the images, trained and tested on the same cohort in cross validation. Its future application could provide clinicians with rapid treatment recommendations to an array of different therapies and importantly, may contribute to advancing precision oncology in developing countries.Statement of SignificanceENLIGHT-DeepPT is the first approach shown to successfully predict response tomultipletargeted and immune cancer therapies from H&E slides. In distinction from all previous H&E slides prediction approaches, it does not require supervised training on a specific cohort for each drug/indication treatment but is trained to predict expression on the TCGA cohort and then can predict response to an array of treatments without any further training. ENLIGHT-DeepPT can provide rapid treatment recommendations to oncologists and help advance precision oncology in underserved regions and low-income countries.
- Published
- 2022
- Full Text
- View/download PDF
76. Tumor-derived biomarkers beyond antigen expression enhance efficacy of CD276/B7H3 antibody-drug conjugate in metastatic prostate cancer
- Author
-
Supreet Agarwal, Lei Fang, Kerry McGowen, JuanJuan Yin, Joel Bowman, Anson T. Ku, Aian Neil Alilin, Eva Corey, Martine Roudier, Lawrence True, Ruthy Dumpit, Ilsa Coleman, John Lee, Peter S. Nelson, Brian J. Capaldo, Aida Mariani, Clare Hoover, Ilya S. Senatorov, Michael Beshiri, Adam G. Sowalsky, Elaine M Hurt, and Kathleen Kelly
- Abstract
Antibody-drug conjugates (ADCs) are promising targeted cancer therapy; however, patient selection based solely on target antigen expression without consideration for cytotoxic payload vulnerabilities has plateaued clinical benefits. Biomarkers to capture patients who might benefit from specific ADCs have not been systematically determined for any cancer. We present a comprehensive therapeutic and biomarker analysis of a B7H3-ADC with pyrrolobenzodiazepine(PBD) payload in 26 treatment-resistant, metastatic prostate cancer(mPC) models. B7H3 is a tumor-specific surface protein widely expressed in mPC, and PBD is a DNA cross-linking agent. B7H3 expression was necessary but not sufficient for B7H3-PBD-ADC responsiveness. RB1 deficiency and/or replication stress, characteristics of poor prognosis, conferred sensitivity and were associated with complete tumor regression in both neuroendocrine (NEPC) and androgen receptor positive(ARPC) prostate cancer models, even with low B7H3 levels. Non-ARPC models, which are currently lacking efficacious treatment, demonstrated the highest replication stress and were most sensitive to treatment. In RB1 wild-type ARPC tumors, SLFN11 expression or select DNA repair mutations in SLFN11 non-expressors governed response. Significantly, wild-type TP53 predicted non-responsiveness (7/8 models). Overall, biomarker-focused selection of models led to high efficacy of in vivo treatment. These data enable a paradigm shift to biomarker-driven trial designs for maximizing clinical benefit of ADC therapies.
- Published
- 2022
- Full Text
- View/download PDF
77. MEIS1 down-regulation by MYC mediates prostate cancer development through elevated HOXB13 expression and AR activity
- Author
-
Rayann Atway, Huihui Ye, Elizabeth D. Walton, Nicole V. Carrabba, S. Thomas Hennigan, Nicholas T. Terrigino, Nichelle C. Whitlock, Brian J. Capaldo, Ross Lake, Shana Y. Trostel, Adam G. Sowalsky, Scott Wilkinson, and Berkley E. Gryder
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Down-Regulation ,Biology ,medicine.disease_cause ,Article ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Downregulation and upregulation ,Prostate ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Myeloid Ecotropic Viral Integration Site 1 Protein ,Cancer genetics ,Molecular Biology ,Transcription factor ,Homeodomain Proteins ,Regulation of gene expression ,Gene knockdown ,Gene Expression Profiling ,Prostatic Neoplasms ,medicine.disease ,Phenotype ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Cancer research ,Carcinogenesis - Abstract
Localized prostate cancer develops very slowly in most men, with the androgen receptor (AR) and MYC transcription factors amongst the most well-characterized drivers of prostate tumorigenesis. Canonically, MYC up-regulation in luminal prostate cancer cells functions to oppose the terminally differentiating effects of AR. However, the effects of MYC up-regulation are pleiotropic and inconsistent with a poorly proliferative phenotype. Here we show that increased MYC expression and activity are associated with the down-regulation of MEIS1, a HOX-family transcription factor. Using RNA-seq to profile a series of human prostate cancer specimens laser capture microdissected on the basis of MYC immunohistochemistry, MYC activity, and MEIS1 expression were inversely correlated. Knockdown of MYC expression in prostate cancer cells increased the expression of MEIS1 and increased the occupancy of MYC at the MEIS1 locus. Finally, we show in laser capture microdissected human prostate cancer samples and the prostate TCGA cohort that MEIS1 expression is inversely proportional to AR activity as well as HOXB13, a known interacting protein of both AR and MEIS1. Collectively, our data demonstrate that elevated MYC in a subset of primary prostate cancers functions in a negative role in regulating MEIS1 expression, and that this down-regulation may contribute to MYC-driven development and progression.
- Published
- 2020
- Full Text
- View/download PDF
78. A cancer stem cell population underlies a multi-lineage phenotype and drug resistance in prostate cancer
- Author
-
Michael L. Beshiri, Brian J. Capaldo, Ross Lake, Anson T. Ku, Danielle Burner, Caitlin M. Tice, Crystal Tran, Julianna Kostas, Aian Neil Alilin, JuanJuan Yin, Supreet Agarwal, Samantha A. Morris, Fatima H. Karzai, Tamara L. Lotan, William L. Dahut, Adam G. Sowalsky, and Kathleen Kelly
- Abstract
SUMMARYTo resist lineage-dependent therapies, cancer cells adopt a plastic stem-like state, leading to phenotypic heterogeneity. Here we dissect the cellular origins of such heterogeneity in a metastatic castration-resistant prostate cancer (CRPC) patient-derived adenocarcinoma organoid model displaying a range of luminal and neuroendocrine phenotypes and driven by mutations in cell cycle (CDKN1B) and epigenetic (ARID1A, and ARID1B) regulators. As shown by lineage tracing, metastatic tumor heterogeneity originated from distinct subclones of infrequent stem/progenitor cells that each produced a full distribution of differentiated lineage markers, suggesting multiclonal evolution to a relatively stable bipotential state. Single cell ATAC-seq analyses revealed the co-occurrence of transcription factor activities associated with multiple disparate lineages in the stem/progenitors: WNT and RXR stem factors, AR and FOXA1 luminal epithelial drivers, and NR2F1 and ASCL1 neural factors. Inhibition of AR in combination with AURKA but not EZH2 blocked tumor growth. These data provide insight into the origins and dynamics of cancer cell plasticity and stem targeted therapy.
- Published
- 2022
- Full Text
- View/download PDF
79. Defining cellular population dynamics at single cell resolution during prostate cancer progression
- Author
-
Alexandre A. Germanos, Sonali Arora, Ye Zheng, Erica T. Goddard, Ilsa M. Coleman, Anson T. Ku, Scott Wilkinson, Robert A. Amezquita, Michael Zager, Annalysa Long, Yu Chi Yang, Jason H. Bielas, Raphael Gottardo, Cyrus M. Ghajar, Peter S. Nelson, Adam G. Sowalsky, Manu Setty, and Andrew C. Hsieh
- Abstract
SummaryAdvanced prostate malignancies are a leading cause of cancer-related deaths in men, in large part due to our incomplete understanding of cellular drivers of disease progression. We investigated prostate cancer cell dynamics at single-cell resolution from disease onset to the development of androgen independence in vivo. We observe a dramatic expansion of a castration-resistant intermediate luminal cell type that correlates with treatment resistance and poor prognosis in human patients. Moreover, transformed epithelial cells and associated fibroblasts create a microenvironment conducive to pro-tumorigenic immune infiltration, which is in part androgen responsive. Androgen independent prostate cancer leads to significant diversification of intermediate luminal cell populations characterized by a range of androgen signaling activity inversely correlated with proliferation and mRNA translation. Accordingly, distinct epithelial populations are exquisitely sensitive to translation inhibition which leads to epithelial cell death, loss of pro-tumorigenic signaling, and decreased tumor heterogeneity. Our findings reveal a complex tumor environment largely dominated by castration-resistant luminal cells and immunosuppressive infiltrates.
- Published
- 2022
- Full Text
- View/download PDF
80. Prostate-Specific Membrane Antigen is a Biomarker for Residual Disease Following Neoadjuvant Intense Androgen Deprivation Therapy in Prostate Cancer
- Author
-
John R. Bright, Rosina T. Lis, Anson T. Ku, Nicholas T. Terrigino, Nichelle C. Whitlock, Shana Y. Trostel, Nicole V. Carrabba, Stephanie A. Harmon, Baris Turkbey, Scott Wilkinson, and Adam G. Sowalsky
- Subjects
Male ,Prostatectomy ,Neoplasm, Residual ,Urology ,Androgens ,Prostate ,Humans ,Prostatic Neoplasms ,Androgen Antagonists ,Prostate-Specific Antigen ,urologic and male genital diseases ,Article ,Neoadjuvant Therapy - Abstract
PURPOSE: Neoadjuvant intense androgen deprivation therapy (iADT) can exert a wide range of histologic responses, which in turn are reflected in the final prostatectomy specimen. Accurate identification and measurement of residual tumor volumes are critical for tracking and stratifying patient outcomes. MATERIALS AND METHODS: The goal of this current study was to evaluate the ability of antibodies against prostate-specific membrane antigen (PSMA) to specifically detect residual tumor in a cohort of 35 patients treated with iADT plus enzalutamide for six months prior to radical prostatectomy. RESULTS: Residual carcinoma was detected in 31 patients, and PSMA reacted positively with tumor in all cases. PSMA staining was 96% sensitive for tumor, with approximately 82% of benign regions showing no reactivity. By contrast, PSMA positively reacted with 72% of benign regions in a control cohort of 37 untreated cases, resulting in 28% specificity for tumor. PSMA further identified highly dedifferentiated prostate carcinomas including tumors with evidence of neuroendocrine differentiation. CONCLUSIONS: We propose that anti-PSMA immunostaining be a standardized marker for identifying residual cancer in the setting of neoadjuvant intense androgen deprivation therapy.
- Published
- 2022
81. Prostate-specific membrane antigen is a biomarker for residual disease following neoadjuvant intense androgen deprivation therapy in prostate cancer
- Author
-
Scott Wilkinson, John R. Bright, Anson Ku, Stephanie Harmon, Rosina T. Lis, Baris Turkbey, Adam G. Sowalsky, Nicole V. Carrabba, Nicholas T. Terrigino, and Shana Y. Trostel
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Prostatectomy ,medicine.medical_treatment ,urologic and male genital diseases ,medicine.disease ,Neuroendocrine differentiation ,Androgen deprivation therapy ,Prostate cancer ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Prostate ,Internal medicine ,medicine ,Carcinoma ,Enzalutamide ,Biomarker (medicine) ,business - Abstract
Neoadjuvant intense androgen deprivation therapy can exert a wide range of histologic responses, which in turn are reflected in the final prostatectomy specimen. Accurate identification and measurement of residual tumor volumes are critical for tracking and stratifying patient outcomes. The goal of this current study was to evaluate the ability of antibodies against prostate-specific membrane antigen (PSMA) to detect residual tumor in a cohort of 35 patients treated with androgen deprivation therapy plus enzalutamide for six months prior to radical prostatectomy. Residual carcinoma was detected in 31 patients, and PSMA reacted positively with tumor in all cases. PSMA staining was 95.5% sensitive for tumor, with approximately 81.6% of benign regions showing no reactivity. By contrast, PSMA positively reacted with 72.2% of benign regions in a control cohort of 37 untreated cases, resulting in 27.8% specificity for tumor. PSMA further identified highly dedifferentiated prostate carcinomas including tumors with evidence of neuroendocrine differentiation. We propose that anti-PSMA immunostaining be a standardized marker for identifying residual cancer in the setting of neoadjuvant intense androgen deprivation therapy.
- Published
- 2021
- Full Text
- View/download PDF
82. Metastatic Castration-Resistant Prostate Cancer Remains Dependent on Oncogenic Drivers Found in Primary Tumors
- Author
-
Seiji Arai, Olga Voznesensky, Adam G. Sowalsky, David J. Einstein, Carla Calagua, Fang Xie, Christina Luffman, Brian J. Capaldo, Joshua W. Russo, Steven P. Balk, and Jonathan L. Hecht
- Subjects
Male ,Cancer Research ,business.industry ,Carcinogenesis ,Biopsy ,ORIGINAL REPORTS ,Castration resistant ,medicine.disease ,Androgen receptor ,Prostate cancer ,Prostatic Neoplasms, Castration-Resistant ,Oncology ,Cancer research ,Medicine ,Humans ,Precision Medicine ,business ,Phylogeny - Abstract
PURPOSE Metastatic prostate cancer is initially sensitive to androgen receptor inhibition, but eventually becomes castration-resistant prostate cancer (mCRPC). Early use of more intensive therapies targeting androgen receptor and other oncogenic drivers in treatment-naïve primary prostate cancer (PC) may be more effective than that in advanced mCRPC. However, analysis of primary tumors may not reveal targetable metastatic drivers that are subclonal in the primary tumor or acquired at metastatic sites. METHODS PC samples spanning one patient's clinical course: diagnostic biopsies, pre- or post-enzalutamide metastatic biopsies, and rapid autopsy samples including a patient-derived xenograft (PDX) were analyzed by targeted exome sequencing followed by phylogenetic analysis. RESULTS Left- and right-lobe primary PC tumors appeared to diverge, with the right acquiring additional shared mutations and striking differences in copy number alterations that later appeared in metastatic samples during the treatment course and at autopsy, whereas the left base tumor maintained a quiet copy number alteration landscape and partitioned into a dead-end node. RB1 loss, a common finding in advanced castration-resistant disease, was identified throughout mCRPC samples, but not in the primary tumor. Significantly, a truncal EGFR-activating mutation (R108K) was identified in the primary tumor and was also found to be maintained in the mCRPC samples and in a PDX model. Furthermore, the PDX model remained sensitive to the EGFR inhibitor erlotinib, despite the presence of both RB1 and BRCA2 losses. CONCLUSION These findings indicate that truncal alterations identified in primary PC can drive advanced mCRPC, even in the presence of additional strong oncogenic drivers (ie, RB1 and BRCA2 loss), and suggest that earlier detection and targeting of these truncal alterations may be effective at halting disease progression., "Truncal EGFR (R108K) drives advanced CRPC, even in the presence of additional oncogenic drivers (RB1, BRCA2 loss)."
- Published
- 2021
83. MP33-01 METASTATIC CASTRATION-RESISTANT PROSTATE CANCER REMAINS DEPENDENT ON ONCOGENIC DRIVERS IN PRIMARY TUMORS
- Author
-
Steven P. Balk, Joshua W. Russo, Adam G. Sowalsky, Seiji Arai, Kazuhiro Suzuki, and David J. Einstein
- Subjects
Androgen receptor ,Prostate cancer ,business.industry ,Urology ,Cancer research ,medicine ,food and beverages ,Castration resistant ,medicine.disease ,business - Abstract
INTRODUCTION AND OBJECTIVE:Localized prostate cancer (PC) can be curable, but metastatic recurrence is unfortunately common. Metastatic PC is initially sensitive to androgen receptor inhibition, bu...
- Published
- 2021
- Full Text
- View/download PDF
84. Low Abundance of Circulating Tumor DNA in Localized Prostate Cancer
- Author
-
Glenn J. Bubley, David J. Einstein, Shana Y. Trostel, Nichelle C. Whitlock, Huihui Ye, James L. Gulley, Adam G. Sowalsky, Nicole V. Carrabba, Peter Chang, Amalia R. Sweet, Olga Voznesensky, Ross Lake, Scott Wilkinson, Rachel J. Schaefer, Rayann Atway, Nicholas T. Terrigino, Steven P. Balk, Steven Shema, Fatima Karzai, and S. Thomas Hennigan
- Subjects
0301 basic medicine ,Cancer Research ,Somatic cell ,Disease ,Article ,chemistry.chemical_compound ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Text mining ,Prostate ,Abundance (ecology) ,Medicine ,030304 developmental biology ,Whole genome sequencing ,0303 health sciences ,business.industry ,medicine.disease ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Oncology ,Circulating tumor DNA ,Localized disease ,030220 oncology & carcinogenesis ,Cancer research ,business ,DNA - Abstract
PURPOSE Despite decreased screening-based detection of clinically insignificant tumors, most diagnosed prostate cancers are still indolent, indicating a need for better strategies for detection of clinically significant disease before treatment. We hypothesized that patients with detectable circulating tumor DNA (ctDNA) were more likely to harbor aggressive disease. METHODS We applied ultra-low-pass whole-genome sequencing to profile cell-free DNA from 112 patients diagnosed with localized prostate cancer and performed targeted resequencing of plasma DNA for somatic mutations previously identified in matched solid tumor in nine cases. We also performed similar analyses of data from patients with metastatic prostate cancer. RESULTS In all cases of localized prostate cancer, even in clinically high-risk patients who subsequently had recurrent disease, ultra-low-pass whole-genome sequencing and targeted resequencing did not detect ctDNA in plasma acquired before surgery or before recurrence. In contrast, using both approaches, ctDNA was detected in patients with metastatic prostate cancer. CONCLUSION Our findings demonstrate clear differences between localized and advanced prostate cancer with respect to the dissemination and detectability of ctDNA. Because allele-specific alterations in ctDNA are below the threshold for detection in localized prostate cancer, other approaches to identify cell-free nucleic acids of tumor origin may demonstrate better specificity for aggressive disease.
- Published
- 2019
- Full Text
- View/download PDF
85. Integrative Genomic Analysis of Coincident Cancer Foci Implicates CTNNB1 and PTEN Alterations in Ductal Prostate Cancer
- Author
-
Adam G. Sowalsky, Justin B. Lack, Jose A. Rodriguez-Nieves, David J. VanderWeele, Gladell P. Paner, Donald J. Vander Griend, David M. Hatcher, Marc Gillard, Divya Gandla, and Andrea Pontier
- Subjects
Male ,0301 basic medicine ,Urology ,Acinar adenocarcinoma ,Article ,Neoplasms, Multiple Primary ,Transcriptome ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Exome Sequencing ,medicine ,Humans ,Tensin ,PTEN ,beta Catenin ,Exome sequencing ,Aged ,Laser capture microdissection ,Prostatectomy ,biology ,Carcinoma, Acinar Cell ,business.industry ,PTEN Phosphohydrolase ,Wnt signaling pathway ,High-Throughput Nucleotide Sequencing ,Prostatic Neoplasms ,Middle Aged ,medicine.disease ,Carcinoma, Ductal ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,business - Abstract
Background Ductal adenocarcinoma of the prostate is an aggressive subtype, with high rates of biochemical recurrence and overall poor prognosis. It is frequently found coincident with conventional acinar adenocarcinoma. The genomic features driving evolution to its ductal histology and the biology associated with its poor prognosis remain unknown. Objective To characterize genomic features distinguishing ductal adenocarcinoma from coincident acinar adenocarcinoma foci from the same patient. Design, setting, and participants Ten patients with coincident acinar and ductal prostate cancer underwent prostatectomy. Laser microdissection was used to separately isolate acinar and ductal foci. DNA and RNA were extracted, and used for integrative genomic and transcriptomic analyses. Outcome measurements and statistical analysis Single nucleotide mutations, small indels, copy number estimates, and expression profiles were identified. Phylogenetic relationships between coincident foci were determined, and characteristics distinguishing ductal from acinar foci were identified. Results and limitations Exome sequencing, copy number estimates, and fusion genes demonstrated coincident ductal and acinar adenocarcinoma diverged from a common progenitor, yet they harbored distinct alterations unique to each focus. AR expression and activity were similar in both histologies. Nine of 10 cases had mutually exclusive CTNNB1 hotspot mutations or phosphatase and tensin homolog ( PTEN ) alterations in the ductal component, and these were absent in the acinar foci. These alterations were associated with changes in expression in WNT- and PI3K-pathway genes. Conclusions Coincident ductal and acinar histologies typically are clonally related and thus arise from the same cell of origin. Ductal foci are enriched for cases with either a CTNNB1 hotspot mutation or a PTEN alteration, and are associated with WNT- or PI3K-pathway activation. These alterations are mutually exclusive and may represent distinct subtypes. Patient summary The aggressive subtype ductal adenocarcinoma is closely related to conventional acinar prostate cancer. Ductal foci contain additional alterations, however, leading to frequent activation of two targetable pathways.
- Published
- 2019
- Full Text
- View/download PDF
86. Autocrine Canonical Wnt Signaling Primes Noncanonical Signaling through ROR1 in Metastatic Castration-Resistant Prostate Cancer
- Author
-
Fen Ma, Seiji Arai, Keshan Wang, Carla Calagua, Amanda R. Yuan, Larysa Poluben, Zhongkai Gu, Joshua W. Russo, David J. Einstein, Huihui Ye, Meng Xiao He, Yu Liu, Eliezer Van Allen, Adam G. Sowalsky, Manoj K. Bhasin, Xin Yuan, and Steven P. Balk
- Subjects
Male ,Cancer Research ,Autocrine Communication ,Prostatic Neoplasms, Castration-Resistant ,Oncology ,Humans ,Receptor Tyrosine Kinase-like Orphan Receptors ,Wnt Signaling Pathway ,beta Catenin ,Article - Abstract
Wnt signaling driven by genomic alterations in genes including APC and CTNNB, which encodes β-catenin, have been implicated in prostate cancer development and progression to metastatic castration-resistant prostate cancer (mCRPC). However, nongenomic drivers and downstream effectors of Wnt signaling in prostate cancer and the therapeutic potential of targeting this pathway in prostate cancer have not been fully established. Here we analyzed Wnt/β-catenin signaling in prostate cancer and identified effectors distinct from those found in other tissues, including aryl hydrocarbon receptor and RUNX1, which are linked to stem cell maintenance, and ROR1, a noncanonical Wnt5a coreceptor. Wnt/β-catenin signaling–mediated increases in ROR1 enhanced noncanonical responses to Wnt5a. Regarding upstream drivers, APC genomic loss, but not its epigenetic downregulation commonly observed in prostate cancer, was strongly associated with Wnt/β-catenin pathway activation in clinical samples. Tumor cell upregulation of the Wnt transporter Wntless (WLS) was strongly associated with Wnt/β-catenin pathway activity in primary prostate cancer but also associated with both canonical and noncanonical Wnt signaling in mCRPC. IHC confirmed tumor cell WLS expression in primary prostate cancer and mCRPC, and patient-derived prostate cancer xenografts expressing WLS were responsive to treatment with Wnt synthesis inhibitor ETC-1922159. These findings reveal that Wnt/β-catenin signaling in prostate cancer drives stem cell maintenance and invasion and primes for noncanonical Wnt signaling through ROR1. They further show that autocrine Wnt production is a nongenomic driver of canonical and noncanonical Wnt signaling in prostate cancer, which can be targeted with Wnt synthesis inhibitors to suppress tumor growth. Significance: This work provides fundamental insights into Wnt signaling and prostate cancer cell biology and indicates that a subset of prostate cancer driven by autocrine Wnt signaling is sensitive to Wnt synthesis inhibitors.
- Published
- 2021
87. Quantitative and Qualitative Analysis of Blood-based Liquid Biopsies to Inform Clinical Decision-making in Prostate Cancer
- Author
-
Marzia Del Re, David T. Miyamoto, Kenneth J. Pienta, Alejandro Athie, Arnulf Stenzl, Adam G. Sowalsky, Alexander W. Wyatt, Paul C. Boutros, Irene Casanova-Salas, Edwin M. Posadas, Joaquin Mateo, Institut Català de la Salut, [Casanova-Salas I, Athie A, Mateo J] Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Vall d’Hebron Hospital Universitari, Barcelona, Spain. [Boutros PC] Departments of Human Genetics and Urology, Institute for Precision Health and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA. [Del Re M] Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy. [Miyamoto DT] Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA. [Pienta KJ] The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Oncology ,Male ,diagnóstico::toma de decisiones clínicas [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,030232 urology & nephrology ,Neoplastic Cells ,Circulating Tumor DNA ,Transcriptome ,Prostate cancer ,0302 clinical medicine ,Circulating tumor cell ,Diagnosis::Diagnostic Techniques and Procedures::Clinical Laboratory Techniques::Cytological Techniques::Cytodiagnosis::Biopsy::Liquid Biopsy [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Circulating ,Cancer ,screening and diagnosis ,Tumor ,Otros calificadores::Otros calificadores::/genética [Otros calificadores] ,Precision medicine ,Genomics ,Extracellular vesicles ,Urology & Nephrology ,Neoplastic Cells, Circulating ,diagnóstico::técnicas y procedimientos diagnósticos::técnicas de laboratorio clínico::técnicas citológicas::citodiagnóstico::biopsia::biopsia líquida [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Detection ,030220 oncology & carcinogenesis ,Diagnosis::Clinical Decision-Making [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Pròstata - Càncer - Aspectes genètics ,Biòpsia ,Pròstata - Càncer - Presa de decisions ,Biotechnology ,4.2 Evaluation of markers and technologies ,Urologic Diseases ,medicine.medical_specialty ,Neoplasms::Neoplasms by Site::Urogenital Neoplasms::Genital Neoplasms, Male::Prostatic Neoplasms [DISEASES] ,Urology ,Clinical Decision-Making ,Clinical Sciences ,Context (language use) ,03 medical and health sciences ,Clinical Research ,Internal medicine ,Biomarkers, Tumor ,Other subheadings::Other subheadings::/genetics [Other subheadings] ,medicine ,Genetics ,Humans ,Liquid biopsy ,Circulating tumor DNA ,business.industry ,Human Genome ,Prostatic Neoplasms ,Epigenome ,medicine.disease ,4.1 Discovery and preclinical testing of markers and technologies ,neoplasias::neoplasias por localización::neoplasias urogenitales::neoplasias de los genitales masculinos::neoplasias de la próstata [ENFERMEDADES] ,Good Health and Well Being ,Liquid Biopsy ,Tumor progression ,business ,Biomarkers - Abstract
Context Genomic stratification can impact prostate cancer (PC) care through diagnostic, prognostic, and predictive biomarkers that aid in clinical decision-making. The temporal and spatial genomic heterogeneity of PC together with the challenges of acquiring metastatic tissue biopsies hinder implementation of tissue-based molecular profiling in routine clinical practice. Blood-based liquid biopsies are an attractive, minimally invasive alternative. Objective To review the clinical value of blood-based liquid biopsy assays in PC and identify potential applications to accelerate the development of precision medicine. Evidence acquisition A systematic review of PubMed/MEDLINE was performed to identify relevant literature on blood-based circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and extracellular vesicles (EVs) in PC. Evidence synthesis Liquid biopsy has emerged as a practical tool to profile tumor dynamics over time, elucidating features that evolve (genome, epigenome, transcriptome, and proteome) with tumor progression. Liquid biopsy tests encompass analysis of DNA, RNA, and proteins that can be detected in CTCs, ctDNA, or EVs. Blood-based liquid biopsies have demonstrated promise in the context of localized tumors (diagnostic signatures, risk stratification, and disease monitoring) and advanced disease (response/resistance biomarkers and prognostic markers). Conclusions Liquid biopsies have value as a source of prognostic, predictive, and response biomarkers in PC. Most clinical applications have been developed in the advanced metastatic setting, where CTC and ctDNA yields are significantly higher. However, standardization of assays and analytical/clinical validation is necessary prior to clinical implementation. Patient summary Traces of tumors can be isolated from blood samples from patients with prostate cancer either as whole cells or as DNA fragments. These traces provide information on tumor features. These minimally invasive tests can guide diagnosis and treatment selection.
- Published
- 2021
88. A Subset of Localized Prostate Cancer Displays an Immunogenic Phenotype Associated with Losses of Key Tumor Suppressor Genes
- Author
-
Sabina Signoretti, Steven P. Balk, Carla Calagua, Adam G. Sowalsky, Ross Lake, Haydn T. Kissick, David B. Sykes, Olga Voznesensky, Huihui Ye, Scott Wilkinson, Caroline S. Jansen, Taghreed Hirz, David J. Einstein, Anson T. Ku, Miriam Ficial, Philip J. Saylor, and Luke del Balzo
- Subjects
Prostate cancer ,Immune system ,medicine.anatomical_structure ,T cell ,Cancer research ,medicine ,Cytotoxic T cell ,Dendritic cell ,Progenitor cell ,Biology ,medicine.disease ,CD8 ,Immune checkpoint - Abstract
PurposeA subset of primary prostate cancer (PCa) expresses programmed death-ligand 1 (PD-L1), but whether they have unique tumor immune microenvironment (TIME) or genomic features is unclear.Experimental DesignWe selected PD-L1-positive high-grade and/or high-risk primary PCa, characterized tumor-infiltrating lymphocytes (TILS) with multiplex immunofluorescence, and identified genomic alterations in immunogenic and non-immunogenic tumor foci.ResultsOne-quarter of aggressive localized PCa cases (29/115) had tumor PD-L1 expression >5%. This correlated with increased density of CD8+ T cells, a large fraction co-expressing PD-1, versus absent PD-1 expression on sparse CD8 T cells in unselected cases. Most CD8+PD-1+ cells did not express terminal exhaustion markers (TIM-3 or LAG-3), while a subset expressed TCF1. Consistent with these CD8+PD-1+TCF1+ cells being progenitors, they were found in antigen-presenting-cell niches in close proximity to MHC II+ cells. CD8 T cell density in immunogenic PCa and renal cell carcinoma (RCC) was nearly identical. Shallow RB1 and BRCA2 losses, and deep deletions of CHD1, were prevalent; the latter being strongly associated with a dendritic cell gene set in TCGA. Tumor mutation burden was variable; neither high microsatellite instability nor CDK12 alterations were present.ConclusionsA subset of localized PCa is immunogenic, manifested by PD-L1 expression and CD8+ T cell content comparable to RCC. The CD8+ T cells include effector cells and exhausted progenitor cells, which may be expanded by ICIs. Genomic losses of RB1, BRCA2, and CHD1 may be drivers of this phenotype. These findings indicate that immunotherapies may be effective in biomarker-selected subpopulations of localized PCa patients.Statement of Translational RelevanceProstate cancer (PCa) is generally considered poorly immunogenic, with low expression of programmed death-ligand 1 (PD-L1) and low density of tumor-infiltrating immune cells. Accordingly, response rates to PD(L)-1 inhibition in unselected patients with advanced prostate cancer have been low. Here, we find that a substantial subset of aggressive primary PCa exhibits tumor PD-L1 expression and contains a high density of tumor-infiltrating lymphocytes. These lymphocytes contain sub-populations of exhausted progenitor CD8+ T cells and differentiated effector T cells, the hallmarks of ongoing anti-tumor immune response and a prerequisite for response to checkpoint inhibition. Furthermore, we identify genomic alterations that may be contributing to immunogenicity in these cases. These findings point to immune responses elicited in a subset of primary PCa, supporting the development of immune checkpoint blockade clinical trials in early-stage disease, such as biochemically recurrent PCa, that are driven by genomic features of the tumor or the immune microenvironment.
- Published
- 2021
- Full Text
- View/download PDF
89. A Subset of Localized Prostate Cancer Displays an Immunogenic Phenotype Associated with Losses of Key Tumor Suppressor Genes
- Author
-
Caroline S. Jansen, Miriam Ficial, Ross Lake, Scott Wilkinson, Olga Voznesensky, Luke del Balzo, Adam G. Sowalsky, Philip J. Saylor, Steven P. Balk, Carla Calagua, David J. Einstein, Taghreed Hirz, David B. Sykes, Huihui Ye, Haydn T. Kissick, Anson T. Ku, and Sabina Signoretti
- Subjects
Male ,Cancer Research ,Mutation ,LAG3 ,Cell ,Prostatic Neoplasms ,Dendritic cell ,Biology ,CD8-Positive T-Lymphocytes ,medicine.disease ,medicine.disease_cause ,B7-H1 Antigen ,Prostate cancer ,medicine.anatomical_structure ,Lymphocytes, Tumor-Infiltrating ,Phenotype ,Oncology ,medicine ,Cancer research ,Tumor Microenvironment ,Cytotoxic T cell ,Humans ,Genes, Tumor Suppressor ,Progenitor cell ,CD8 - Abstract
Purpose: A subset of primary prostate cancer expresses programmed death-ligand 1 (PD-L1), but whether they have a unique tumor immune microenvironment or genomic features is unclear. Experimental Design: We selected PD-L1–positive high-grade and/or high-risk primary prostate cancer, characterized tumor-infiltrating lymphocytes with multiplex immunofluorescence, and identified genomic alterations in immunogenic and nonimmunogenic tumor foci. Results: One quarter of aggressive localized prostate cancer cases (29/115) had tumor PD-L1 expression more than 5%. This correlated with increased density of CD8+ T cells, a large fraction coexpressing PD-1, versus absent PD-1 expression on sparse CD8 T cells in unselected cases. Most CD8+PD-1+ cells did not express terminal exhaustion markers (TIM3 or LAG3), while a subset expressed TCF1. Consistent with these CD8+PD-1+TCF1+ cells being progenitors, they were found in antigen-presenting cell niches in close proximity to MHC-II+ cells. CD8 T-cell density in immunogenic prostate cancer and renal cell carcinoma (RCC) was nearly identical. Shallow RB1 and BRCA2 losses, and deep deletions of CHD1, were prevalent, the latter being strongly associated with a dendritic cell gene set in The Cancer Genome Atlas. Tumor mutation burden was variable; neither high microsatellite instability nor CDK12 alterations were present. Conclusions: A subset of localized prostate cancer is immunogenic, manifested by PD-L1 expression and CD8+ T-cell content comparable with RCC. The CD8+ T cells include effector cells and exhausted progenitor cells, which may be expanded by immune checkpoint inhibitors (ICI). Genomic losses of RB1, BRCA2, and CHD1 may be drivers of this phenotype. These findings indicate that immunotherapies may be effective in biomarker-selected subpopulations of patients with localized prostate cancer.
- Published
- 2021
90. Reply to Alessia Cimadamore, Liang Cheng, Antonio Lopez-Beltran, Marina Scarpelli, and Rodolfo Montironi’s Letter to the Editor re: Scott Wilkinson, Huihui Ye, Fatima Karzai, et al. Nascent Prostate Cancer Heterogeneity Drives Evolution and Resistance to Intense Hormonal Therapy. Eur Urol. In press. https://doi.org/10.1016/j.eururo.2021.03.009: Focus on Intraductal Carcinoma of the Prostate
- Author
-
Scott Wilkinson, Huihui Ye, Rosina T. Lis, and Adam G. Sowalsky
- Subjects
Urology - Published
- 2021
- Full Text
- View/download PDF
91. Alterations in AR regulome following response to kinase inhibitors
- Author
-
Kathleen Kelly, David J VanderWeele, Adam G. Sowalsky, Berkley E. Gryder, Brian J. Capaldo, and Remi Adelaiye-Ogala
- Published
- 2020
- Full Text
- View/download PDF
92. Nascent Prostate Cancer Heterogeneity Drives Evolution and Resistance to Intense Hormonal Therapy
- Author
-
Harsimar B. Kaur, John R. Bright, Houssein Abdul Sater, Nichelle C. Whitlock, James L. Gulley, Adam G. Sowalsky, Rayann Atway, David Venzon, Daniela C. Salles, Rosina T. Lis, Baris Turkbey, Nicholas T. Terrigino, Stephanie M. Walker, Fatima Karzai, Peter A. Pinto, Peter L. Choyke, Shana Y. Trostel, David J. VanderWeele, Scott Wilkinson, Stephanie Harmon, Nicole V. Carrabba, William L. Dahut, Maria J. Merino, Ravi A. Madan, Guinevere Chun, Tamara L. Lotan, Brian J. Capaldo, and Huihui Ye
- Subjects
Oncology ,Male ,medicine.medical_specialty ,Urology ,030232 urology & nephrology ,Article ,Androgen deprivation therapy ,03 medical and health sciences ,Prostate cancer ,chemistry.chemical_compound ,0302 clinical medicine ,Prostate ,Internal medicine ,Biopsy ,medicine ,Carcinoma ,Enzalutamide ,Humans ,Prospective Studies ,Phylogeny ,Retrospective Studies ,Phylogenetic tree ,medicine.diagnostic_test ,business.industry ,Prostatic Neoplasms ,Androgen Antagonists ,medicine.disease ,Minimal residual disease ,Clinical trial ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Cohort ,Androgens ,Immunohistochemistry ,Hormonal therapy ,business - Abstract
Background Patients diagnosed with high risk localized prostate cancer have variable outcomes following surgery. Trials of intense neoadjuvant androgen deprivation therapy (NADT) have shown lower rates of recurrence among patients with minimal residual disease after treatment. The molecular features that distinguish exceptional responders from poor responders are not known. Objective To identify genomic and histologic features associated with treatment resistance at baseline. Design, setting, and participants Targeted biopsies were obtained from 37 men with intermediate- to high-risk prostate cancer before receiving 6 mo of ADT plus enzalutamide. Biopsy tissues were used for whole-exome sequencing and immunohistochemistry (IHC). Outcome measurements and statistical analysis We assessed the relationship of molecular features with final pathologic response using a cutpoint of 0.05 cm3 for residual cancer burden to compare exceptional responders to incomplete and nonresponders. We assessed intratumoral heterogeneity at the tissue and genomic level, and compared the volume of residual disease to the Shannon diversity index for each tumor. We generated multivariate models of resistance based on three molecular features and one histologic feature, with and without multiparametric magnetic resonance imaging estimates of baseline tumor volume. Results and limitations Loss of chromosome 10q (containing PTEN) and alterations to TP53 were predictive of poor response, as were the expression of nuclear ERG on IHC and the presence of intraductal carcinoma of the prostate. Patients with incompletely and nonresponding tumors harbored greater tumor diversity as estimated via phylogenetic tree reconstruction from DNA sequencing and analysis of IHC staining. Our four-factor binary model (area under the receiver operating characteristic curve [AUC] 0.89) to predict poor response correlated with greater diversity in our cohort and a validation cohort of 57 Gleason score 8–10 prostate cancers from The Cancer Genome Atlas. When baseline tumor volume was added to the model, it distinguished poor response to NADT with an AUC of 0.98. Prospective use of this model requires further retrospective validation with biopsies from additional trials. Conclusions A subset of prostate cancers exhibit greater histologic and genomic diversity at the time of diagnosis, and these localized tumors have greater fitness to resist therapy. Patient summary Some prostate cancer tumors do not respond well to a hormonal treatment called androgen deprivation therapy (ADT). We used tumor volume and four other parameters to develop a model to identify tumors that will not respond well to ADT. Treatments other than ADT should be considered for these patients.
- Published
- 2020
93. Age and Tumor Differentiation-Associated Gene Expression Based Analysis of Non-Familial Prostate Cancers
- Author
-
Shashwat Sharad, Travis C. Allemang, Hua Li, Darryl Nousome, Anson Tai Ku, Nichelle C. Whitlock, Adam G. Sowalsky, Jennifer Cullen, Isabell A. Sesterhenn, David G. McLeod, Shiv Srivastava, and Albert Dobi
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Cancer Research ,Microarray ,Somatic cell ,laser captured microdissection ,age-associated gene expression ,Disease ,lcsh:RC254-282 ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Prostate ,Internal medicine ,Gene expression ,medicine ,Gene ,tumor-associate gene ,Original Research ,Tumor differentiation ,business.industry ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,prostate cancer ,030104 developmental biology ,medicine.anatomical_structure ,tumor differentiation ,030220 oncology & carcinogenesis ,business ,microarray - Abstract
Prostate cancer incidence in young men has increased. Patients diagnosed at an earlier age are likely to have aggressive prostate cancer and treatment decisions are continuing to be weighted by patient age and life expectancy. Identification of age-associated gene-expression signatures hold great potential to augment current and future treatment modalities. To investigate age-specific tumor associated gene signatures and their potential biomarkers for disease aggressiveness, this study was designed and stratified into well and poorly differentiated tumor types of young (42–58 years) and old (66–73 years) prostate cancer patients. The differentially expressed genes related to tumor-normal differences between non-familial prostate cancer patients were identified and several genes uniquely associated with the age and tumor differentiation are markedly polarized. Overexpressed genes known to be associated with somatic genomic alterations was predominantly found in young men, such as TMPRESS2-ERG and c-MYC. On the other hand, old men have mostly down-regulated gene expressions indicating the loss of protective genes and reduced cell mediated immunity indicated by decreased HLA-A and HLA-B expression. The normalization for the benign signatures between the age groups indicates a significant age and tumor dependent heterogeneity exists among the patients with a great potential for age-specific and tumor differentiation-based therapeutic stratification of prostate cancer.
- Published
- 2020
94. A case report of multiple primary prostate tumors with differential drug sensitivity
- Author
-
Guinevere Chun, Maria Merino, Rayann Atway, Ravi A. Madan, James L. Gulley, Nicole V. Carrabba, Nicholas T. Terrigino, Adam G. Sowalsky, Rosina T. Lis, Ross Lake, Fatima Karzai, David J. VanderWeele, Shana Y. Trostel, Scott Wilkinson, Peter A. Pinto, Stephanie Harmon, John R. Bright, William L. Dahut, Baris Turkbey, Peter L. Choyke, and Huihui Ye
- Subjects
Male ,0301 basic medicine ,Oncology ,medicine.medical_treatment ,General Physics and Astronomy ,Drug resistance ,Neoplasms, Multiple Primary ,Androgen deprivation therapy ,Prostate cancer ,0302 clinical medicine ,Prostate ,Cancer genomics ,Medicine ,Neoplasm ,lcsh:Science ,Neoadjuvant therapy ,Multidisciplinary ,biology ,Neoadjuvant Therapy ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,medicine.medical_specialty ,Tumour heterogeneity ,Science ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Internal medicine ,Humans ,PTEN ,neoplasms ,Aged ,business.industry ,PTEN Phosphohydrolase ,Prostatic Neoplasms ,Diagnostic markers ,Androgen Antagonists ,Sequence Analysis, DNA ,General Chemistry ,medicine.disease ,030104 developmental biology ,Drug Resistance, Neoplasm ,Mutation ,biology.protein ,lcsh:Q ,Cancer imaging ,Neoplasm Grading ,Tumor Suppressor Protein p53 ,business ,Gene Deletion - Abstract
Localized prostate cancers are genetically variable and frequently multifocal, comprising spatially distinct regions with multiple independently-evolving clones. To date there is no understanding of whether this variability can influence management decisions for patients with prostate tumors. Here, we present a single case from a clinical trial of neoadjuvant intense androgen deprivation therapy. A patient was diagnosed with a large semi-contiguous tumor by imaging, histologically composed of a large Gleason score 9 tumor with an adjacent Gleason score 7 nodule. DNA sequencing demonstrates these are two independent tumors, as only the Gleason 9 tumor harbors single-copy losses of PTEN and TP53. The PTEN/TP53-deficient tumor demonstrates treatment resistance, selecting for subclones with mutations to the remaining copies of PTEN and TP53, while the Gleason 7 PTEN-intact tumor is almost entirely ablated. These findings indicate that spatiogenetic variability is a major confounder for personalized treatment of patients with prostate cancer., Prostate cancer is often a multifocal disease but how best to manage this clinically remains unclear. Here, the authors report a single case study of a patient with two genetically diverse tumours which showed differential response to therapy.
- Published
- 2020
- Full Text
- View/download PDF
95. The source within - intratumoural stem-like T cells give rise to differentiated T cells
- Author
-
Donald J. McGuire, Rajesh M. Valanparambil, Se Jin Im, Mehrdad Alemozaffar, Christopher P. Filson, Caroline S. Jansen, Rama Akondy, Yun Min Chang, Ross Lake, Yuan Liu, Maria Cardenas, Kevin Richard Melnick, Adam G. Sowalsky, Haydn T. Kissick, Patrick Mullane, Jennifer W Carlisle, Nataliya Prokhnevska, Carla LaShannon Ellis, Martin G. Sanda, Scott Wilkinson, Adeboye O. Osunkoya, Amir Ishaq Khan, Viraj A. Master, Alice Kim, Mehmet Asim Bilen, Alice O. Kamphorst, Adriana Moon Reyes, William H. Hudson, and Kyu Seo Kim
- Subjects
0301 basic medicine ,Transcription, Genetic ,Cell division ,General Mathematics ,Cellular differentiation ,Population ,Cell ,Biology ,CD8-Positive T-Lymphocytes ,Article ,Epigenesis, Genetic ,Mice ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Neoplasms ,medicine ,Animals ,Cytotoxic T cell ,Humans ,In patient ,Hepatocyte Nuclear Factor 1-alpha ,Stem Cell Niche ,education ,Antigen Presentation ,education.field_of_study ,Multidisciplinary ,Applied Mathematics ,Stem Cells ,Cancer ,Cell Differentiation ,medicine.disease ,Immunosurveillance ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Disease Progression ,Cancer research ,Tumor Escape ,Stem cell ,Infiltration (medical) ,CD8 - Abstract
Tumour-infiltrating lymphocytes are associated with a survival benefit in several tumour types and with the response to immunotherapy1–8. However, the reason some tumours have high CD8 T cell infiltration while others do not remains unclear. Here we investigate the requirements for maintaining a CD8 T cell response against human cancer. We find that CD8 T cells within tumours consist of distinct populations of terminally differentiated and stem-like cells. On proliferation, stem-like CD8 T cells give rise to more terminally differentiated, effector-molecule-expressing daughter cells. For many T cells to infiltrate the tumour, it is critical that this effector differentiation process occur. In addition, we show that these stem-like T cells reside in dense antigen-presenting-cell niches within the tumour, and that tumours that fail to form these structures are not extensively infiltrated by T cells. Patients with progressive disease lack these immune niches, suggesting that niche breakdown may be a key mechanism of immune escape. The authors examine the immune cell infiltrates of human tumours and provide evidence for a population of CD8 T cells with stem-cell characteristics and proliferative capacity that reside in an antigen-presenting niche within tumours.
- Published
- 2020
96. Doubling down on tumor suppressor deletion
- Author
-
Adam G. Sowalsky
- Subjects
0301 basic medicine ,business.industry ,General Medicine ,medicine.disease ,law.invention ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,law ,030220 oncology & carcinogenesis ,PARP inhibitor ,Cancer research ,medicine ,Suppressor ,business - Abstract
Codeletion of BRCA2 and RB1 increases prostate cancer sensitivity to PARP inhibitor therapy.
- Published
- 2020
- Full Text
- View/download PDF
97. A comparison of prostate cancer bone metastases on 18F-Sodium Fluoride and Prostate Specific Membrane Antigen (18F-PSMA) PET/CT: Discordant uptake in the same lesion
- Author
-
Stephen Adler, Yolanda McKinney, Joanna H. Shih, Esther Mena, Anna Couvillon, Liza Lindenberg, Baris Turkbey, Martin G. Pomper, Peter L. Choyke, William L. Dahut, Ravi A. Madan, Ronnie C. Mease, Ethan Bergvall, James L. Gulley, Adam G. Sowalsky, Sherif Mehralivand, Janet F. Eary, and Stephanie Harmon
- Subjects
Oncology ,medicine.medical_specialty ,Genitourinary system ,business.industry ,Concordance ,Cancer ,medicine.disease ,030218 nuclear medicine & medical imaging ,3. Good health ,Bone remodeling ,Lesion ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Glutamate carboxypeptidase II ,medicine ,medicine.symptom ,business ,Cancer Imaging Program - Abstract
// Stephanie A. Harmon 1, 2 , Esther Mena 2 , Joanna H. Shih 3 , Stephen Adler 1, 2 , Yolanda McKinney 2 , Ethan Bergvall 2 , Sherif Mehralivand 2 , Adam G. Sowalsky 4 , Anna Couvillon 5 , Ravi A. Madan 5 , James L. Gulley 5 , Janet Eary 6 , Ronnie C. Mease 7 , Martin G. Pomper 7 , William L. Dahut 5 , Baris Turkbey 2 , Liza Lindenberg 2 and Peter L. Choyke 2 1 Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA 2 Molecular Imaging Program, National Cancer Institute, NIH, Bethesda, MD, USA 3 Biometric Research Branch, National Cancer Institute, NIH, Bethesda, MD, USA 4 Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA 5 Genitourinary Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD, USA 6 Cancer Imaging Program, National Cancer Institute, NIH, Rockville, MD, USA 7 Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA Correspondence to: Stephanie A. Harmon, email: stephanie.harmon@nih.gov Keywords: prostate cancer; bone metastases; sodium fluoride; prostate specific membrane antigen; spatial analysis Received: October 01, 2018 Accepted: December 04, 2018 Published: December 28, 2018 ABSTRACT Purpose: Prostate-Specific Membrane Antigen (PSMA) PET/CT has been introduced as a sensitive method for characterizing metastatic prostate cancer. The purpose of this study is to compare the spatial concordance of 18 F-NaF PET/CT and 18 F-PSMA-targeted PET/CT within prostate cancer bone metastases. Methods: Prostate cancer patients with known bone metastases underwent PSMA-targeted PET/CT ( 18 F-DCFBC or 18 F-DCFPyL) and 18 F-NaF PET/CT. In pelvic and spinal lesions detected by both radiotracers, regions-of-interest (ROIs) derived by various thresholds of uptake intensity were compared for spatial colocalization. Overlap volume was correlated with uptake characteristics and disease status. Results: The study included 149 lesions in 19 patients. Qualitatively, lesions exhibited a heterogeneous range of spatial concordance between PSMA and NaF uptake from completely matched to completely discordant. Quantitatively, overlap volume decreased as a function of tracer intensity. and disease status, where lesions from patients with castration-sensitive disease showed higher spatial concordance while lesions from patients with castration-resistant disease demonstrated more frequent spatial discordance. Conclusion: As metastatic prostate cancer progresses from castration-sensitive to castration-resistant, greater discordance is observed between NaF PET and PSMA PET uptake. This may indicate a possible phenotypic shift to tumor growth that is more independent of bone remodeling via osteoblastic formation.
- Published
- 2018
- Full Text
- View/download PDF
98. BMX-Mediated Regulation of Multiple Tyrosine Kinases Contributes to Castration Resistance in Prostate Cancer
- Author
-
Huihui Ye, Xin Yuan, Changmeng Cai, Sen Chen, Steven P. Balk, Nathanael S. Gray, Adam G. Sowalsky, Nicholas I. Simon, and Fen Ma
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Amino Acid Motifs ,Mice, SCID ,Biology ,urologic and male genital diseases ,Antibodies ,Gene Expression Regulation, Enzymologic ,Article ,Receptor tyrosine kinase ,Mice ,03 medical and health sciences ,Piperidines ,Cell Line, Tumor ,Animals ,Humans ,Bruton's tyrosine kinase ,Phosphorylation ,Kinase activity ,Mice, Inbred ICR ,Sequence Analysis, RNA ,Kinase ,Adenine ,Androgen Antagonists ,Protein-Tyrosine Kinases ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,Prostatic Neoplasms, Castration-Resistant ,HEK293 Cells ,Pyrimidines ,030104 developmental biology ,Oncology ,Receptors, Androgen ,Tissue Array Analysis ,Androgens ,biology.protein ,Cancer research ,Pyrazoles ,Signal transduction ,Tyrosine kinase ,Neoplasm Transplantation ,Protein Binding ,Signal Transduction - Abstract
Prostate cancer responds to therapies that suppress androgen receptor (AR) activity (androgen deprivation therapy, ADT) but invariably progresses to castration-resistant prostate cancer (CRPC). The Tec family nonreceptor tyrosine kinase BMX is activated downstream of PI3K and has been implicated in regulation of multiple pathways and in the development of cancers including prostate cancer. However, its precise mechanisms of action, and particularly its endogenous substrates, remain to be established. Here, we demonstrate that BMX expression in prostate cancer is suppressed directly by AR via binding to the BMX gene and that BMX expression is subsequently rapidly increased in response to ADT. BMX contributed to CRPC development in cell line and xenograft models by positively regulating the activities of multiple receptor tyrosine kinases through phosphorylation of a phosphotyrosine-tyrosine (pYY) motif in their activation loop, generating pYpY that is required for full kinase activity. To assess BMX activity in vivo, we generated a BMX substrate–specific antibody (anti-pYpY) and found that its reactivity correlated with BMX expression in clinical samples, supporting pYY as an in vivo substrate. Inhibition of BMX with ibrutinib (developed as an inhibitor of the related Tec kinase BTK) or another BMX inhibitor BMX-IN-1 markedly enhanced the response to castration in a prostate cancer xenograft model. These data indicate that increased BMX in response to ADT contributes to enhanced tyrosine kinase signaling and the subsequent emergence of CRPC, and that combination therapies targeting AR and BMX may be effective in a subset of patients. Significance: The tyrosine kinase BMX is negatively regulated by androgen and contributes to castration-resistant prostate cancer by enhancing the phosphorylation and activation of multiple receptor tyrosine kinases following ADT. Cancer Res; 78(18); 5203–15. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
99. A PDX/Organoid Biobank of Advanced Prostate Cancers Captures Genomic and Phenotypic Heterogeneity for Disease Modeling and Therapeutic Screening
- Author
-
Juan Juan Yin, Fatima Karzai, Supreet Agarwal, Michael L. Beshiri, Kathleen Kelly, Caitlin M. Tice, Eva Corey, Kerry McGowen, Keith H. Jansson, Crystal Tran, Adam G. Sowalsky, Qi Yang, Holly M. Nguyen, William L. Dahut, and Aian Neil Alilin
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Genotype ,Biology ,Article ,Olaparib ,Genetic Heterogeneity ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,Organoid ,medicine ,Animals ,Humans ,Cell Lineage ,Biological Specimen Banks ,Genetic heterogeneity ,Lineage markers ,Prostatic Neoplasms ,Cancer ,medicine.disease ,Organoids ,030104 developmental biology ,Oncology ,chemistry ,PARP inhibitor ,Cancer research ,Heterografts ,Adenocarcinoma - Abstract
Purpose: Prostate cancer translational research has been hampered by the lack of comprehensive and tractable models that represent the genomic landscape of clinical disease. Metastatic castrate-resistant prostate cancer (mCRPC) patient-derived xenografts (PDXs) recapitulate the genetic and phenotypic diversity of the disease. We sought to establish a representative, preclinical platform of PDX-derived organoids that is experimentally facile for high-throughput and mechanistic analysis. Experimental Design: Using 20 models from the LuCaP mCRPC PDX cohort, including adenocarcinoma and neuroendocrine lineages, we systematically tested >20 modifications to prostate organoid conditions. Organoids were evaluated for genomic and phenotypic stability and continued reliance on the AR signaling pathway. The utility of the platform as a genotype-dependent model of drug sensitivity was tested with olaparib and carboplatin. Results: All PDX models proliferated as organoids in culture. Greater than 50% could be continuously cultured long-term in modified conditions; however, none of the PDXs could be established long-term as organoids under previously reported conditions. In addition, the modified conditions improved the establishment of patient biopsies over current methods. The genomic heterogeneity of the PDXs was conserved in organoids. Lineage markers and transcriptomes were maintained between PDXs and organoids. Dependence on AR signaling was preserved in adenocarcinoma organoids, replicating a dominant characteristic of CRPC. Finally, we observed maximum cytotoxicity to the PARP inhibitor olaparib in BRCA2−/− organoids, similar to responses observed in patients. Conclusions: The LuCaP PDX/organoid models provide an expansive, genetically characterized platform to investigate the mechanisms of pathogenesis as well as therapeutic responses and their molecular correlates in mCRPC. Clin Cancer Res; 24(17); 4332–45. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
100. Neoadjuvant-Intensive Androgen Deprivation Therapy Selects for Prostate Tumor Foci with Diverse Subclonal Oncogenic Alterations
- Author
-
Manoj Bhasin, Zhenwei Zhang, Elahe A. Mostaghel, Adam G. Sowalsky, Glenn J. Bubley, Rosina T. Lis, Fen Ma, Olga Voznesensky, Eliezer M. Van Allen, Laleh Montaser-Kouhsari, Steven P. Balk, Carla Calagua, Huihui Ye, Rachel J. Schaefer, Joshua W. Russo, Bruce Montgomery, Massimo Loda, Peter S. Nelson, and Mary-Ellen Taplin
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Carcinogenesis ,medicine.medical_treatment ,Abiraterone Acetate ,Neuroendocrine differentiation ,Article ,Clonal Evolution ,Androgen deprivation therapy ,03 medical and health sciences ,Prostate cancer ,chemistry.chemical_compound ,Prostate ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,Humans ,Medicine ,Cytochrome P450 Family 2 ,Neoadjuvant therapy ,Aged ,Prostatectomy ,business.industry ,Abiraterone acetate ,Cancer ,Androgen Antagonists ,Middle Aged ,medicine.disease ,Neoadjuvant Therapy ,Androgen receptor ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,medicine.anatomical_structure ,Steroid 16-alpha-Hydroxylase ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,Receptors, Androgen ,Cancer research ,Aryl Hydrocarbon Hydroxylases ,Neoplasm Recurrence, Local ,business - Abstract
Primary prostate cancer can have extensive microheterogeneity, but its contribution to the later emergence of metastatic castration-resistant prostate cancer (mCRPC) remains unclear. In this study, we microdissected residual prostate cancer foci in radical prostatectomies from 18 men treated with neoadjuvant-intensive androgen deprivation therapy (leuprolide, abiraterone acetate, and prednisone) and analyzed them for resistance mechanisms. Transcriptome profiling showed reduced but persistent androgen receptor (AR) activity in residual tumors, with no increase in neuroendocrine differentiation. Proliferation correlated negatively with AR activity but positively with decreased RB1 expression, and whole-exome sequencing (WES) further showed enrichment for RB1 genomic loss. In 15 cases where 2 or 3 tumor foci were microdissected, WES confirmed a common clonal origin but identified multiple oncogenic alterations unique to each focus. These findings show that subclones with oncogenic alterations found in mCRPC are present in primary prostate cancer and are selected for by neoadjuvant-intense androgen deprivation therapy. In particular, this study indicates that subclonal RB1 loss may be more common than previously appreciated in intermediate- to high-risk primary prostate cancer and may be an early event, independent of neuroendocrine differentiation, in the development of mCRPC. Comprehensive molecular analyses of primary prostate cancer may detect aggressive subclones and possibly inform adjuvant strategies to prevent recurrence. Significance: Neoadjuvant androgen deprivation therapy for prostate cancer selects for tumor foci with subclonal genomic alterations, which may comprise the origin of metastatic castration-resistant prostate cancer. Cancer Res; 78(16); 4716–30. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.