1
|
Zang PD, Da Silva DM, Liu ZX, Kandukuri S, Tsao-Wei D, D’Souza A, Kast WM, Pal SK, Kefauver C, Juanqueira M, Yang L, Quinn DI, Dorff TB. Immune Modulation During Treatment with Enzalutamide Alone or with Radium-223 in Patients with Castration Resistant Prostate Cancer. Cancers (Basel) 2025; 17:1730. [PMID: 40427227 PMCID: PMC12110403 DOI: 10.3390/cancers17101730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 05/15/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
INTRODUCTION Prostate cancer has been generally resistant to immunotherapy approaches. Radiation can be immunostimulatory, but the extent to which standard prostate cancer treatments induce immune activation has not been well described. The bone-targeted radiopharmaceutical Radium223 (Ra223) has been proposed to enrich immune function, but clinical studies have not fully delineated whether this is true, or by what mechanisms. Enzalutamide has been shown to increase PD-L1 expression on dendritic cells, which could impact immune activation, though the extent to which this is associated with other evidence of immune activation remains uncertain, and combination strategies remain of interest. We performed a randomized phase II trial to evaluate whether Radium223 (Ra223) added to enzalutamide would induce greater immune activation and clinical responses compared to enzalutamide alone in men with metastatic castration-resistant prostate cancer (mCRPC). METHODS Eligible patients were randomized 2:1 to Arm A (enzalutamide 160 mg PO daily + Ra223 55 kBq/kg IV q4 weeks × 6 doses) or Arm B (enzalutamide 160 mg PO daily). Blood was collected at treatment start and during treatment to measure soluble immune checkpoint biomarkers (BTLA, TIM3, HVEM, GITR, LAG3, PD-1, CTLA-4, PD-L1, PD-L2, ICOS). Immunophenotyping by mass cytometry time of flight (CyTOF) was performed to measure peripheral blood mononuclear cell populations before and after treatment. CyTOF was used to determine changes in circulating immune cell population subsets before and after treatment. Biopsies were performed of an active bone metastatic lesion prior to study treatment and after at least 3 months. IHC was subsequently performed to examine changes in immune cell population subsets before and after treatment, and changes in pSTAT3 levels. RESULTS In total, 30 patients were enrolled, with median age 68. The median duration of follow up was 36 months. PSA responses, PFS, and OS were not significantly different between the two arms; however, the study was not powered for clinical endpoints. Peripheral blood and bone biopsy specimens were analyzed for immune correlatives. Soluble receptor concentrations showed significantly increased expression of PDL-2 in the combination arm, but this was not seen on CyTOF. Otherwise, there were no significant differences in markers of immune activation/exhaustion or immune cell population subsets in the combination arm and enzalutamide monotherapy arm. IHC also did not show a significant difference in immune cell population subsets in bone biopsy specimens before and after treatment in both arms. However, treatment with the combination arm did show significantly increased levels of pSTAT3 (p = 0.04), which was not seen in the enzalutamide monotherapy arm. CONCLUSIONS Our study showed an overall lack of evidence for immune activation or cytokine induction with the combination, which does not make a strong case for combinatorial immunotherapy approaches. However, the combination did induce higher levels of pSTAT3, which has been implicated in radio-resistance. Therefore, the addition of a STAT3 inhibitor to the combination may be of interest to improve efficacy.
Collapse
Affiliation(s)
- Peter D. Zang
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Diane M. Da Silva
- Department of Obstetrics & Gynecology, USC/Norris Comprehensive Cancer Center, 1450 Biggy Street, Los Angeles, CA 90033, USA
- Beckman Center for Immune Monitoring, USC/Norris Comprehensive Cancer Center, 1450 Biggy Street, Los Angeles, CA 90033, USA
| | - Zhang-Xu Liu
- Beckman Center for Immune Monitoring, USC/Norris Comprehensive Cancer Center, 1450 Biggy Street, Los Angeles, CA 90033, USA
| | - Shivani Kandukuri
- Clinical Pathology, USC/Norris Comprehensive Cancer Center, 1500 San Pablo Street, Los Angeles, CA 90033, USA
| | - Denice Tsao-Wei
- Department of Preventative Medicine, USC/Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, CA 90089, USA
| | - Anishka D’Souza
- Clinical Medicine, Department of Medical Oncology, USC/Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, CA 90089, USA
| | - W. Martin Kast
- Beckman Center for Immune Monitoring, USC/Norris Comprehensive Cancer Center, 1450 Biggy Street, Los Angeles, CA 90033, USA
- Department of Molecular Microbiology & Immunology and Urology, USC/Norris Comprehensive Cancer Center, 1450 Biggy Street, Los Angeles, CA 90033, USA
| | - Sumanta K. Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Cheryl Kefauver
- USC/Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, CA 90089, USA
| | - Maribel Juanqueira
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Lixin Yang
- Department of Pathology, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - David I. Quinn
- USC/Norris Comprehensive Cancer Center, 1450 Biggy Street, Los Angeles, CA 90033, USA
- Abbvie, 1000 Gateway Boulevard, South San Francisco, CA 94080, USA
| | - Tanya B. Dorff
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| |
Collapse
|
2
|
Elbialy A, Sood A, Wang SJ, Wang P, Fadiel A, Parwani AV, Huang S, Shvets G, Putluri N, Li J, Liu X. Unveiling racial disparities in prostate cancer using an integrative genomic and transcriptomic analysis. CELL INSIGHT 2025; 4:100238. [PMID: 40104216 PMCID: PMC11914995 DOI: 10.1016/j.cellin.2025.100238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 03/20/2025]
Abstract
Prostate cancer exhibits significant racial disparities, with African American (AA) individuals showing ∼64% higher incidence and 2.3 times greater mortality rates compared to their Caucasian (CA) counterparts. Understanding the complex interplay of genetic, environmental, lifestyle, socioeconomic, and healthcare access factors is crucial for developing effective interventions to reduce this disproportionate burden. This study aims to uncover the genetic and transcriptomic differences driving these disparities through a comprehensive analysis using RNA sequencing (RNA-seq) and exome sequencing of prostate cancer tissues from both Black and White patients. Our transcriptomics analysis revealed enhanced activity in pathways linked to immune response and cellular interactions in AA prostate cancer samples, with notable regulation by histone-associated transcription factors (HIST1H1A, HIST1H1D, and HIST1H1B) suggests potential involvement of histone modification mechanisms. Additionally, pseudogenes and long non-coding RNAs (lncRNAs) among the regulated genes indicate non-coding elements' role in these disparities. Exome sequencing identified unique variants in AA patient samples within key genes, including TP73 (tumor suppression), XYLB (metabolism), ALDH4A1 (oxidative stress), PTPRB (cellular signaling), and HLA-DRB5 (immune response). These genetic variations likely contribute to disease progression and therapy response disparities. This study highlights the importance of considering genetic and epigenetic variations in developing tailored therapeutic approaches to improve treatment efficacy and reduce mortality rates across diverse populations.
Collapse
Affiliation(s)
- Abdalla Elbialy
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Computational Oncology Unit, The University of Chicago Comprehensive Cancer Center, 900 E 57th Street, KCBD Bldg., STE 4144, Chicago, IL, 60637, USA
| | - Akshay Sood
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Shang-Jui Wang
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Peng Wang
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Department of Medicine, College of Medicine, The Ohio State University, Columbus, OH, 3210, USA
| | - Ahmed Fadiel
- Computational Oncology Unit, The University of Chicago Comprehensive Cancer Center, 900 E 57th Street, KCBD Bldg., STE 4144, Chicago, IL, 60637, USA
| | - Anil V Parwani
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Departments of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Steven Huang
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY, 14850, USA
| | - Gennady Shvets
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY, 14850, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jenny Li
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Departments of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Xuefeng Liu
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Departments of Pathology, Urology, and Radiation Oncology, College of Medicine, The Ohio StateUniversity, Columbus, OH, 43210, USA
| |
Collapse
|
3
|
Benabdallah N, Lu P, Abou DS, Zhang H, Ulmert D, Hobbs RF, Gay HA, Simons BW, Saeed MA, Rogers BE, Jha AK, Tai YC, Malone CD, Ippolito JE, Michalski J, Jennings JW, Baumann BC, Pachynski RK, Thorek DLJ. Beyond Average: α-Particle Distribution and Dose Heterogeneity in Bone Metastatic Prostate Cancer. J Nucl Med 2024; 65:245-251. [PMID: 38124163 PMCID: PMC10858382 DOI: 10.2967/jnumed.123.266571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/23/2023] [Indexed: 12/23/2023] Open
Abstract
α-particle emitters are emerging as a potent modality for disseminated cancer therapy because of their high linear energy transfer and localized absorbed dose profile. Despite great interest and pharmaceutical development, there is scant information on the distribution of these agents at the scale of the α-particle pathlength. We sought to determine the distribution of clinically approved [223Ra]RaCl2 in bone metastatic castration-resistant prostate cancer at this resolution, for the first time to our knowledge, to inform activity distribution and dose at the near-cell scale. Methods: Biopsy specimens and blood were collected from 7 patients 24 h after administration. 223Ra activity in each sample was recorded, and the microstructure of biopsy specimens was analyzed by micro-CT. Quantitative autoradiography and histopathology were segmented and registered with an automated procedure. Activity distributions by tissue compartment and dosimetry calculations based on the MIRD formalism were performed. Results: We revealed the activity distribution differences across and within patient samples at the macro- and microscopic scales. Microdistribution analysis confirmed localized high-activity regions in a background of low-activity tissue. We evaluated heterogeneous α-particle emission distribution concentrated at bone-tissue interfaces and calculated spatially nonuniform absorbed-dose profiles. Conclusion: Primary patient data of radiopharmaceutical therapy distribution at the small scale revealed that 223Ra uptake is nonuniform. Dose estimates present both opportunities and challenges to enhance patient outcomes and are a first step toward personalized treatment approaches and improved understanding of α-particle radiopharmaceutical therapies.
Collapse
Affiliation(s)
- Nadia Benabdallah
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Peng Lu
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Diane S Abou
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Hanwen Zhang
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - David Ulmert
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Robert F Hobbs
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Hiram A Gay
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Brian W Simons
- Center for Comparative Medicine, Baylor University, Houston, Texas
| | - Muhammad A Saeed
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Buck E Rogers
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Abhinav K Jha
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Yuan-Chuan Tai
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Christopher D Malone
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Joseph E Ippolito
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Jeff Michalski
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Jack W Jennings
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Brian C Baumann
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
- Department of Radiation Oncology, Springfield Clinic, Springfield, Illinois; and
| | - Russell K Pachynski
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Daniel L J Thorek
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri;
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
- Oncologic Imaging Program, Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| |
Collapse
|
4
|
Gupta S, Fernandez L, Bourdon D, Hamid AA, Pasam A, Lam E, Wenstrup R, Sandhu S. Detection of PSMA expression on circulating tumor cells by blood-based liquid biopsy in prostate cancer. J Circ Biomark 2024; 13:1-6. [PMID: 38415240 PMCID: PMC10895373 DOI: 10.33393/jcb.2024.2636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 01/08/2024] [Indexed: 02/29/2024] Open
Abstract
Background For patients with mCRPC, PSMA-targeted radioligand treatment has significantly improved the clinical outcome. A blood-based liquid biopsy assay for recognizing PSMA protein expression on circulating tumor cells may be beneficial for better informing therapeutic decision-making and identifying the patients most likely to benefit from PSMA-targeted radioligand therapy. Methods Using high-throughput imaging and digital AI pathology algorithms, a four-color immunofluorescence assay has been developed to find PSMA protein expression on CTCs on a glass slide. Cell line cells (LNCaP/PC3s/22Rv1) spiked into healthy donor blood were used to study the precision, specificity, sensitivity, limit of detection, and overall accuracy of the assay. Clinical validation and low-pass whole-genome sequencing were performed in PSMA-PET-positive patients with high-risk mCRPC (N = 24) utilizing 3 mL of blood. Results The PSMA CTC IF assay achieved analytical specificity, sensitivity, and overall accuracy above 99% with high precision. In the clinical validation, 76% (16/21) of the cases were PSMA positive with CTC heterogeneity, and 88% (21/24) of the patients contained at least one conventional CTC per milliliter of blood. Thirty-six low-pass-sequenced CTCs from 11 individuals with mCRPC frequently exhibited copy number increases in AR and MYC and losses in RB1, PTEN, TP53, and BRCA2 locus. Conclusions The analytical validation utilizing Epic Sciences' liquid biopsy CTC platform demonstrated the potential to detect PSMA protein expression in CTCs from patients with mCRPC. This assay is positioned as an effective research tool to evaluate PSMA expression, heterogeneity, and therapeutic response in many ongoing clinical studies to target tumors that express PSMA.
Collapse
Affiliation(s)
- Santosh Gupta
- Translational Research and Assay Development, Epic Sciences, San Diego, California - USA
| | - Luisa Fernandez
- Translational Research and Assay Development, Epic Sciences, San Diego, California - USA
| | - David Bourdon
- Translational Research and Assay Development, Epic Sciences, San Diego, California - USA
| | - Anis A Hamid
- Department of Medical Oncology, University of Melbourne, Melbourne, Victoria - Australia
| | - Anupama Pasam
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria - Australia
| | - Ernest Lam
- Translational Research and Assay Development, Epic Sciences, San Diego, California - USA
| | - Richard Wenstrup
- Translational Research and Assay Development, Epic Sciences, San Diego, California - USA
| | - Shahneen Sandhu
- Department of Medical Oncology, University of Melbourne, Melbourne, Victoria - Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria - Australia
| |
Collapse
|
5
|
de Jong AC, Isebia KT, Ling SW, de Weerd V, Van NM, Kraan J, Martens JWM, Mehra N, Hamberg P, Lolkema MP, de Wit R, van der Veldt AAM, Wilting SM. Liquid Biopsies for Early Response Evaluation of Radium-223 in Metastatic Prostate Cancer. JCO Precis Oncol 2023; 7:e2300156. [PMID: 38061007 DOI: 10.1200/po.23.00156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/07/2023] [Accepted: 09/26/2023] [Indexed: 12/18/2023] Open
Abstract
PURPOSE Reliable biomarkers for response monitoring during radium-223 treatment in patients with metastatic castration-resistant prostate cancer (mCRPC) are lacking. Circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA), obtained from liquid biopsies, are shown to have prognostic value in mCRPC. The aim of this study was to determine the value of CTCs and ctDNA for response evaluation of radium-223. METHODS In this prospective multicenter study, longitudinal blood draws and imaging were performed in 28 patients with mCRPC and predominantly bone disease, who were treated with radium-223. CTCs were counted (CELLSEARCH CTC test), while fraction of ctDNA was estimated by measuring aneuploidy of cell-free DNA (cfDNA; modified Fast Aneuploidy Screening Test-Sequencing System). CTC counts and aneuploidy score (AS) were categorized as low (<5) and high (≥5). Primary and secondary clinical end points were failure-free survival (FFS), and overall survival (OS) and development of extraosseous metastases, respectively. Additionally, CTC count and AS were related to alkaline phosphatase (ALP) and total tumor volume in bone (TTVbone) on positron emission tomography-computed tomography with 68gallium prostate-specific membrane antigen. RESULTS FFS was longer in patients with a low CTC count or AS either at baseline or after 12 weeks, whereas for OS, only a significant association with CTC count was observed. Liquid biopsy results correlated well with ALP and TTVbone at baseline, but not with change in both parameters after three cycles of radium-223. AS and CTC count were significantly correlated. CONCLUSION CTC count and AS of cfDNA at baseline and during treatment predict clinical response to radium-223 in patients with mCRPC, warranting future evaluation of their value in treatment guidance.
Collapse
Affiliation(s)
- Anouk C de Jong
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Khrystany T Isebia
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Sui Wai Ling
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Vanja de Weerd
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ngoc M Van
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Jaco Kraan
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud UMC, Nijmegen, the Netherlands
| | - Paul Hamberg
- Department of Internal Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, the Netherlands
| | - Martijn P Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ronald de Wit
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Saskia M Wilting
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| |
Collapse
|
6
|
|
7
|
Hawkey NM, Broderick A, George DJ, Sartor O, Armstrong AJ. The Value of Phenotypic Precision Medicine in Prostate Cancer. Oncologist 2022; 28:93-104. [PMID: 36200788 PMCID: PMC9907055 DOI: 10.1093/oncolo/oyac198] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/12/2022] [Indexed: 11/14/2022] Open
Abstract
Prostate cancer is the most common cancer among men and the second leading cause of cancer-related death. For patients who develop metastatic disease, tissue-based and circulating-tumor-based molecular and genomic biomarkers have emerged as a means of improving outcomes through the application of precision medicine. However, the benefit is limited to a minority of patients. An additional approach to further characterize the biology of advanced prostate cancer is through the use of phenotypic precision medicine, or the identification and targeting of phenotypic features of an individual patient's cancer. In this review article, we will discuss the background, potential clinical benefits, and limitations of genomic and phenotypic precision medicine in prostate cancer. We will also highlight how the emergence of image-based phenotypic medicine may lead to greater characterization of advanced prostate cancer disease burden and more individualized treatment approaches in patients.
Collapse
Affiliation(s)
- Nathan M Hawkey
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Amanda Broderick
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Daniel J George
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, USA,Duke Cancer Institute Center for Prostate and Urologic Cancer, Duke University, Durham, NC, USA
| | - Oliver Sartor
- Tulane Cancer Center, Division of Genitourinary Oncology, New Orleans, LA, USA
| | - Andrew J Armstrong
- Corresponding author: Andrew J. Armstrong, MD, ScM, FACP, Department of Medicine, Surgery, Pharmacology and Cancer Biology, Director of Research, the Duke Cancer Institute Center for Prostate and Urologic Cancers, Divisions of Medical Oncology and Urology, Duke University, DUMC Box 103861, Durham, NC 27710, USA;
| |
Collapse
|
8
|
Gupta S, Halabi S, Kemeny G, Anand M, Giannakakou P, Nanus DM, George DJ, Gregory SG, Armstrong AJ. Circulating Tumor Cell Genomic Evolution and Hormone Therapy Outcomes in Men with Metastatic Castration-Resistant Prostate Cancer. Mol Cancer Res 2021; 19:1040-1050. [PMID: 33771885 PMCID: PMC8178231 DOI: 10.1158/1541-7786.mcr-20-0975] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/11/2021] [Accepted: 03/01/2021] [Indexed: 01/24/2023]
Abstract
Men with circulating tumor cell (CTC) AR-V7-positive metastatic castration-resistant prostate cancer (mCRPC) have worse outcomes when treated with enzalutamide/abiraterone. However, most men lack CTC AR-V7 detection, and additional predictive biomarkers are needed. We conducted a retrospective secondary analysis of the prospective PROPHECY trial (NCT02269982) of men with mCRPC undergoing treatment with enzalutamide/abiraterone, analyzing pooled CTC and germline DNA for whole-genome copy-number alterations (CNA) in 73 samples from 48 men over time along with pooled CTC and germline whole-exome sequencing on 22 paired samples before and following progression on androgen receptor (AR) inhibitor therapy to identify somatic genomic alterations associated with acquired resistance. We observed broad interpatient and longitudinal CTC genomic heterogeneity from AR-V7-negative men with mCRPC, including common gains of KDM6A, MYCN, and AR, and loss of ZFHX3, BRCA1, and PTEN. Men who had progression-free survival of ≤3 months despite enzalutamide/abiraterone treatment were more likely to have baseline CTC genomic loss of CHD1, PTEN, PHLPP1, and ZFHX3 and gains of BRCA2, KDM5D, MYCN, and SPARC. After progression on abiraterone/enzalutamide, we observed clonal evolution of CTCs harboring TP53 mutations and gain of ATM, KDM6A, and MYC, and loss of NCOR1, PTEN, RB1, and RUNX2. CTC genomic findings were independently confirmed in a separate cohort of mCRPC men who progressed despite prior treatment with abiraterone/enzalutamide (NCT02204943). IMPLICATIONS: We identified common and reproducible genomic alterations in CTCs from AR-V7-negative mCRPC men associated with poor outcomes during enzalutamide/abiraterone treatment, including CNAs in genes linked to lineage plasticity and epigenetic signaling, DNA repair, AR, TP53/RB1, PTEN, and WNT pathways.
Collapse
Affiliation(s)
- Santosh Gupta
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolin
- Epic Sciences, San Diego, California
| | - Susan Halabi
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina
| | - Gabor Kemeny
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Monika Anand
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | | | - David M Nanus
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Daniel J George
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
- Departments of Medicine, Surgery, Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Simon G Gregory
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolin
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina.
- Departments of Medicine, Surgery, Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| |
Collapse
|
9
|
Marques IA, Abrantes AM, Pires AS, Neves AR, Caramelo FJ, Rodrigues T, Matafome P, Tavares-da-Silva E, Gonçalves AC, Pereira CC, Teixeira JP, Seiça R, Costa G, Figueiredo A, Botelho MF. Kinetics of radium-223 and its effects on survival, proliferation and DNA damage in lymph-node and bone metastatic prostate cancer cell lines. Int J Radiat Biol 2021; 97:714-726. [PMID: 33764249 DOI: 10.1080/09553002.2021.1906462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/15/2021] [Accepted: 03/15/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Metastatic castration-resistant prostate cancer (mCRPC) is associated with a very unfavorable prognosis. At this advanced stage of the disease, there are several therapeutic strategies approved in recent times, being one of them Radium-223 dichloride (Radium-223). However, its mechanisms of action and the process that conducts to cell death are not fully understood. Given this, our main goal is to characterize the radiobiological effects induced by Radium-223 and to evaluate its kinetics on metastatic Prostate Cancer (mPCa) cells. MATERIALS AND METHODS In vitro studies were conducted using two mPCa cell lines, the LNCaP and PC3, the first being derived from lymph node metastasis and the second from bone metastasis. Kinetic studies were conducted to access the capacity of these cell lines to uptake, retain and internalize the Radium-223. For the assessment of radiobiological effects, cells were first exposed to different doses of Radium-223 and the clonogenic assay was done to evaluate cell survival and to determine lethal doses (LD50). Then, the effects were also evaluated in terms of proliferation, oxidative stress, morphological changes and cell damage. RESULTS Radium-223 is uptaken by mPCa cells and reaches the nucleus, where it is retained over time. Irradiation decreases cell survival and proliferation, with LNCaP cells (LD50 = 1.73mGy) being more radiosensitive than PC3 cells (LD50 = 4.20mGy). Irradiated cells showed morphological changes usually associated with apoptosis and a dose-dependent increase in DNA damage. Moreover, activation of cell cycle checkpoints occurs through ATM/CHK2 pathway, which is involved in cell cycle arrest and cell death. CONCLUSIONS The cytotoxic and anti-proliferative effects on both cell lines showed that Radium-223 can decrease the aggressiveness of tumor cells by decreasing the cell survival and proliferation and, also, by increasing the DNA damage. The similar results observed in both cell lines indicated that Radium-223 may have the potential to be used as a therapeutic option also for mCRPC patients with lymph node metastasis. The activation of DNA Damage Response pathways allows the possibility to understand the importance of these checkpoints as targets for new combined therapies.
Collapse
Affiliation(s)
- Inês A Marques
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| | - Ana M Abrantes
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Ana S Pires
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Ana R Neves
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Project Development Office, Department of Mathematics and Computer Science, Eindhoven University of Technology (TU/e), Eindhoven, The Netherlands
| | - Francisco J Caramelo
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Laboratory of Biostatistics and Medical Informatics of Faculty of Medicine, Coimbra, Portugal
| | - Tiago Rodrigues
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Laboratory of Physiology of Faculty of Medicine, Coimbra, Portugal
| | - Paulo Matafome
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Laboratory of Physiology of Faculty of Medicine, Coimbra, Portugal
| | - Edgar Tavares-da-Silva
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra (CHUC), Department of Urology and Renal Transplantation, Coimbra, Portugal
| | - Ana C Gonçalves
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Laboratory of Oncobiology and Hematology and University Clinic of Hematology of Faculty of Medicine, Coimbra, Portugal
| | - Cristiana C Pereira
- National Institute of Health, Environmental Health Department, Porto, Portugal
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
| | - João P Teixeira
- National Institute of Health, Environmental Health Department, Porto, Portugal
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
| | - Raquel Seiça
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Laboratory of Physiology of Faculty of Medicine, Coimbra, Portugal
| | - Grancinda Costa
- Centro Hospitalar e Universitário de Coimbra (CHUC), Department of Nuclear Medicine, Coimbra, Portugal
| | - Arnaldo Figueiredo
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra (CHUC), Department of Urology and Renal Transplantation, Coimbra, Portugal
| | - Maria F Botelho
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| |
Collapse
|
10
|
Ashe H, Krakowiak P, Hasterok S, Sleppy R, Roller DG, Gioeli D. Role of the runt-related transcription factor (RUNX) family in prostate cancer. FEBS J 2021; 288:6112-6126. [PMID: 33682350 DOI: 10.1111/febs.15804] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/22/2021] [Accepted: 03/05/2021] [Indexed: 12/13/2022]
Abstract
Prostate cancer (PCa) is a very complex disease that is a major cause of death in men worldwide. Currently, PCa dependence on the androgen receptor (AR) has resulted in use of AR antagonists and antiandrogen therapies that reduce endogenous steroid hormone production. However, within two to three years of receiving first-line androgen deprivation therapy, the majority of patients diagnosed with PCa progress to castration-resistant prostate cancer (CRPC). There is an urgent need for therapies that are more durable than antagonism of the AR axis. Studies of runt-related transcription factors (RUNX) and their heterodimerization partner, core-binding factor subunit b (CBFβ), are revealing that the RUNX family are drivers of CRPC. In this review, we describe what is presently understood about RUNX members in PCa, including what regulates and is regulated by RUNX proteins, and the role of RUNX proteins in the tumor microenvironment and AR signaling. We discuss the implications for therapeutically targeting RUNX, the potential for RUNX as PCa biomarkers, and the current pressing questions in the field.
Collapse
Affiliation(s)
- Hannah Ashe
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Patryk Krakowiak
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Sylwia Hasterok
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Rosalie Sleppy
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Devin G Roller
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Daniel Gioeli
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA.,University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
11
|
Theil G, Fornara P, Bialek J. Position of Circulating Tumor Cells in the Clinical Routine in Prostate Cancer and Breast Cancer Patients. Cancers (Basel) 2020; 12:cancers12123782. [PMID: 33333999 PMCID: PMC7765455 DOI: 10.3390/cancers12123782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Many different therapies are applied to fight tumor disease. Blood-based biosources, like circulating tumor cells (CTCs), offer the opportunity to monitor the healing progression and the real-time response to the therapy. In this review, we analyze the outcomes of the clinical trials and scientific studies of prostate and breast cancer performed in the decade between April 2010 and April 2020. Additionally, we describe the abstracts from the 4th Advances in Circulating Tumor Cells (ACTC) meeting in 2019. We discuss the potential therapeutic opportunities related to the CTCs and the challenges ahead in the routine treatment of cancer. Abstract Prostate cancer and breast cancer are the most common cancers worldwide. Anti-tumor therapies are long and exhaustive for the patients. The real-time monitoring of the healing progression could be a useful tool to evaluate therapeutic response. Blood-based biosources like circulating tumor cells (CTCs) may offer this opportunity. Application of CTCs for the clinical diagnostics could improve the sequenced screening, provide additional valuable information of tumor dynamics, and help personalized management for the patients. In the past decade, CTCs as liquid biopsy (LB) has received tremendous attention. Many different isolation and characterization platforms are developed but the clinical validation is still missing. In this review, we focus on the clinical trials of circulating tumor cells that have the potential to monitor and stratify patients and lead to implementation into clinical practice.
Collapse
|
12
|
Liu X, Mondal AM. Conditional cell reprogramming for modeling host-virus interactions and human viral diseases. J Med Virol 2020; 92:2440-2452. [PMID: 32478897 PMCID: PMC7586785 DOI: 10.1002/jmv.26093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 01/08/2023]
Abstract
Conventional cancer and transformed cell lines are widely used in cancer biology and other fields within biology. These cells usually have abnormalities from the original tumor itself, but may also develop abnormalities due to genetic manipulation, or genetic and epigenetic changes during long-term passages. Primary cultures may maintain lineage functions as the original tissue types, yet they have a very limited life span or population doubling time because of the nature of cellular senescence. Primary cultures usually have very low yields, and the high variability from any original tissue specimens, largely limiting their applications in research. Animal models are often used for studies of virus infections, disease modeling, development of antiviral drugs, and vaccines. Human viruses often need a series of passages in vivo to adapt to the host environment because of variable receptors on the cell surface and may have intracellular restrictions from the cell types or host species. Here, we describe a long-term cell culture system, conditionally reprogrammed cells (CRCs), and its applications in modeling human viral diseases and drug discovery. Using feeder layer coculture in presence of Y-27632 (conditional reprogramming, CR), CRCs can be obtained and rapidly propagated from surgical specimens, core or needle biopsies, and other minimally invasive or noninvasive specimens, for example, nasal cavity brushing. CRCs preserve their lineage functions and provide biologically relevant and physiological conditions, which are suitable for studies of viral entry and replication, innate immune responses of host cells, and discovery of antiviral drugs. In this review, we summarize the applications of CR technology in modeling host-virus interactions and human viral diseases including severe acute respiratory syndrome coronavirus-2 and coronavirus disease-2019, and antiviral discovery.
Collapse
Affiliation(s)
- Xuefeng Liu
- Department of Pathology, Center for Cell ReprogrammingGeorgetown University Medical CenterWashingtonDC
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| | - Abdul M. Mondal
- Department of Pathology, Center for Cell ReprogrammingGeorgetown University Medical CenterWashingtonDC
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| |
Collapse
|
13
|
Tang L, Xu M, Zhang L, Qu L, Liu X. Role of αVβ3 in Prostate Cancer: Metastasis Initiator and Important Therapeutic Target. Onco Targets Ther 2020; 13:7411-7422. [PMID: 32801764 PMCID: PMC7395689 DOI: 10.2147/ott.s258252] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022] Open
Abstract
In prostate cancer, distant organ metastasis is the leading cause of patient death. Although the mechanism of malignant tumor metastasis is unclear, studies have confirmed that integrin αVβ3 plays an important role in this process. In prostate cancer, αVβ3 mediates adhesion, invasion, immune escape and neovascularization through interactions with different ligands. Among these ligands and in addition to proteins that are directly related to tumor invasion, other proteins that contain the RGD structure could also bind to αVβ3 and cause a number of biological effects. In this article, we summarized the ligand and downstream proteins related to αVβ3-mediated prostate cancer metastasis as well as some diagnostic and therapeutic measures targeting αVβ3.
Collapse
Affiliation(s)
- Lin Tang
- College of Mathematics and Computer Science, Chifeng University, Chifeng, The Inner Mongol Autonomous Region 024005, People's Republic of China
| | - Meng Xu
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, People's Republic of China.,R&D Department, Seekgene Technology Co., Ltd, Beijing 100000, People's Republic of China
| | - Long Zhang
- Department of Hepatobiliary Surgery, Yidu Central Hospital, Weifang, Shandong 262500, People's Republic of China
| | - Lin Qu
- Department of Orthopaedic Surgery, Anshan Hospital of the First Hospital of China Medical University, Anshan, Liaoning 114000, People's Republic of China
| | - Xiaoyan Liu
- Department of Pathology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100000, People's Republic of China
| |
Collapse
|
14
|
Gupta S, Hovelson DH, Kemeny G, Halabi S, Foo WC, Anand M, Somarelli JA, Tomlins SA, Antonarakis ES, Luo J, Dittamore RV, George DJ, Rothwell C, Nanus DM, Armstrong AJ, Gregory SG. Discordant and heterogeneous clinically relevant genomic alterations in circulating tumor cells vs plasma DNA from men with metastatic castration resistant prostate cancer. Genes Chromosomes Cancer 2019; 59:225-239. [PMID: 31705765 DOI: 10.1002/gcc.22824] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Circulating tumor cell (CTC) and cell-free (cf) DNA-based genomic alterations are increasingly being used for clinical decision-making in oncology. However, the concordance and discordance between paired CTC and cfDNA genomic profiles remain largely unknown. We performed comparative genomic hybridization (CGH) on CTCs and cfDNA, and low-pass whole genome sequencing (lpWGS) on cfDNA to characterize genomic alterations (CNA) and tumor content in two independent prospective studies of 93 men with mCRPC treated with enzalutamide/abiraterone, or radium-223. Comprehensive analysis of 69 patient CTCs and 72 cfDNA samples from 93 men with mCRPC, including 64 paired samples, identified common concordant gains in FOXA1, AR, and MYC, and losses in BRCA1, PTEN, and RB1 between CTCs and cfDNA. Concordant PTEN loss and discordant BRCA2 gain were associated with significantly worse outcomes in Epic AR-V7 negative men with mCRPC treated with abiraterone/enzalutamide. We identified and externally validated CTC-specific genomic alternations that were discordant in paired cfDNA, even in samples with high tumor content. These CTC/cfDNA-discordant regions included key genomic regulators of lineage plasticity, osteomimicry, and cellular differentiation, including MYCN gain in CTCs (31%) that was rarely detected in cfDNA. CTC MYCN gain was associated with poor clinical outcomes in AR-V7 negative men and small cell transformation. In conclusion, we demonstrated concordance of multiple genomic alterations across CTC and cfDNA platforms; however, some genomic alterations displayed substantial discordance between CTC DNA and cfDNA despite the use of identical copy number analysis methods, suggesting tumor heterogeneity and divergent evolution associated with poor clinical outcomes.
Collapse
Affiliation(s)
- Santosh Gupta
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina.,Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Daniel H Hovelson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Gabor Kemeny
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina
| | - Wen-Chi Foo
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Monika Anand
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Jason A Somarelli
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina.,Department of Medicine, Surgery, Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Scott A Tomlins
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Emmanuel S Antonarakis
- Prostate Cancer Research Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jun Luo
- James Buchanan Brady Urological Institute and the Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Daniel J George
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - Colin Rothwell
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - David M Nanus
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina.,Department of Medicine, Surgery, Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Simon G Gregory
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina.,Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| |
Collapse
|