201
|
Kong Y, Cheng L, Mao F, Zhang Z, Zhang Y, Farah E, Bosler J, Bai Y, Ahmad N, Kuang S, Li L, Liu X. Inhibition of cholesterol biosynthesis overcomes enzalutamide resistance in castration-resistant prostate cancer (CRPC). J Biol Chem 2018; 293:14328-14341. [PMID: 30089652 DOI: 10.1074/jbc.ra118.004442] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/02/2018] [Indexed: 01/01/2023] Open
Abstract
Enzalutamide, a nonsteroidal second-generation antiandrogen, has been recently approved for the management of castration-resistant prostate cancer (CRPC). Although patients can benefit from enzalutamide at the beginning of this therapy, acquired enzalutamide resistance usually occurs within a short period. This motivated us to investigate the mechanism involved and possible approaches for overcoming enzalutamide resistance in CRPC. In the present study, we found that 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR), a crucial enzyme in the mevalonate pathway for sterol biosynthesis, is elevated in enzalutamide-resistant prostate cancer cell lines. HMGCR knockdown could resensitize these cells to the drug, and HMGCR overexpression conferred resistance to it, suggesting that aberrant HMGCR expression is an important enzalutamide-resistance mechanism in prostate cancer cells. Furthermore, enzalutamide-resistant prostate cancer cells were more sensitive to statins, which are HMGCR inhibitors. Of note, a combination of simvastatin and enzalutamide significantly inhibited the growth of enzalutamide-resistant prostate cancer cells in vitro and tumors in vivo Mechanistically, simvastatin decreased protein levels of the androgen receptor (AR), which was further reduced in combination with enzalutamide. We observed that the decrease in AR may occur through simvastatin-mediated inhibition of the mTOR pathway, whose activation was associated with increased HMGCR and AR expression. These results indicate that simvastatin enhances the efficacy of enzalutamide-based therapy, highlighting the therapeutic potential of statins to overcome enzalutamide resistance in CRPC.
Collapse
Affiliation(s)
- Yifan Kong
- From the Departments of Biochemistry and.,Animal Sciences and
| | - Lijun Cheng
- the Department of Biomedical Informatics, Ohio State University, Columbus, Ohio 43210, and
| | - Fengyi Mao
- From the Departments of Biochemistry and.,Animal Sciences and
| | | | | | - Elia Farah
- From the Departments of Biochemistry and
| | | | | | - Nihal Ahmad
- the Department of Dermatology, University of Wisconsin, Madison, Wisconsin 53715
| | - Shihuan Kuang
- Animal Sciences and.,the Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| | - Lang Li
- the Department of Biomedical Informatics, Ohio State University, Columbus, Ohio 43210, and
| | - Xiaoqi Liu
- From the Departments of Biochemistry and .,the Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| |
Collapse
|
202
|
Plyduang T, Armiñán A, Movellan J, England RM, Wiwattanapatapee R, Vicent MJ. Polyacetal-Based Combination Therapy for the Treatment of Prostate Cancer. Macromol Rapid Commun 2018; 39:e1800265. [DOI: 10.1002/marc.201800265] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/11/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Thipapun Plyduang
- Department of Pharmaceutical Technology; Faculty of Pharmaceutical Sciences; Prince of Songkla University; Hat Yai Songkla 90112 Thailand
- School of Pharmacy; Walailak University; Thasala Nakhon Si Thammarat 80161 Thailand
| | - Ana Armiñán
- Polymer Therapeutics Laboratory; Centro de Investigación Príncipe Felipe; Av Eduardo Primo Yúfera 3 46012 Valencia Spain
| | - Julie Movellan
- Polymer Therapeutics Laboratory; Centro de Investigación Príncipe Felipe; Av Eduardo Primo Yúfera 3 46012 Valencia Spain
| | - Richard M. England
- Polymer Therapeutics Laboratory; Centro de Investigación Príncipe Felipe; Av Eduardo Primo Yúfera 3 46012 Valencia Spain
| | - Ruedeekorn Wiwattanapatapee
- Department of Pharmaceutical Technology; Faculty of Pharmaceutical Sciences; Prince of Songkla University; Hat Yai Songkla 90112 Thailand
| | - María J. Vicent
- Polymer Therapeutics Laboratory; Centro de Investigación Príncipe Felipe; Av Eduardo Primo Yúfera 3 46012 Valencia Spain
| |
Collapse
|
203
|
Singh KB, Ji X, Singh SV. Therapeutic Potential of Leelamine, a Novel Inhibitor of Androgen Receptor and Castration-Resistant Prostate Cancer. Mol Cancer Ther 2018; 17:2079-2090. [PMID: 30030299 DOI: 10.1158/1535-7163.mct-18-0117] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/19/2018] [Accepted: 07/16/2018] [Indexed: 01/31/2023]
Abstract
Clinical management of castration-resistant prostate cancer (CRPC) resulting from androgen deprivation therapy remains challenging. CRPC is driven by aberrant activation of androgen receptor (AR) through mechanisms ranging from its amplification, mutation, post-translational modification, and expression of splice variants (e.g., AR-V7). Herein, we present experimental evidence for therapeutic vulnerability of CRPC to a novel phytochemical, leelamine (LLM), derived from pine tree bark. Exposure of human prostate cancer cell lines LNCaP (an androgen-responsive cell line with mutant AR), C4-2B (an androgen-insensitive variant of LNCaP), and 22Rv1 (a CRPC cell line with expression of AR-Vs), and a murine prostate cancer cell line Myc-CaP to plasma achievable concentrations of LLM resulted in ligand-dependent (LNCaP) and ligand-independent (22Rv1) growth inhibition in vitro that was accompanied by downregulation of mRNA and/or protein levels of full-length AR as well as its splice variants, including AR-V7. LLM treatment resulted in apoptosis induction in the absence and presence of R1881. In silico modeling followed by luciferase reporter assay revealed a critical role for noncovalent interaction of LLM with Y739 in AR activity inhibition. Substitution of the amine group with an isothiocyanate functional moiety abolished AR and cell viability inhibition by LLM. Administration of LLM resulted in 22Rv1 xenograft growth suppression that was statistically insignificant but was associated with a significant decrease in Ki-67 expression, mitotic activity, expression of full-length AR and AR-V7 proteins, and secretion of PSA. This study identifies a novel chemical scaffold for the treatment of CRPC. Mol Cancer Ther; 17(10); 2079-90. ©2018 AACR.
Collapse
Affiliation(s)
- Krishna B Singh
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xinhua Ji
- Macromolecular Crystallography Laboratory, National Cancer Institute, Frederick, Maryland
| | - Shivendra V Singh
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. .,UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
204
|
Kallio HML, Hieta R, Latonen L, Brofeldt A, Annala M, Kivinummi K, Tammela TL, Nykter M, Isaacs WB, Lilja HG, Bova GS, Visakorpi T. Constitutively active androgen receptor splice variants AR-V3, AR-V7 and AR-V9 are co-expressed in castration-resistant prostate cancer metastases. Br J Cancer 2018; 119:347-356. [PMID: 29988112 PMCID: PMC6070921 DOI: 10.1038/s41416-018-0172-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/08/2018] [Accepted: 06/13/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND A significant subset of prostate cancer (PC) patients with a castration-resistant form of the disease (CRPC) show primary resistance to androgen receptor (AR)-targeting drugs developed against CRPC. As one explanation could be the expression of constitutively active androgen receptor splice variants (AR-Vs), our current objectives were to study AR-Vs and other AR aberrations to better understand the emergence of CRPC. METHODS We analysed specimens from different stages of prostate cancer by next-generation sequencing and immunohistochemistry. RESULTS AR mutations and copy number variations were detected only in CRPC specimens. Genomic structural rearrangements of AR were observed in 5/30 metastatic CRPC patients, but they were not associated with expression of previously known AR-Vs. The predominant AR-Vs detected were AR-V3, AR-V7 and AR-V9, with the expression levels being significantly higher in CRPC cases compared to prostatectomy samples. Out of 25 CRPC metastases that expressed any AR variant, 17 cases harboured expression of all three of these AR-Vs. AR-V7 protein expression was highly heterogeneous and higher in CRPC compared to hormone-naïve tumours. CONCLUSIONS AR-V3, AR-V7 and AR-V9 are co-expressed in CRPC metastases highlighting the fact that inhibiting AR function via regions common to all AR-Vs is likely to provide additional benefit to patients with CRPC.
Collapse
Affiliation(s)
- Heini M L Kallio
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland.
| | - Reija Hieta
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Leena Latonen
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Anniina Brofeldt
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Matti Annala
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Kati Kivinummi
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Teuvo L Tammela
- Department of Urology, University of Tampere, Tampere University Hospital, Tampere, Finland
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - William B Isaacs
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hans G Lilja
- Departments of Surgery (Urology), Laboratory Medicine and Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.,Department of Translational Medicine, Lund University, Malmö, Sweden.,Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - G Steven Bova
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Tapio Visakorpi
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland.,Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
205
|
Systems pharmacology using mass spectrometry identifies critical response nodes in prostate cancer. NPJ Syst Biol Appl 2018; 4:26. [PMID: 29977602 PMCID: PMC6026592 DOI: 10.1038/s41540-018-0064-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/17/2018] [Accepted: 05/23/2018] [Indexed: 01/27/2023] Open
Abstract
In the United States alone one in five newly diagnosed cancers in men are prostate carcinomas (PCa). Androgen receptor (AR) status and the PI3K-AKT-mTOR signal transduction pathway are critical in PCa. After initial response to single drugs targeting these pathways resistance often emerges, indicating the need for combination therapy. Here, we address the question of efficacy of drug combinations and development of resistance mechanisms to targeted therapy by a systems pharmacology approach. We combine targeted perturbation with detailed observation of the molecular response by mass spectrometry. We hypothesize that the molecular short-term (24 h) response reveals details of how PCa cells adapt to counter the anti-proliferative drug effect. With focus on six drugs currently used in PCa treatment or targeting the PI3K-AKT-mTOR signal transduction pathway, we perturbed the LNCaP clone FGC cell line by a total of 21 treatment conditions using single and paired drug combinations. The molecular response was analyzed by the mass spectrometric quantification of 52 proteins. Analysis of the data revealed a pattern of strong responders, i.e., proteins that were consistently downregulated or upregulated across many of the perturbation conditions. The downregulated proteins, HN1, PAK1, and SPAG5, are potential early indicators of drug efficacy and point to previously less well-characterized response pathways in PCa cells. Some of the upregulated proteins such as 14-3-3 proteins and KLK2 may be useful early markers of adaptive response and indicate potential resistance pathways targetable as part of combination therapy to overcome drug resistance. The potential of 14-3-3ζ (YWHAZ) as a target is underscored by the independent observation, based on cancer genomics of surgical specimens, that its DNA copy number and transcript levels tend to increase with PCa disease progression. The combination of systematic drug perturbation combined with detailed observation of short-term molecular response using mass spectrometry is a potentially powerful tool to discover response markers and anti-resistance targets. Metastatic prostate cancer is often treated with pharmacological agents to prevent the tumor from expanding; however, despite advances in drug development patients often die of the disease. An international research team lead by Ruedi Aebersold (ETH Zürich, Switzerland) and Chris Sander (Dana Faber Cancer Institute, Boston, USA) asked how prostate cancer cells adapt to pharmacological treatment on the molecular protein level and find a general response in their prostate cancer model. Next, they asked if similar changes are found in prostate cancer patients. Indeed, the same proteins upregulated in prostate cancer models are also upregulated in prostate cancer patients. Immediately, this has implications for patient treatment stratification and opens new avenues for drug developments in metastatic prostate cancer.
Collapse
|
206
|
Shore ND, Tammela TL, Massard C, Bono P, Aspegren J, Mustonen M, Fizazi K. Safety and Antitumour Activity of ODM-201 (BAY-1841788) in Chemotherapy-naïve and CYP17 Inhibitor-naïve Patients: Follow-up from the ARADES and ARAFOR Trials. Eur Urol Focus 2018; 4:547-553. [DOI: 10.1016/j.euf.2017.01.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/04/2017] [Accepted: 01/24/2017] [Indexed: 12/12/2022]
|
207
|
Verma K, Gupta N, Zang T, Wangtrakluldee P, Srivastava SK, Penning TM, Trippier PC. AKR1C3 Inhibitor KV-37 Exhibits Antineoplastic Effects and Potentiates Enzalutamide in Combination Therapy in Prostate Adenocarcinoma Cells. Mol Cancer Ther 2018; 17:1833-1845. [PMID: 29891491 DOI: 10.1158/1535-7163.mct-17-1023] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/01/2018] [Accepted: 06/04/2018] [Indexed: 11/16/2022]
Abstract
Aldo-keto reductase 1C3 (AKR1C3), also known as type 5 17 β-hydroxysteroid dehydrogenase, is responsible for intratumoral androgen biosynthesis, contributing to the development of castration-resistant prostate cancer (CRPC) and eventual chemotherapeutic failure. Significant upregulation of AKR1C3 is observed in CRPC patient samples and derived CRPC cell lines. As AKR1C3 is a downstream steroidogenic enzyme synthesizing intratumoral testosterone (T) and 5α-dihydrotestosterone (DHT), the enzyme represents a promising therapeutic target to manage CRPC and combat the emergence of resistance to clinically employed androgen deprivation therapy. Herein, we demonstrate the antineoplastic activity of a potent, isoform-selective and hydrolytically stable AKR1C3 inhibitor (E)-3-(4-(3-methylbut-2-en-1-yl)-3-(3-phenylpropanamido)phenyl)acrylic acid (KV-37), which reduces prostate cancer cell growth in vitro and in vivo and sensitizes CRPC cell lines (22Rv1 and LNCaP1C3) toward the antitumor effects of enzalutamide. Crucially, KV-37 does not induce toxicity in nonmalignant WPMY-1 prostate cells nor does it induce weight loss in mouse xenografts. Moreover, KV-37 reduces androgen receptor (AR) transactivation and prostate-specific antigen expression levels in CRPC cell lines indicative of a therapeutic effect in prostate cancer. Combination studies of KV-37 with enzalutamide reveal a very high degree of synergistic drug interaction that induces significant reduction in prostate cancer cell viability via apoptosis, resulting in >200-fold potentiation of enzalutamide action in drug-resistant 22Rv1 cells. These results demonstrate a promising therapeutic strategy for the treatment of drug-resistant CRPC that invariably develops in prostate cancer patients following initial treatment with AR antagonists such as enzalutamide. Mol Cancer Ther; 17(9); 1833-45. ©2018 AACR.
Collapse
Affiliation(s)
- Kshitij Verma
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas
| | - Nehal Gupta
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas
| | - Tianzhu Zang
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Phumvadee Wangtrakluldee
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sanjay K Srivastava
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas.,Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Abilene, Texas
| | - Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas. .,Center for Chemical Biology, Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas
| |
Collapse
|
208
|
Brown LC, Sonpavde G, Armstrong AJ. Can RECIST response predict success in phase 3 trials in men with metastatic castration-resistant prostate cancer? Prostate Cancer Prostatic Dis 2018; 21:419-430. [PMID: 29858595 DOI: 10.1038/s41391-018-0049-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/11/2018] [Accepted: 03/19/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND Intermediate endpoints are needed in early phase studies of men with metastatic castration-resistant prostate cancer (mCRPC) that can reliably predict success in phase 3 trials. Among men with measurable disease, objective response may provide information as to whether a treatment is likely to be successful. METHODS We conducted a systematic review of systemic agents that have proceeded to phase 3 trials in men with mCRPC and examined the relationship between improvements in measurable disease response in phase 2 trials and successful phase 3 trials leading to regulatory approval. Only trials that included men with radiographically measurable disease were included. RESULTS We examined 31 eligible mCRPC phase 3 trials between 1992 and 2017 and 29 of the preceding phase 2 trials for RECIST responses. Measurable tumor responses in phase 2 trials were higher for successful therapies in phase 3 trials in chemotherapy-naive men with mCRPC, but were less correlated with success in trials investigating docetaxel combination regimens or the post chemotherapy mCRPC setting. Many failed agents did not produce higher than expected response rates over control arms; however, several agents such as anti-angiogenic therapies or orteronel produced higher than expected responses without survival benefit. CONCLUSIONS Objective responses in men with mCRPC may be associated with prolonged survival, but this association is mechanism dependent and inconsistent across trials or disease states. These data support considering RECIST response as a supportive but not sole endpoint in phase 2 trials to support launching phase 3 trials.
Collapse
Affiliation(s)
- Landon C Brown
- Department of Medicine, School of Medicine, Duke University, Durham, NC, USA
| | | | - Andrew J Armstrong
- Department of Medicine, School of Medicine, Duke University, Durham, NC, USA. .,Duke Cancer Institute and the Duke Prostate and Urologic Cancer Center, Duke University, Durham, NC, USA.
| |
Collapse
|
209
|
Sumanasuriya S, De Bono J. Treatment of Advanced Prostate Cancer-A Review of Current Therapies and Future Promise. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a030635. [PMID: 29101113 DOI: 10.1101/cshperspect.a030635] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Despite many recent advances in the therapy for metastatic castration-resistant prostate cancer (mCRPC), the disease remains incurable, although men suffering from this disease are living considerably longer. In this review, we discuss the current treatment options available for this disease, such as taxane-based chemotherapy, the novel hormone therapies abiraterone and enzalutamide, and treatments such as radium-223 and sipuleucel-T. We also highlight the need for ongoing research in this field, because, despite numerous recent advances, the prognosis for mCRPC remains poor. Furthermore, as a growing body of evidence shows the increasing heterogeneity of the disease, and highlights the ongoing need for disease molecular stratification and validation/qualification of predictive biomarkers, we explore this burgeoning research space that is likely to transform how we treat this disease. We describe putative predictive biomarkers, including androgen receptor splice variants, phosphatase and tensin homolog (PTEN) loss, homologous recombination repair defects, including BRCA2 loss, and mismatch repair defects. The development of next-generation sequencing techniques and the routine biopsy of metastatic disease have driven significant advances in our understanding of the genomics of cancer, and are now poised to transform our treatment of this disease.
Collapse
Affiliation(s)
- Semini Sumanasuriya
- Division of Clinical Studies, The Institute of Cancer Research, Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, United Kingdom
| | - Johann De Bono
- Division of Clinical Studies, The Institute of Cancer Research, Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, United Kingdom
| |
Collapse
|
210
|
Kulkarni HR, Singh A, Langbein T, Schuchardt C, Mueller D, Zhang J, Lehmann C, Baum RP. Theranostics of prostate cancer: from molecular imaging to precision molecular radiotherapy targeting the prostate specific membrane antigen. Br J Radiol 2018; 91:20180308. [PMID: 29762048 DOI: 10.1259/bjr.20180308] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Alterations at the molecular level are a hallmark of cancer. Prostate cancer is associated with the overexpression of prostate-specific membrane antigen (PSMA) in a majority of cases, predominantly in advanced tumors, increasing with the grade or Gleason's score. PSMA can be selectively targeted using radiolabeled PSMA ligands. These small molecules binding the PSMA can be radiolabeled with γ-emitters like 99mTc and 111In or positron emitters like 68Ga and 18F for diagnosis as well as with their theranostic pairs such as 177Lu (β-emitter) or 225Ac (α-emitter) for therapy. This review summarizes the theranostic role of PSMA ligands for molecular imaging and targeted molecular radiotherapy, moving towards precision oncology.
Collapse
Affiliation(s)
- Harshad R Kulkarni
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Aviral Singh
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Thomas Langbein
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Christiane Schuchardt
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Dirk Mueller
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Jingjing Zhang
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Coline Lehmann
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Richard P Baum
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| |
Collapse
|
211
|
Okegawa T, Ninomiya N, Masuda K, Nakamura Y, Tambo M, Nutahara K. AR-V7 in circulating tumor cells cluster as a predictive biomarker of abiraterone acetate and enzalutamide treatment in castration-resistant prostate cancer patients. Prostate 2018; 78:576-582. [PMID: 29508425 DOI: 10.1002/pros.23501] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 02/12/2018] [Indexed: 11/10/2022]
Abstract
OBJECTIVE We examined whether androgen receptor splice variant 7 (AR-V7) in circulating tumor cell(CTC)clusters can be used to predict survival in patients with bone metastatic castration resistant-prostate cancer (mCRPC) treated with abiraterone or enzalutamide. METHODS We retrospectively enrolled 98 patients with CRPC on abiraterone or enzalutamide, and investigated the prognostic value of CTC cluster detection (+ v -) and AR-V7 detection (+ v -) using a CTC cluster detection - based AR-V7 mRNA assay. We examined ≤50% prostate-specific antigen (PSA) responses, PSA progression-free survival (PSA-PFS), clinical and radiological progression-free survival (radiologic PSF), and overall survival (OS). We then assessed whether AR-V7 expression in CTC clusters identified after On-chip multi-imaging flow cytometry was related to disease progression and survival after first-line systemic therapy. RESULTS All abiraterone-treated or enzalutamide-treated patients received prior docetaxel. The median follow-up was 20.7 (range: 3.0-37.0) months in the abiraterone and enzalutamide cohorts, respectively. Forty-nine of the 98 men (50.0%) were CTC cluster (-), 23 of the 98 men (23.5%) were CTC cluster(+)/AR-V7(-), and 26 of the 98 men (26.5%) were CTC cluster(+)/AR-V7(+). CTC cluster(+)/AR-V7(+) patients were more likely to have EOD ≥3 at diagnosis (P = 0.003), pain (P = 0.023), higher alkaline phosphatase levels (P < 0.001), and visceral metastases (P < 0.001). On multivariable analysis, pretherapy CTC cluster(+), CTC cluster(+)/AR-V7(-), and ALP >UNL were independently associated with a poor PSA-PFS, radiographic PFS, and OS in abiraterone-treated patients and enzalutamide-treated patients. CONCLUSION The CTC clusters and AR-V7-positive CTC clusters detected were important for assessing the response to abiraterone or enzalutamide therapy and for predicting disease outcome.
Collapse
Affiliation(s)
- Takatsugu Okegawa
- Department of Urology, The University of Kyorin, Mitaka, Tokyo, Japan
| | - Naoki Ninomiya
- Department of Urology, The University of Kyorin, Mitaka, Tokyo, Japan
| | - Kazuki Masuda
- Department of Urology, The University of Kyorin, Mitaka, Tokyo, Japan
| | - Yu Nakamura
- Department of Urology, The University of Kyorin, Mitaka, Tokyo, Japan
| | - Mitsuhiro Tambo
- Department of Urology, The University of Kyorin, Mitaka, Tokyo, Japan
| | - Kikuo Nutahara
- Department of Urology, The University of Kyorin, Mitaka, Tokyo, Japan
| |
Collapse
|
212
|
Zhang Y, Pitchiaya S, Cieślik M, Niknafs YS, Tien JCY, Hosono Y, Iyer MK, Yazdani S, Subramaniam S, Shukla SK, Jiang X, Wang L, Liu TY, Uhl M, Gawronski AR, Qiao Y, Xiao L, Dhanasekaran SM, Juckette KM, Kunju LP, Cao X, Patel U, Batish M, Shukla GC, Paulsen MT, Ljungman M, Jiang H, Mehra R, Backofen R, Sahinalp CS, Freier SM, Watt AT, Guo S, Wei JT, Feng FY, Malik R, Chinnaiyan AM. Analysis of the androgen receptor-regulated lncRNA landscape identifies a role for ARLNC1 in prostate cancer progression. Nat Genet 2018; 50:814-824. [PMID: 29808028 PMCID: PMC5980762 DOI: 10.1038/s41588-018-0120-1] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/23/2018] [Indexed: 12/23/2022]
Abstract
The androgen receptor (AR) plays a critical role in the development of the normal prostate as well as prostate cancer. Using an integrative transcriptomic analysis of prostate cancer cell lines and tissues, we identified ARLNC1 (AR-regulated long non-coding RNA 1) as an important long non-coding RNA that is strongly associated with AR signaling in prostate cancer progression. Not only was ARLNC1 induced by AR protein, ARLNC1 stabilized the AR transcript via RNA-RNA interaction. ARLNC1 knockdown suppressed AR expression, global AR signaling, and prostate cancer growth in vitro and in vivo. Taken together, these data support a role for ARLNC1 in maintaining a positive feedback loop that potentiates AR signaling during prostate cancer progression, and identifies ARLNC1 as a novel therapeutic target.
Collapse
Affiliation(s)
- Yajia Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Pathology, University of Michigan, Ann Arbor, MI, USA.,Molecular and Cellular Pathology Program, University of Michigan, Ann Arbor, MI, USA.,Department of Computational Medicine and Bioinformatics, Ann Arbor, MI, USA
| | - Sethuramasundaram Pitchiaya
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Marcin Cieślik
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yashar S Niknafs
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jean C-Y Tien
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yasuyuki Hosono
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew K Iyer
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Computational Medicine and Bioinformatics, Ann Arbor, MI, USA
| | - Sahr Yazdani
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Shruthi Subramaniam
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Sudhanshu K Shukla
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Biosciences and Bioengineering, Indian Institute of Technology Dharwad, Dharwad, India
| | - Xia Jiang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Lisha Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Tzu-Ying Liu
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Michael Uhl
- Department of Computer Science and Centre for Biological Signaling Studies (BIOSS), University of Freiburg, Freiburg, Germany
| | - Alexander R Gawronski
- School of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Pathology, University of Michigan, Ann Arbor, MI, USA.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Lanbo Xiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Saravana M Dhanasekaran
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Kristin M Juckette
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Lakshmi P Kunju
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Pathology, University of Michigan, Ann Arbor, MI, USA.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Utsav Patel
- New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Mona Batish
- New Jersey Medical School, Rutgers University, Newark, NJ, USA.,Department of Medical Laboratory Sciences, University of Delaware, Newark, DE, USA
| | - Girish C Shukla
- Department of Biological, Geological and Environmental Sciences, Center for Gene Regulation in Health and Disease, Cleveland State Univesity, Cleveland, OH, USA
| | - Michelle T Paulsen
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.,Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Mats Ljungman
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.,Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Hui Jiang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.,Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Rolf Backofen
- Department of Computer Science and Centre for Biological Signaling Studies (BIOSS), University of Freiburg, Freiburg, Germany
| | - Cenk S Sahinalp
- School of Informatics and Computing, Indiana University, Bloomington, IN, USA.,Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | | | | | | | - John T Wei
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Felix Y Feng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.,Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA.,Breast Oncology Program, University of Michigan, Ann Arbor, MI, USA.,Departments of Radiation Oncology, Urology, and Medicine, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA
| | - Rohit Malik
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Bristol-Myers Squibb, Princeton, NJ, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA. .,Department of Pathology, University of Michigan, Ann Arbor, MI, USA. .,Department of Computational Medicine and Bioinformatics, Ann Arbor, MI, USA. .,Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA. .,Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA. .,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA. .,Department of Urology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
213
|
Ban F, Dalal K, LeBlanc E, Morin H, Rennie PS, Cherkasov A. Cheminformatics Driven Development of Novel Therapies for Drug Resistant Prostate Cancer. Mol Inform 2018; 37:e1800043. [PMID: 29733509 DOI: 10.1002/minf.201800043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 04/18/2018] [Indexed: 11/07/2022]
Abstract
Androgen receptor (AR) is a master regulator of prostate cancer (PCa), and therefore is a pivotal drug target for the treatment of PCa including its castration-resistance form (CRPC). The development of acquired resistance is a major challenge in the use of the current antiandrogens. The recent advancements in inhibiting AR activity with small molecules specifically designed to target areas distinct from the receptor's androgen binding site are carefully discussed. Our new classes of AR inhibitors of AF2 and BF3 functional sites and DBD domains designed using cheminformatics techniques are promising to circumvent various AR-dependent resistance mechanisms.
Collapse
Affiliation(s)
- Fuqiang Ban
- Vancouver Prostate Centre (VPC), 2660 Oak Street, Vancouver, British Columbia, V6H3Z6, Canada
| | - Kush Dalal
- Vancouver Prostate Centre (VPC), 2660 Oak Street, Vancouver, British Columbia, V6H3Z6, Canada
| | - Eric LeBlanc
- Vancouver Prostate Centre (VPC), 2660 Oak Street, Vancouver, British Columbia, V6H3Z6, Canada
| | - Hélène Morin
- Vancouver Prostate Centre (VPC), 2660 Oak Street, Vancouver, British Columbia, V6H3Z6, Canada
| | - Paul S Rennie
- Vancouver Prostate Centre (VPC), 2660 Oak Street, Vancouver, British Columbia, V6H3Z6, Canada.,Department of Urology, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia, V6H 3Z6, Canada
| | - Artem Cherkasov
- Vancouver Prostate Centre (VPC), 2660 Oak Street, Vancouver, British Columbia, V6H3Z6, Canada.,Department of Urology, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia, V6H 3Z6, Canada
| |
Collapse
|
214
|
Crawford ED, Schellhammer PF, McLeod DG, Moul JW, Higano CS, Shore N, Denis L, Iversen P, Eisenberger MA, Labrie F. Androgen Receptor Targeted Treatments of Prostate Cancer: 35 Years of Progress with Antiandrogens. J Urol 2018; 200:956-966. [PMID: 29730201 DOI: 10.1016/j.juro.2018.04.083] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2018] [Indexed: 11/25/2022]
Abstract
PURPOSE Antiandrogens inhibit the androgen receptor and have an important role in the treatment of prostate cancer. This review provides a historical perspective on the development and clinical benefit of antiandrogens in the treatment of prostate cancer. MATERIALS AND METHODS We searched PubMed® for clinical trials with the search terms antiandrogens and prostate cancer combined with drug names for antiandrogens. This article represents a collaboration of clinical investigators who have made critical scientific contributions leading to the approval of antiandrogens for treating patients with prostate cancer. RESULTS Antiandrogens differ in chemical structure and exert varying efficacy and safety profiles. The unfavorable therapeutic index of steroidal antiandrogens led to replacement by safer nonsteroidal agents. Flutamide, nilutamide and bicalutamide, which were designed to target the androgen receptor, were developed primarily for use in combination with castration to provide combined androgen blockade. Modest clinical benefits were observed with the combination of first generation antiandrogens and castration vs castration alone. With increased knowledge of androgen receptor structure and its biological functions a new generation of antiandrogens without agonist activity was designed to provide more potent inhibition of the androgen receptor. Randomized clinical trials in patients with metastatic, castration resistant prostate cancer showed significant survival benefits, which led to the approval of enzalutamide in August 2012. Apalutamide was recently approved while darolutamide is not yet approved in the United States. These next generation antiandrogens are being actively tested in earlier disease states such as nonmetastatic prostate cancer. Evolving knowledge of resistance mechanisms to androgen receptor targeted treatments will stimulate research and drug discovery for additional compounds. Further testing in nonmetastatic castration resistant prostate cancer as well as castration sensitive disease states will hopefully augment our ability to treat a broader spectrum of patients with prostate cancer. CONCLUSIONS Antiandrogens have already provided important benefits for prostate cancer treatment. Greater knowledge about the structural and functional biology of the androgen receptor in prostate cancer will facilitate further discovery and development of further improved antiandrogens with enhanced clinical activity in patients with advanced metastatic disease. Testing these new agents earlier in the course of prostate cancer may further improve the survival and quality of life of patients with current local and/or systemic treatment modalities.
Collapse
Affiliation(s)
| | | | - David G McLeod
- Center for Prostate Disease Research, Uniformed Services University of the Health Sciences, Bethesda
| | - Judd W Moul
- Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Celestia S Higano
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, South Carolina
| | - Louis Denis
- Europa Uomo, Oncology Centre Antwerp, Antwerp, Belgium
| | - Peter Iversen
- Copenhagen Prostate Cancer Center, University of Copenhagen, Copenhagen, Denmark
| | - Mario A Eisenberger
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | | |
Collapse
|
215
|
Wu W, Karelia D, Pramanik K, Amin SG, Sharma AK, Jiang C, Lu J. Phenylbutyl isoselenocyanate induces reactive oxygen species to inhibit androgen receptor and to initiate p53-mediated apoptosis in LNCaP prostate cancer cells. Mol Carcinog 2018; 57:1055-1066. [DOI: 10.1002/mc.22825] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/09/2018] [Accepted: 04/14/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Wei Wu
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
| | - Deepkamal Karelia
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
| | - Kartick Pramanik
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
| | - Shantu G. Amin
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
- Penn State Cancer Institute; Pennsylvania State College of Medicine; Hershey Pennsylvania
| | - Arun K. Sharma
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
- Penn State Cancer Institute; Pennsylvania State College of Medicine; Hershey Pennsylvania
| | - Cheng Jiang
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
| | - Junxuan Lu
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
- Penn State Cancer Institute; Pennsylvania State College of Medicine; Hershey Pennsylvania
| |
Collapse
|
216
|
Qin X, Fang L, Zhao J, Gou S. Theranostic Pt(IV) Conjugate with Target Selectivity for Androgen Receptor. Inorg Chem 2018; 57:5019-5029. [DOI: 10.1021/acs.inorgchem.8b00083] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
217
|
Guo J, Hu J, Cao R, Chen Q, Li K. Androgen Receptor Is Inactivated and Degraded in Bladder Cancer Cells by Phenyl Glucosamine via miR-449a Restoration. Med Sci Monit 2018; 24:2294-2301. [PMID: 29659560 PMCID: PMC5918918 DOI: 10.12659/msm.906836] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Bladder cancer caused by exposure to aniline dyes, chronic cystitis, and smoking is detected in approximately 70 000 new cases annually. In the USA alone, it leads to 15 000 deaths every year. In the present study, we investigated the role of 3-((4′-amino-[1,1′-biphenyl]-4-yl)amino)-4-bromo-5-oxo-2,5-dihydrofuran-2-yl acetate (ABDHFA) in the inhibition of bladder cancer cell viability. Material/Methods Viability of cells was examined using MTT assay and distribution of cell cycle was assessed by flow cytometry. Expression of cyclin D1, androgen, prostate-specific antigen (PSA), and miR-449a was analyzed using Western blot and quantitative real-time polymerase chain reaction assays. Results The results demonstrated that ABDHFA treatment inhibited viability of UMUC3 and TCCSUP AR-positive bladder cancer cells. ABDHFA treatment led to break-down of AR in UMUC3 and TCCSUP cells after 48 h in a dose-dependent manner. Up-regulation of miR-449a by lentivirus transfection down-regulated the AR signalling pathway. In UMUC3 and TCCSUP cells, ABDHFA treatment led to inhibition of mRNA and protein expression corresponding to AR. Conclusions In summary, the present study demonstrates that proliferation of AR-positive bladder carcinoma cells is markedly reduced by ABDHFA treatment through arrest of cell cycle and degradation of AR protein. Thus, ABDHFA, a novel compound, can be used for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Ju Guo
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Jieping Hu
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Runfu Cao
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Qingsheng Chen
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Kanghua Li
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| |
Collapse
|
218
|
Thangavel C, Perepelyuk M, Boopathi E, Liu Y, Polischak S, Deshpande DA, Rafiq K, Dicker AP, Knudsen KE, Shoyele SA, Den RB. Improvement in Therapeutic Efficacy and Reduction in Cellular Toxicity: Introduction of a Novel Anti-PSMA-Conjugated Hybrid Antiandrogen Nanoparticle. Mol Pharm 2018; 15:1778-1790. [PMID: 29616555 DOI: 10.1021/acs.molpharmaceut.7b01024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Second generation antiandrogens have improved overall survival for men with metastatic castrate resistant prostate cancer; however, the antiandrogens result in suppression of androgen receptor (AR) activity in all tissues resulting in dose limiting toxicity. We sought to overcome this limitation through encapsulation in a prostate specific membrane antigen (PSMA)-conjugated nanoparticle. We designed and characterized a novel nanoparticle containing an antiandrogen, enzalutamide. Selectivity and enhanced efficacy was achieved through coating the particle with PSMA. The PSMA-conjugated nanoparticle was internalized selectively in AR expressing prostate cancer cells. It did not elicit an inflammatory effect. The efficacy of enzalutamide was not compromised through insertion into the nanoparticle; in fact, lower systemic drug concentrations of enzalutamide resulted in comparable clinical activity. Normal muscle cells were not impacted by the PSMA-conjugated containing antiandrogen. This approach represents a novel strategy to increase the specificity and effectiveness of antiandrogen treatment for men with castrate resistant prostate cancer. The ability to deliver higher drug concentrations in prostate cancer cells may translate into improved clinical end points including overall survival.
Collapse
|
219
|
Bremmer F, Jarry H, Unterkircher V, Kaulfuss S, Burfeind P, Radzun HJ, Ströbel P, Thelen P. Testosterone metabolites inhibit proliferation of castration- and therapy-resistant prostate cancer. Oncotarget 2018; 9:16951-16961. [PMID: 29682196 PMCID: PMC5908297 DOI: 10.18632/oncotarget.24763] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/27/2018] [Indexed: 01/16/2023] Open
Abstract
Novel treatments for castration-resistant prostate cancer (CRPC) such as abiraterone acetate (AA) or enzalutamide effectively target the androgen pathway to arrest aberrant signalling and cell proliferation. Testosterone is able to inhibit tumour cell growth in CRPC. Estrogen receptor-beta (ERβ) binds the testosterone-metabolites 3β-androstanediol and 3α-androstanediol in parallel to the canonical estradiol. In the prostate it is widely accepted that ERβ regulates estrogen signalling, mediating anti-proliferative effects. We used the prostate cancer cell lines LNCaP, PC-3, VCaP, and the non-neoplastic BPH-1. VCaP cells were treated with 1 nmol/L testosterone over 20 passages, yielding the cell line VCaPrev, sensitive to hormone therapies. In contrast, LNCaP cells were grown for more than 100 passages yielding a high passage therapy resistant cell line (hiPLNCaP). VCaP and hiPLNCaP cell lines were treated with 5 μmol/L AA for more than 20 passages, respectively, generating the AA-tolerant-subtypes VCaPAA and hiPLNCaPAA. Cell lines were treated with testosterone, dihydrotestosterone (DHT), R1881, and the androgen-metabolites 3β-androstanediol and 3α-androstanediol. 3β-androstanediol or 3α-androstanediol significantly reduced proliferation in all cell lines except the BPH-1 and androgen receptor-negative PC-3 and markedly downregulated AR and estrogen receptor alpha (ERα). Whereas ERβ expression was increased in all cell lines except BPH-1 or PC-3. In summary, 3β-adiol or 3α-adiol, as well as DHT and R1881, significantly reduced tumour cell growth in CRPC cells. Thus, these compounds represent novel potential therapeutic approaches to overcome drug-resistance in CRPC, especially with regard to AR-V7 function in therapy resistance. Furthermore, these data confirm the tumour suppressor properties of ERβ in CRPC.
Collapse
Affiliation(s)
- Felix Bremmer
- Institute of Pathology, University Medical Center, Göttingen 37075, Germany
| | - Hubertus Jarry
- Department of Experimental Endocrinology, University Medical Center, Göttingen 37075, Germany
| | | | - Silke Kaulfuss
- Institute of Human Genetics, University Medical Center, Göttingen 37073, Germany
| | - Peter Burfeind
- Institute of Human Genetics, University Medical Center, Göttingen 37073, Germany
| | | | - Philipp Ströbel
- Institute of Pathology, University Medical Center, Göttingen 37075, Germany
| | - Paul Thelen
- Department of Urology, University Medical Center, Göttingen 37075, Germany
| |
Collapse
|
220
|
Burvenich IJG, Parakh S, Parslow AC, Lee ST, Gan HK, Scott AM. Receptor Occupancy Imaging Studies in Oncology Drug Development. AAPS JOURNAL 2018. [DOI: 10.1208/s12248-018-0203-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
221
|
Logan IR, McClurg UL, Jones DL, O'Neill DJ, Shaheen FS, Lunec J, Gaughan L, Robson CN. Nutlin-3 inhibits androgen receptor-driven c-FLIP expression, resulting in apoptosis of prostate cancer cells. Oncotarget 2018; 7:74724-74733. [PMID: 27729622 PMCID: PMC5342697 DOI: 10.18632/oncotarget.12542] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 09/26/2016] [Indexed: 01/10/2023] Open
Abstract
Inhibition of androgen receptor (AR) signalling represents the conventional medical management of prostate cancer. Ultimately this treatment fails because tumors develop an incurable, castrate resistant phenotype, resulting in an unmet need for new treatments in prostate cancer. The AR remains a viable therapeutic target in castrate resistant disease, such that novel ways of downregulating AR activities are attractive as potential treatments. Here we describe a mechanism by which the AR can be downregulated by the MDM2 antagonist Nutlin-3, resulting in loss of pro-survival c-FLIP gene expression and apoptosis. We additionally show that loss of c-FLIP sensitises prostate cancer cells to Nutlin-3. Finally, we demonstrate that the unrelated MDM2 antagonist Mi-63 also impinges upon AR signalling, supporting the concept of future treatment of prostate cancer with MDM2 antagonists.
Collapse
Affiliation(s)
- Ian R Logan
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, Tyne and Wear, United Kingdom
| | - Urszula L McClurg
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, Tyne and Wear, United Kingdom
| | - Dominic L Jones
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, Tyne and Wear, United Kingdom
| | - Daniel J O'Neill
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, Tyne and Wear, United Kingdom
| | - Fadhel S Shaheen
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, Tyne and Wear, United Kingdom
| | - John Lunec
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, Tyne and Wear, United Kingdom
| | - Luke Gaughan
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, Tyne and Wear, United Kingdom
| | - Craig N Robson
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, Tyne and Wear, United Kingdom
| |
Collapse
|
222
|
Ito Y, Sadar MD. Enzalutamide and blocking androgen receptor in advanced prostate cancer: lessons learnt from the history of drug development of antiandrogens. Res Rep Urol 2018; 10:23-32. [PMID: 29497605 PMCID: PMC5818862 DOI: 10.2147/rru.s157116] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Enzalutamide is a nonsteroidal antiandrogen for the treatment of metastatic castration-resistant prostate cancer (mCRPC) both before and after chemotherapy. Enzalutamide is more effective than its predecessor bicalutamide, which was analyzed in head-to-head studies of patients with CRPC. This family of nonsteroidal antiandrogens is now comprised of four drugs approved by the US Food and Drug Administration with two investigational drugs in clinical trials. Antiandrogens have been employed clinically for more than five decades to provide a rich resource of information. Steady-state concentration minimums (Cmin or trough) in the range of ~1–13 μg/mL are measured in patients at therapeutic doses. Interestingly, enzalutamide which is considered to have strong affinity for the androgen receptor (AR) requires Cmin levels >10 μg/mL. The sequence of antiandrogens and the clinical order of application in regard to other drugs that target the androgen axis remain of high interest. One novel first-in-class drug, called ralaniten, which binds to a unique region in the N-terminus domain of both the full-length and the truncated constitutively active splice variants of the AR, is currently in clinical trials for patients who previously received abiraterone, enzalutamide, or both. This highlights the trend to develop drugs with novel mechanisms of action and potentially differing mechanisms of resistance compared with antiandrogens. Better and more complete inhibition of the transcriptional activity of the AR appears to continue to provide improvements in the clinical management of mCRPC.
Collapse
Affiliation(s)
- Yusuke Ito
- Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | | |
Collapse
|
223
|
Hong JH. Pharmacokinetic/pharmacodynamic drug evaluation of enzalutamide for treating prostate cancer. Expert Opin Drug Metab Toxicol 2018; 14:361-369. [PMID: 29431540 DOI: 10.1080/17425255.2018.1440288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Enzalutamide is the first approved second-generation androgen receptor (AR) antagonist in the treatment of metastatic castration-resistant prostate cancer (mCRPC) with or without docetaxel-based chemotherapy. Over the past 5 years, a number of attempts were made to determine the efficacy of enzalutamide in the different clinical settings. Areas covered: A literature search was performed at the PubMed, Embase, and Web of Science database to collect the most relevant and impactful studies, including basic science investigations, clinical trials, and reviews. This article focuses on the pharmacology, efficacy, tolerability, and future perspective of enzalutamide. Expert opinion: The treatment paradigm of CRPC has been dramatically challenged of late. Enzalutamide are in wide use because of its favorable efficacy and safety, but primary or acquired resistance to the drug will eventually develop. Further studies are thus necessary to identify appropriate patients who can achieve apparent benefits from enzalutamide alone or in combination with other drugs.
Collapse
Affiliation(s)
- Jeong Hee Hong
- a Department of Urology , Dankook University College of Medicine , Cheonan , Republic of Korea
| |
Collapse
|
224
|
Abstract
Prostate cancer is a common malignancy with various treatments from surveillance, surgery, radiation and chemotherapy. The institution of appropriate, effective treatment relies in part on accurate imaging. Molecular imaging techniques offer an opportunity for increased timely detection of prostate cancer, its recurrence, as well as metastatic disease. Advancements within the field of molecular imaging have been complex with some agents targeting receptors and others acting as metabolic intermediaries. In this article, we provide an overview of the most clinically relevant radiotracers to date based on a combination of the five states model and the National Comprehensive Cancer Network Guidelines.
Collapse
Affiliation(s)
- Anne Marie Boustani
- 1 Department of Radiology and Biomedical Imaging, Yale University School of Medicine , New Haven, CT , USA
| | - Darko Pucar
- 1 Department of Radiology and Biomedical Imaging, Yale University School of Medicine , New Haven, CT , USA
| | - Lawrence Saperstein
- 1 Department of Radiology and Biomedical Imaging, Yale University School of Medicine , New Haven, CT , USA
| |
Collapse
|
225
|
Maximov PY, Abderrahman B, Curpan RF, Hawsawi YM, Fan P, Jordan VC. A unifying biology of sex steroid-induced apoptosis in prostate and breast cancers. Endocr Relat Cancer 2018; 25:R83-R113. [PMID: 29162647 PMCID: PMC5771961 DOI: 10.1530/erc-17-0416] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 11/21/2017] [Indexed: 12/13/2022]
Abstract
Prostate and breast cancer are the two cancers with the highest incidence in men and women, respectively. Here, we focus on the known biology of acquired resistance to antihormone therapy of prostate and breast cancer and compare laboratory and clinical similarities in the evolution of the disease. Laboratory studies and clinical observations in prostate and breast cancer demonstrate that cell selection pathways occur during acquired resistance to antihormonal therapy. Following sex steroid deprivation, both prostate and breast cancer models show an initial increased acquired sensitivity to the growth potential of sex steroids. Subsequently, prostate and breast cancer cells either become dependent upon the antihormone treatment or grow spontaneously in the absence of hormones. Paradoxically, the physiologic sex steroids now kill a proportion of selected, but vulnerable, resistant tumor cells. The sex steroid receptor complex triggers apoptosis. We draw parallels between acquired resistance in prostate and breast cancer to sex steroid deprivation. Clinical observations and patient trials confirm the veracity of the laboratory studies. We consider therapeutic strategies to increase response rates in clinical trials of metastatic disease that can subsequently be applied as a preemptive salvage adjuvant therapy. The goal of future advances is to enhance response rates and deploy a safe strategy earlier in the treatment plan to save lives. The introduction of a simple evidence-based enhanced adjuvant therapy as a global healthcare strategy has the potential to control recurrence, reduce hospitalization, reduce healthcare costs and maintain a healthier population that contributes to society.
Collapse
Affiliation(s)
- Philipp Y Maximov
- Department of Breast Medical OncologyMD Anderson Cancer Centre, Houston, Texas, USA
| | - Balkees Abderrahman
- Department of Breast Medical OncologyMD Anderson Cancer Centre, Houston, Texas, USA
| | | | - Yousef M Hawsawi
- Department of GeneticsKing Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Ping Fan
- Department of Breast Medical OncologyMD Anderson Cancer Centre, Houston, Texas, USA
| | - V Craig Jordan
- Department of Breast Medical OncologyMD Anderson Cancer Centre, Houston, Texas, USA
| |
Collapse
|
226
|
Dysregulated fibronectin trafficking by Hsp90 inhibition restricts prostate cancer cell invasion. Sci Rep 2018; 8:2090. [PMID: 29391407 PMCID: PMC5794796 DOI: 10.1038/s41598-018-19871-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/08/2018] [Indexed: 12/17/2022] Open
Abstract
The molecular chaperone Hsp90 is overexpressed in prostate cancer (PCa) and is responsible for the folding, stabilization and maturation of multiple oncoproteins, which are implicated in PCa progression. Compared to first-in-class Hsp90 inhibitors such as 17-allylamino-demethoxygeldanamycin (17-AAG) that were clinically ineffective, second generation inhibitor AUY922 has greater solubility and efficacy. Here, transcriptomic and proteomic analyses of patient-derived PCa explants identified cytoskeletal organization as highly enriched with AUY922 treatment. Validation in PCa cell lines revealed that AUY922 caused marked alterations to cell morphology, and suppressed cell motility and invasion compared to vehicle or 17-AAG, concomitant with dysregulation of key extracellular matrix proteins such as fibronectin (FN1). Interestingly, while the expression of FN1 was increased by AUY922, FN1 secretion was significantly decreased. This resulted in cytosolic accumulation of FN1 protein within late endosomes, suggesting that AUY922 disrupts vesicular secretory trafficking pathways. Depletion of FN1 by siRNA knockdown markedly reduced the invasive capacity of PCa cells, phenocopying AUY922. These results highlight a novel mechanism of action for AUY922 beyond its established effects on cellular mitosis and survival and, furthermore, identifies extracellular matrix cargo delivery as a potential therapeutic target for the treatment of aggressive PCa.
Collapse
|
227
|
Dharmani C, Bonafede M, Krivoshik A. Risk Factors for and Incidence of Seizures in Metastatic Castration-Resistant Prostate Cancer: A Real-World Retrospective Cohort Study. Clin Drug Investig 2018; 37:1183-1190. [PMID: 29027610 DOI: 10.1007/s40261-017-0578-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND OBJECTIVE This real-world study assessed the prevalence, risk factors for, and incidence of seizures in patients with metastatic castration-resistant prostate cancer (mCRPC). METHODS Patients with mCRPC were selected from MarketScan Commercial and Medicare Supplemental Databases between 1 January 2009 and 31 July 2012. Prevalence of seizure risk factors were described separately and in combination with other risk factors. Seizure incidence was calculated overall and for each risk factor group. RESULTS The most common risk factors were history of seizure threshold-lowering medication use (35%), history of loss of consciousness (6%), history of transient ischemic attack or cerebrovascular accident (2%), treated brain metastasis (0.9%), history of seizure (0.6%), and dementia (0.5%). Overall, seizure incidence was 1.8 per 100 person-years (PYs) (95% confidence interval [CI] 1.5-2.1), being higher among patients with at least one risk factor (2.8 per 100 PYs; 95% CI 2.2-3.4) than those without risk factors (1.2 per 100 PYs; 95% CI 1.0-1.6). Seizure incidence was highest among a few patients (0.6%) with a history of seizure (82.0 per 100 PYs; 95% CI 45.9-135.2) and within this small subpopulation, higher among those with a history of anticonvulsant use (120.9 per 100 PYs; 95% CI 60.3-216.3) than without anticonvulsant use (43.5 per 100 PYs; 95% CI 11.9-111.3). CONCLUSION History of seizure is an important risk factor for seizure occurrence in patients with mCRPC, particularly in those with a history of anticonvulsant use. These findings improve understanding of the risk of seizure occurrence in patients with mCRPC, who are potential users of androgen receptor antagonists, including enzalutamide.
Collapse
Affiliation(s)
- Charles Dharmani
- Astellas Pharma, Inc., 1 Astellas Way, Northbrook, IL, 60062, USA.
| | | | - Andrew Krivoshik
- Astellas Pharma, Inc., 1 Astellas Way, Northbrook, IL, 60062, USA
| |
Collapse
|
228
|
Waaijer SJ, Kok IC, Eisses B, Schröder CP, Jalving M, Brouwers AH, Lub-de Hooge MN, de Vries EG. Molecular Imaging in Cancer Drug Development. J Nucl Med 2018; 59:726-732. [DOI: 10.2967/jnumed.116.188045] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/16/2018] [Indexed: 12/23/2022] Open
|
229
|
Xie J, Mølck C, Paquet-Fifield S, Butler L, Sloan E, Ventura S, Hollande F. High expression of TROP2 characterizes different cell subpopulations in androgen-sensitive and androgen-independent prostate cancer cells. Oncotarget 2018; 7:44492-44504. [PMID: 27283984 PMCID: PMC5190113 DOI: 10.18632/oncotarget.9876] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/19/2016] [Indexed: 12/15/2022] Open
Abstract
Progression of castration-resistant tumors is frequent in prostate cancer. Current systemic treatments for castration-resistant prostate cancer only produce modest increases in survival time and self-renewing Tumor-Initiating Cells (TICs) are suspected to play an important role in resistance to these treatments. However it remains unclear whether the same TICs display both chemo-resistance and self-renewing abilities throughout progression from early stage lesions to late, castration resistant tumors. Here, we found that treatment of mice bearing LNCaP-derived xenograft tumors with cytotoxic (docetaxel) and anti-androgen (flutamide) compounds enriched for cells that express TROP2, a putative TIC marker. Consistent with a tumor-initiating role, TROP2high cells from androgen-sensitive prostate cancer cell lines displayed an enhanced ability to re-grow in culture following treatment with taxane-based chemotherapy with or without androgen blockade. TROP2 down-regulation in these cells reduced their ability to recur after treatment with docetaxel, in the presence or absence of flutamide. Accordingly, in silico analysis of published clinical data revealed that prostate cancer patients with poor prognosis exhibit significantly elevated TROP2 expression level compared to low-risk patients, particularly in the case of patients diagnosed with early stage tumors. In contrast, in androgen-independent prostate cancer cell lines, TROP2high cells did not exhibit a differential treatment response but were characterized by their high self-renewal ability. Based on these findings we propose that high TROP2 expression identifies distinct cell sub-populations in androgen-sensitive and androgen-independent prostate tumors and that it may be a predictive biomarker for prostate cancer treatment response in androgen-sensitive tumors.
Collapse
Affiliation(s)
- Jinhan Xie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia.,Current address: Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Kensington, Australia
| | - Christina Mølck
- Department of Pathology, The University of Melbourne, Parkville, Australia
| | | | - Lisa Butler
- School of Medicine, South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | | | - Erica Sloan
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia.,Cousins Center for PNI, UCLA Semel Institute, Jonsson Comprehensive Cancer Center, and UCLA AIDS Institute, University of California Los Angeles, Los Angeles, CA, USA.,Peter MacCallum Cancer Centre, Division of Cancer Surgery, East Melbourne, Victoria, Australia
| | - Sabatino Ventura
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Frédéric Hollande
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia.,Department of Pathology, The University of Melbourne, Parkville, Australia
| |
Collapse
|
230
|
Systemic Treatment of Castration-Resistant Metastatic Prostate Cancer. Urol Oncol 2018. [DOI: 10.1007/978-3-319-42603-7_76-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
231
|
Thorek DLJ, Watson PA, Lee SG, Ku AT, Bournazos S, Braun K, Kim K, Sjöström K, Doran MG, Lamminmäki U, Santos E, Veach D, Turkekul M, Casey E, Lewis JS, Abou DS, van Voss MRH, Scardino PT, Strand SE, Alpaugh ML, Scher HI, Lilja H, Larson SM, Ulmert D. Internalization of secreted antigen-targeted antibodies by the neonatal Fc receptor for precision imaging of the androgen receptor axis. Sci Transl Med 2017; 8:367ra167. [PMID: 27903863 DOI: 10.1126/scitranslmed.aaf2335] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 07/14/2016] [Accepted: 09/13/2016] [Indexed: 12/13/2022]
Abstract
Targeting the androgen receptor (AR) pathway prolongs survival in patients with prostate cancer, but resistance rapidly develops. Understanding this resistance is confounded by a lack of noninvasive means to assess AR activity in vivo. We report intracellular accumulation of a secreted antigen-targeted antibody (SATA) that can be used to characterize disease, guide therapy, and monitor response. AR-regulated human kallikrein-related peptidase 2 (free hK2) is a prostate tissue-specific antigen produced in prostate cancer and androgen-stimulated breast cancer cells. Fluorescent and radio conjugates of 11B6, an antibody targeting free hK2, are internalized and noninvasively report AR pathway activity in metastatic and genetically engineered models of cancer development and treatment. Uptake is mediated by a mechanism involving the neonatal Fc receptor. Humanized 11B6, which has undergone toxicological tests in nonhuman primates, has the potential to improve patient management in these cancers. Furthermore, cell-specific SATA uptake may have a broader use for molecularly guided diagnosis and therapy in other cancers.
Collapse
Affiliation(s)
- Daniel L J Thorek
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Radiological Science, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.,Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Philip A Watson
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sang-Gyu Lee
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anson T Ku
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stylianos Bournazos
- Leonard Wagner Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA
| | - Katharina Braun
- Department of Urology, University Hospital of the Ruhr-University of Bochum, Marien Hospital Herne, Herne, Germany
| | - Kwanghee Kim
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Michael G Doran
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Urpo Lamminmäki
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Elmer Santos
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Darren Veach
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mesruh Turkekul
- Molecular Cytology Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Emily Casey
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jason S Lewis
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Radiochemistry and Imaging Sciences Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Diane S Abou
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Radiological Science, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Marise R H van Voss
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Radiological Science, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.,Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Peter T Scardino
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Urology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Sven-Erik Strand
- Department of Medical Radiation Physics, Lund University, Lund, Sweden
| | - Mary L Alpaugh
- Departments of Biology and Biomedical and Translational Sciences, Rowan University, Glassboro, NJ 08028, USA
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Hans Lilja
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. .,Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Nuffield Department of Surgical Sciences, University of Oxford, Oxford, U.K.,Department of Laboratory Medicine, Lund University, Malmö, Sweden
| | - Steven M Larson
- Nuclear Medicine Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. .,Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - David Ulmert
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. .,Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Division of Urological Research, Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
232
|
Heller G, McCormack R, Kheoh T, Molina A, Smith MR, Dreicer R, Saad F, de Wit R, Aftab DT, Hirmand M, Limon A, Fizazi K, Fleisher M, de Bono JS, Scher HI. Circulating Tumor Cell Number as a Response Measure of Prolonged Survival for Metastatic Castration-Resistant Prostate Cancer: A Comparison With Prostate-Specific Antigen Across Five Randomized Phase III Clinical Trials. J Clin Oncol 2017; 36:572-580. [PMID: 29272162 DOI: 10.1200/jco.2017.75.2998] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose Measures of response that are clinically meaningful and occur early are an unmet need in metastatic castration-resistant prostate cancer clinical research and practice. We explored, using individual patient data, week 13 circulating tumor cell (CTC) and prostate-specific antigen (PSA) response end points in five prospective randomized phase III trials that enrolled a total of 6,081 patients-COU-AA-301, AFFIRM, ELM-PC-5, ELM-PC-4, and COMET-1- ClinicalTrials.Gov identifiers: NCT00638690, NCT00974311, NCT01193257, NCT01193244, and NCT01605227, respectively. Methods Eight response end points were explored. CTC nonzero at baseline and 0 at 13 weeks (CTC0); CTC conversion (≥ 5 CTCs at baseline, ≤ 4 at 13 weeks-the US Food and Drug Administration cleared response measure); a 30%, 50%, and 70% decrease in CTC count; and a 30%, 50%, and 70% decrease in PSA level. Patients missing week-13 values were considered nonresponders. The discriminatory strength of each end point with respect to overall survival in each trial was assessed using the weighted c-index. Results Of the eight response end points, CTC0 and CTC conversion had the highest weighted c-indices, with smaller standard deviations. For CTC0, the mean (standard deviation) was 0.81 (0.04); for CTC conversion, 0.79 (0.03); for 30% decrease in CTC count, 0.72 (0.06); for 50% decrease in CTC count, 0.72 (0.06); for 70% decrease in CTC count, 0.73 (0.05); for 30% decrease in PSA level, 0.71 (0.03); for 50% decrease in PSA level, 0.72 (0.06); and for 70% decrease in PSA level, 0.74 (0.05). Seventy-five percent of eligible patients could be evaluated with the CTC0 end point, compared with 51% with the CTC conversion end point. Conclusion The CTC0 and CTC conversion end points had the highest discriminatory power for overall survival. Both are robust and meaningful response end points for early-phase metastatic castration-resistant prostate cancer clinical trials. CTC0 is applicable to a significantly higher percentage of patients than CTC conversion.
Collapse
Affiliation(s)
- Glenn Heller
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Robert McCormack
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Thian Kheoh
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Arturo Molina
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Matthew R Smith
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Robert Dreicer
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Fred Saad
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Ronald de Wit
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Dana T Aftab
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Mohammad Hirmand
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Ana Limon
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Karim Fizazi
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Martin Fleisher
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Johann S de Bono
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Howard I Scher
- Glenn Heller, Martin Fleisher, and Howard I. Scher, Memorial Sloan Kettering Cancer Center; Howard I. Scher, Weill Cornell Medical College, New York, NY; Robert McCormack, Janssen Research & Development, Raritan, NJ; Thian Kheoh and Arturo Molina, Janssen Research & Development, Los Angeles; Dana T. Aftab, Exelixis, South San Francisco; Mohammad Hirmand, Medivation, San Francisco, CA; Matthew R. Smith, Massachusetts General Hospital, Boston; Ana Limon, Takeda Oncology, Cambridge, MA; Robert Dreicer, University of Virginia Medical School, Charlottesville, VA; Fred Saad, University of Montreal, Montreal, Quebec, Canada; Ronald de Wit, Erasmus Medical Center, Rotterdam, the Netherlands; Karim Fizazi, Institut Gustave Roussy, Villejuif, and University of Paris Sud, Orsay, France; and Johann S. de Bono, The Institute of Cancer Research, and The Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| |
Collapse
|
233
|
Kivinummi K, Urbanucci A, Leinonen K, Tammela TLJ, Annala M, Isaacs WB, Bova GS, Nykter M, Visakorpi T. The expression of AURKA is androgen regulated in castration-resistant prostate cancer. Sci Rep 2017; 7:17978. [PMID: 29269934 PMCID: PMC5740165 DOI: 10.1038/s41598-017-18210-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 12/06/2017] [Indexed: 11/13/2022] Open
Abstract
Although second generation endocrine therapies have significantly improved survival, castration-resistant prostate cancer (CRPC) cells are eventually able to escape available hormonal treatments due to reactivation of androgen receptor (AR) signaling. Identification of novel, non-classical and druggable AR-target genes may provide new approaches to treat CRPC. Our previous analyses suggested that Aurora kinase A (AURKA) is regulated by androgens in prostate cancer cells that express high levels of AR. Here, we provide further evidence that AURKA is significantly overexpressed in AR-positive CRPC samples carrying amplification of AR gene and/or expressing AR in high levels. We also demonstrate androgen-induced AR binding in the intronic region of AURKA. The expression of AURKA is increased upon androgen stimulation in LNCaP-ARhi cells that express high levels of AR. The growth of the cells was also significantly inhibited by an AURKA specific inhibitor, alisertib (MLN8237). Together, these findings suggest that the expression of AURKA is regulated by androgen in prostate cancer cells that highly express AR, emphasizing its potential as a therapeutic target in patients with CRPC.
Collapse
Affiliation(s)
- Kati Kivinummi
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland.
| | - Alfonso Urbanucci
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland.,Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forksningsparken, University of Oslo, Oslo, Norway.,Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,K.G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | - Katri Leinonen
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Teuvo L J Tammela
- Department of Urology, Tampere University Hospital and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Matti Annala
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - William B Isaacs
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - G Steven Bova
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Matti Nykter
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Tapio Visakorpi
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
234
|
Liu CY, Lau KY, Hsu CC, Chen JL, Lee CH, Huang TT, Chen YT, Huang CT, Lin PH, Tseng LM. Combination of palbociclib with enzalutamide shows in vitro activity in RB proficient and androgen receptor positive triple negative breast cancer cells. PLoS One 2017; 12:e0189007. [PMID: 29261702 PMCID: PMC5737960 DOI: 10.1371/journal.pone.0189007] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 11/16/2017] [Indexed: 11/29/2022] Open
Abstract
Objectives Triple negative breast cancer (TNBC) lacks specific drug targets and remains challenging. Palbociclib, a cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitor is approved for metastatic estrogen receptor (ER)-positive and human epithermal growth factor 2 (HER2)-negative breast cancer. The nature of cell cycle inhibition by palbociclib suggests its potential in TNBC cells. Retinoblastoma (RB, a known substrate of CDK4/6) pathway deregulation is a frequent occurrence in TNBC and studies have revealed that pharmacological CDK4/6 inhibition induces a cooperative cytostatic effect with doxorubicin in RB-proficient TNBC models. In addition, recent studies reported that anti-androgen therapy shows preclinical efficacy in androgen-receptor (AR)-positive TNBC cells. Here we examined the effect of palbociclib in combination with an anti-androgen enzalutamide in TNBC cells. Method MDA-MB-453, BT-549, MDA-MB-231 and MDA-MB-468 TNBC cell lines were used for in vitro studies. Protein expressions were assessed by Western blot analysis. Cytostatic effect was examined by MTT assay. Cell cycle and apoptosis were examined by flow cytometry. Results Palbociclib showed inhibitory effect in RB-proficient TNBC cells, and enzalutamide inhibited cell viability in AR-positive TNBC cells. Enzalutamide treatment could enhance the palbociclib-induced cytostatic effect in AR-positive/RB-proficient TNBC cells. In addition, palbociclib-mediated G1 arrest in AR-positive/RB-proficient TNBC cells was attenuated by RB knockdown. Conclusion Our study provided a preclinical rationale in selecting patients who might have therapeutic benefit from combining CDK4/6 inhibitors with AR antagonists.
Collapse
Affiliation(s)
- Chun-Yu Liu
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ka-Yi Lau
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Chi Hsu
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ji-Lin Chen
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Han Lee
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzu-Ting Huang
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Ting Chen
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Teng Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Hematology & Oncology, Department of Medicine, Yang-Ming Branch of Taipei City Hospital, Taipei, Taiwan
| | - Po-Han Lin
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Ling-Ming Tseng
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
235
|
Herbrink M, de Vries N, Rosing H, Huitema ADR, Nuijen B, Schellens JHM, Beijnen JH. Development and validation of a liquid chromatography-tandem mass spectrometry analytical method for the therapeutic drug monitoring of eight novel anticancer drugs. Biomed Chromatogr 2017; 32. [PMID: 29165815 DOI: 10.1002/bmc.4147] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 11/03/2017] [Accepted: 11/13/2017] [Indexed: 11/09/2022]
Abstract
To support therapeutic drug monitoring of patients with cancer, a fast and accurate method for simultaneous quantification of the registered anticancer drugs afatinib, axitinib, ceritinib, crizotinib, dabrafenib, enzalutamide, regorafenib and trametinib in human plasma using liquid chromatography tandem mass spectrometry was developed and validated. Human plasma samples were collected from treated patients and stored at -20°C. Analytes and internal standards (stable isotopically labeled analytes) were extracted with acetonitrile. An equal amount of 10 mm NH4 CO3 was added to the supernatant to yield the final extract. A 2 μL aliquot of this extract was injected onto a C18 -column, gradient elution was applied and triple-quadrupole mass spectrometry in positive-ion mode was used for detection. All results were within the acceptance criteria of the latest US Food and Drug Administration guidance and European Medicines Agency guidelines on method validation, except for the carry-over of ceritinib and crizotinib. These were corrected for by the injection order of samples. Additional stability tests were carried out for axitinib and dabrafenib in relation to their reported photostability. In conclusion, the described method to simultaneously quantify the eight selected anticancer drugs in human plasma was successfully validated and applied for therapeutic drug monitoring in cancer patients treated with these drugs.
Collapse
Affiliation(s)
- M Herbrink
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Amsterdam, The Netherlands
| | - N de Vries
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Amsterdam, The Netherlands
| | - H Rosing
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Amsterdam, The Netherlands
| | - A D R Huitema
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands
| | - B Nuijen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Amsterdam, The Netherlands
| | - J H M Schellens
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Amsterdam, The Netherlands.,Division of Pharmacoepidemiology and Clinical Pharmacology, Science Faculty, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - J H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital and MC Slotervaart, Amsterdam, The Netherlands.,Division of Pharmacoepidemiology and Clinical Pharmacology, Science Faculty, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
236
|
Mohammad OS, Nyquist MD, Schweizer MT, Balk SP, Corey E, Plymate S, Nelson PS, Mostaghel EA. Supraphysiologic Testosterone Therapy in the Treatment of Prostate Cancer: Models, Mechanisms and Questions. Cancers (Basel) 2017; 9:E166. [PMID: 29210989 PMCID: PMC5742814 DOI: 10.3390/cancers9120166] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 12/13/2022] Open
Abstract
Since Huggins defined the androgen-sensitive nature of prostate cancer (PCa), suppression of systemic testosterone (T) has remained the most effective initial therapy for advanced disease although progression inevitably occurs. From the inception of clinical efforts to suppress androgen receptor (AR) signaling by reducing AR ligands, it was also recognized that administration of T in men with castration-resistant prostate cancer (CRPC) could result in substantial clinical responses. Data from preclinical models have reproducibly shown biphasic responses to T administration, with proliferation at low androgen concentrations and growth inhibition at supraphysiological T concentrations. Many questions regarding the biphasic response of PCa to androgen treatment remain, primarily regarding the mechanisms driving these responses and how best to exploit the biphasic phenomenon clinically. Here we review the preclinical and clinical data on high dose androgen growth repression and discuss cellular pathways and mechanisms likely to be involved in mediating this response. Although meaningful clinical responses have now been observed in men with PCa treated with high dose T, not all men respond, leading to questions regarding which tumor characteristics promote response or resistance, and highlighting the need for studies designed to determine the molecular mechanism(s) driving these responses and identify predictive biomarkers.
Collapse
Affiliation(s)
- Osama S Mohammad
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
- Faculty of Medicine, Benha University, Benha 13518, Egypt.
| | | | - Michael T Schweizer
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
- School of Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Stephen P Balk
- Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA 98195, USA.
| | - Stephen Plymate
- School of Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Peter S Nelson
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Elahe A Mostaghel
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
- School of Medicine, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
237
|
Drug development for noncastrate prostate cancer in a changed therapeutic landscape. Nat Rev Clin Oncol 2017; 15:168-182. [PMID: 29039422 DOI: 10.1038/nrclinonc.2017.160] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The unprecedented progress in the treatment of metastatic castration-resistant prostate cancer is only beginning to be realized in patients with noncastrate disease. This slow progress in part reflects the use of trial objectives focused on time-to-event end points, such as time to metastasis and overall survival, which require long follow-up durations and large sample sizes, and has been further delayed by the use of approved therapies that are effective at the time of progression. Our central hypotheses are that progress can be accelerated, and that outcomes can be improved by shifting trial objectives to response measures occurring early that solely reflect the effects of the treatment. To test these hypotheses, a continuously enrolling multi-arm, multi-stage randomized trial design, analogous to that used in the STAMPEDE trial, has been developed. Eligibility is focused on patients with incurable disease or those with a high risk of death with any form of monotherapy alone. The primary objective is to eliminate all disease using a multimodality treatment strategy. End points include pathological complete response and an undetectable level of serum prostate-specific antigen, with recovery of serum testosterone levels. Both are binary, objective, and provide an early, quantitative indication of efficacy.
Collapse
|
238
|
Abstract
INTRODUCTION AKR1C3 is a drug target in hormonal and hormonal independent malignancies and acts as a major peripheral 17β-hydroxysteroid dehydrogenase to yield the potent androgens testosterone and dihydrotestosterone, and as a prostaglandin (PG) F synthase to produce proliferative ligands for the PG FP receptor. AKR1C3 inhibitors may have distinct advantages over existing therapeutics for the treatment of castration resistant prostate cancer, breast cancer and acute myeloid leukemia. Area covered: This article reviews the patent literature on AKR1C3 inhibitors using SciFinder which identified inhibitors in the following chemical classes: N-phenylsulfonyl-indoles, N-(benzimidazoylylcarbonyl)- N-(indoylylcarbonyl)- and N-(pyridinepyrrolyl)- piperidines, N-benzimidazoles and N-benzindoles, repurposed nonsteroidal antiinflammatory drugs (indole acetic acids, N-phenylanthranilates and aryl propionic acids), isoquinolines, and nitrogen and sulfur substituted estrenes. The article evaluates inhibitor AKR potency, specificity, efficacy in cell-based and xenograft models and clinical utility. The advantage of bifunctional compounds that either competitively inhibit AKR1C3 and block its androgen receptor (AR) coactivator function or act as AKR1C3 inhibitors and direct acting AR antagonists are discussed. Expert opinion: A large number of potent and selective inhibitors of AKR1C3 have been described however, preclinical optimization, is required before their benefit in human disease can be assessed.
Collapse
Affiliation(s)
- Trevor M Penning
- a Center of Excellence in Environmental Toxicology & Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine , University of Pennsylvania , Philadelphia , PA , USA
| |
Collapse
|
239
|
Venema CM, Mammatas LH, Schröder CP, van Kruchten M, Apollonio G, Glaudemans AW, Bongaerts AH, Hoekstra OS, Verheul HM, Boven E, van der Vegt B, de Vries EF, de Vries EG, Boellaard R, Menke van der Houven van Oordt CW, Hospers GA. Androgen and Estrogen Receptor Imaging in Metastatic Breast Cancer Patients as a Surrogate for Tissue Biopsies. J Nucl Med 2017; 58:1906-1912. [DOI: 10.2967/jnumed.117.193649] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/24/2017] [Indexed: 11/16/2022] Open
|
240
|
Ketola K, Munuganti RSN, Davies A, Nip KM, Bishop JL, Zoubeidi A. Targeting Prostate Cancer Subtype 1 by Forkhead Box M1 Pathway Inhibition. Clin Cancer Res 2017; 23:6923-6933. [PMID: 28899970 DOI: 10.1158/1078-0432.ccr-17-0901] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/27/2017] [Accepted: 08/31/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Prostate cancer was recently classified to three clinically relevant subtypes (PCS) demarcated by unique pathway activation and clinical aggressiveness. In this preclinical study, we investigated molecular targets and therapeutics for PCS1, the most aggressive and lethal subtype, with no treatment options available in the clinic.Experimental Design: We utilized the PCS1 gene set and our model of enzalutamide (ENZR) castration-resistant prostate cancer (CRPC) to identify targetable pathways and inhibitors for PCS1. The findings were evaluated in vitro and in the ENZR CRPC xenograft model in vivoResults: The results revealed that ENZR CRPC cells are enriched with PCS1 signature and that Forkhead box M1 (FOXM1) pathway is the central driver of this subtype. Notably, we identified Monensin as a novel FOXM1-binding agent that selectively targets FOXM1 to reverse the PCS1 signature and its associated stem-like features and reduces the growth of ENZR CRPC cells and xenograft tumors.Conclusions: Our preclinical data indicate FOXM1 pathway as a master regulator of PCS1 tumors, namely in ENZR CRPC, and targeting FOXM1 reduces cell growth and stemness in ENZR CRPC in vitro and in vivo These preclinical results may guide clinical evaluation of targeting FOXM1 to eradicate highly aggressive and lethal PCS1 prostate cancer tumors. Clin Cancer Res; 23(22); 6923-33. ©2017 AACR.
Collapse
Affiliation(s)
- Kirsi Ketola
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | | | - Alastair Davies
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Ka Mun Nip
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | | | - Amina Zoubeidi
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada. .,Department of Urology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
241
|
Benoist GE, Hendriks RJ, Mulders PFA, Gerritsen WR, Somford DM, Schalken JA, van Oort IM, Burger DM, van Erp NP. Pharmacokinetic Aspects of the Two Novel Oral Drugs Used for Metastatic Castration-Resistant Prostate Cancer: Abiraterone Acetate and Enzalutamide. Clin Pharmacokinet 2017; 55:1369-1380. [PMID: 27106175 PMCID: PMC5069300 DOI: 10.1007/s40262-016-0403-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Two novel oral drugs that target androgen signaling have recently become available for the treatment of metastatic castration-resistant prostate cancer (mCRPC). Abiraterone acetate inhibits the synthesis of the natural ligands of the androgen receptor, whereas enzalutamide directly inhibits the androgen receptor by several mechanisms. Abiraterone acetate and enzalutamide appear to be equally effective for patients with mCRPC pre- and postchemotherapy. Rational decision making for either one of these drugs is therefore potentially driven by individual patient characteristics. In this review, an overview of the pharmacokinetic characteristics is given for both drugs and potential and proven drug–drug interactions are presented. Additionally, the effect of patient-related factors on drug disposition are summarized and the limited data on the exposure–response relationships are described. The most important pharmacological feature of enzalutamide that needs to be recognized is its capacity to induce several key enzymes in drug metabolism. The potency to cause drug–drug interactions needs to be addressed in patients who are treated with multiple drugs simultaneously. Abiraterone has a much smaller drug–drug interaction potential; however, it is poorly absorbed, which is affected by food intake, and a large interpatient variability in drug exposure is observed. Dose reductions of abiraterone or, alternatively, the selection of enzalutamide, should be considered in patients with hepatic dysfunction. Understanding the pharmacological characteristics and challenges of both drugs could facilitate decision making for either one of the drugs.
Collapse
Affiliation(s)
- Guillemette E Benoist
- Department of Clinical Pharmacy, Radboud University Medical Centre, Route 864, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Rianne J Hendriks
- Department of Urology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Peter F A Mulders
- Department of Urology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Winald R Gerritsen
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Diederik M Somford
- Department of Urology, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Jack A Schalken
- Department of Urology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Inge M van Oort
- Department of Urology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - David M Burger
- Department of Clinical Pharmacy, Radboud University Medical Centre, Route 864, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Nielka P van Erp
- Department of Clinical Pharmacy, Radboud University Medical Centre, Route 864, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
242
|
Behl AS, Ellis LA, Pilon D, Xiao Y, Lefebvre P. Medication Adherence, Treatment Patterns, and Dose Reduction in Patients with Metastatic Castration-Resistant Prostate Cancer Receiving Abiraterone Acetate or Enzalutamide. AMERICAN HEALTH & DRUG BENEFITS 2017; 10:296-303. [PMID: 28975013 PMCID: PMC5620511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 06/20/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND The efficacy of and overall survival associated with metastatic castration-resistant prostate cancer (CRPC) treatments rely on patients' consistent adherence to the recommended dosage regimens. OBJECTIVES To evaluate treatment patterns and patient adherence to abiraterone acetate or enzalutamide therapy in real-world practice, and to examine the factors that may be associated with medication dose reduction in patients with metastatic CRPC. METHODS Retrospective analyses were conducted using the Truven Health MarketScan research databases among patients with metastatic CRPC who initiated treatment with abiraterone acetate or enzalutamide between October 1, 2012, and December 31, 2014 (index date). The patients were followed for up to 12 months, and their baseline characteristics were assessed during the 6 months before the index date. Medication adherence was measured at 3, 6, 9, and 12 months postindex using medication possession ratios (MPRs), and dose reduction was measured using refill gaps and relative dose intensity over the entire observation period. Kaplan-Meier survival analyses and Cox proportional hazards models were used to assess the association between the initial treatment and the risk for dose reduction. RESULTS The study included 2591 and 807 patients who initiated treatment with abiraterone acetate and enzalutamide, respectively. At 6, 9, and 12 months postindex, the patients who initiated abiraterone acetate had higher MPRs than the patients who initiated enzalutamide. In addition, the patients who initiated abiraterone acetate had lower Kaplan-Meier rates of dose reduction across 4 measurements for dose reduction. All hazard ratios for treatment (abiraterone acetate vs enzalutamide) were significantly lower than 1 (range, 0.57-0.80), indicating a lower risk for dose reduction associated with abiraterone acetate. CONCLUSION Patients who initiated abiraterone acetate therapy had higher medication adherence and lower risk for dose reduction than those who initiated enzalutamide therapy. Improved medication adherence may be associated with longer duration of treatment, which in turn may lead to better survival. Further research is needed to assess the potential effect of medication adherence on the overall survival of patients with metastatic CRPC.
Collapse
Affiliation(s)
- Ajay S Behl
- Director of US Oncology Economics and Outcomes Research, Janssen Scientific Affairs, Titusville, NJ
| | - Lorie A Ellis
- Director of US Rheumatology Economics and Outcomes Research, Janssen Scientific Affairs
| | | | | | | |
Collapse
|
243
|
Eiber M, Fendler WP, Rowe SP, Calais J, Hofman MS, Maurer T, Schwarzenboeck SM, Kratowchil C, Herrmann K, Giesel FL. Prostate-Specific Membrane Antigen Ligands for Imaging and Therapy. J Nucl Med 2017; 58:67S-76S. [DOI: 10.2967/jnumed.116.186767] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/20/2017] [Indexed: 02/07/2023] Open
|
244
|
Zhou J, Gao S, Hsieh CL, Malla M, Shemshedini L. Peptide B targets soluble guanylyl cyclase α1 and kills prostate cancer cells. PLoS One 2017; 12:e0184088. [PMID: 28859127 PMCID: PMC5578680 DOI: 10.1371/journal.pone.0184088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/17/2017] [Indexed: 11/30/2022] Open
Abstract
Among androgen-regulated genes, soluble guanylyl cyclase α1 (sGCα1) is significant in promoting the survival and growth of prostate cancer cells and does so independent of nitric oxide (NO) signaling. Peptides were designed targeting sGCα1 to block its pro-cancer functions and one peptide is discussed here. Peptide B-8R killed both androgen-dependent and androgen-independent prostate cancer cells that expressed sGCα1, but not cells that do not express this gene. Peptide B-8R induced apoptosis of prostate cancer cells. Importantly, Peptide B-8R does not affect nor its cytotoxicity depend on NO signaling, despite the fact that it associates with sGCα1, which dimerizes with sGCβ1 to form the sGC enzyme. Just as with a previously studied Peptide A-8R, Peptide B-8R induced elevated levels of reactive oxygen species (ROS) in prostate cancer cells, but using a ROS-sequestering agent showed that ROS was not responsible the cytotoxic activity of Peptide B-8R. Interestingly, Peptide B-8R induced elevated levels of p53 and phosphorylated p38, but neither of these changes is the cause of the peptide’s cytotoxicity. Additional drugs were used to alter levels of iron levels in cells and these studies showed that Peptide B-8R activity does not depend on Ferroptosis. Thus, future work will be directed at defining the mechanism of cytotoxic action of Peptide B-8R against prostate cancer cells.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Shuai Gao
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Chen-Lin Hsieh
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Mamata Malla
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Lirim Shemshedini
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
- * E-mail:
| |
Collapse
|
245
|
Chen J, Li L, Yang Z, Luo J, Yeh S, Chang C. Androgen-deprivation therapy with enzalutamide enhances prostate cancer metastasis via decreasing the EPHB6 suppressor expression. Cancer Lett 2017; 408:155-163. [PMID: 28826721 DOI: 10.1016/j.canlet.2017.08.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/04/2017] [Accepted: 08/09/2017] [Indexed: 10/19/2022]
Abstract
Early studies suggested that using ADT with the recently developed anti-androgen Enzalutamide (Enz, also named as MDV3100 could extent castration resistant prostate cancer (CRPC) patients' survival an extra 4.8 months. Yet the therapy in most patients might eventually fail due to development of Enz-resistance. Here we found Enz might also increase some unwanted side-effects via increasing the CRPC cell invasion that might involve altering the Enz-mediated androgen receptor (AR)/EPHB6 suppressor/JNK signaling. Results from multiple clinical surveys also indicated that EPHP6 might function as a suppressor of PCa metastasis. Mechanism dissection revealed that Enz-mediated AR might function via binding to the androgen-response-element (ARE) on the EPHB6 promoter to decrease EPHB6 suppressor expression, which might then activate the phosphorylation of JNK signals to increase the CRPC cell invasion. Targeting this newly identified AR/EPHB6/JNK signaling with JNK inhibitor (SP600125) may then block/reverse the Enz-increased CRPC cell invasion. Collectively, our results suggest that Enz may increase CRPC cell invasion via altering the AR/EPHB6/JNK/MMP9 signaling and targeting this newly identified signaling may help us to increase the Enz efficacy to better suppress the CRPC at the later metastatic stage.
Collapse
Affiliation(s)
- Jiaqi Chen
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China; George Whipple Lab for Cancer Research, Departments of Pathology and Urology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Lei Li
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China; George Whipple Lab for Cancer Research, Departments of Pathology and Urology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| | - Zhao Yang
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China; George Whipple Lab for Cancer Research, Departments of Pathology and Urology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Jie Luo
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA; Sex Hormone Research Center, China Medical University/Hospital, Taichung, 404, Taiwan.
| |
Collapse
|
246
|
Abstract
Despite a growing number of treatment options, metastatic castrate resistant prostate cancer remains almost universally fatal. Dose individualization ensures patients receive the maximal benefit from each line of treatment potentially leading to improved outcomes, a reduction in quality of life impairment and minimization of premature cessation for avoidable toxicity. Herein, we review drug-specific issues that may be associated with unexpected or unrecognized variations in drug systemic exposure despite the use of protocol doses. In particular, we discuss the potential for under-exposure of docetaxel and cabazitaxel; over-exposure of enzalutamide; and varied absorption of abiraterone acetate.
Collapse
Affiliation(s)
- Megan Crumbaker
- Crown Princess Mary Cancer Care Centre, Westmead Hospital, New South Wales, Australia
- Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Howard Gurney
- Crown Princess Mary Cancer Care Centre, Westmead Hospital, New South Wales, Australia
- Macquarie University, New South Wales, Australia
| |
Collapse
|
247
|
|
248
|
Targeting androgen receptor versus targeting androgens to suppress castration resistant prostate cancer. Cancer Lett 2017; 397:133-143. [DOI: 10.1016/j.canlet.2017.03.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/11/2017] [Accepted: 03/13/2017] [Indexed: 12/31/2022]
|
249
|
Crona DJ, Whang YE. Androgen Receptor-Dependent and -Independent Mechanisms Involved in Prostate Cancer Therapy Resistance. Cancers (Basel) 2017; 9:cancers9060067. [PMID: 28604629 PMCID: PMC5483886 DOI: 10.3390/cancers9060067] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/07/2017] [Accepted: 06/07/2017] [Indexed: 12/12/2022] Open
Abstract
Despite the initial efficacy of androgen deprivation in prostate cancer, virtually all patients progress to castration-resistant prostate cancer (CRPC). Androgen receptor (AR) signaling is critically required for CRPC. A new generation of medications targeting AR, such as abiraterone and enzalutamide, has improved survival of metastatic CRPC (mCRPC) patients. However, a significant proportion of patients presents with primary resistance to these agents, and in the remainder, secondary resistance will invariably develop, which makes mCRPC the lethal form of the disease. Mechanisms underlying progression to mCRPC and treatment resistance are extremely complex. AR-dependent resistance mechanisms include AR amplification, AR point mutations, expression of constitutively active AR splice variants, and altered intratumoral androgen biosynthesis. AR-independent resistance mechanisms include glucocorticoid receptor activation, immune-mediated resistance, and neuroendocrine differentiation. The development of novel agents, such as seviteronel, apalutamide, and EPI-001/EPI-506, as well as the identification and validation of novel predictive biomarkers of resistance, may lead to improved therapeutics for mCRPC patients.
Collapse
Affiliation(s)
- Daniel J Crona
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Young E Whang
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
250
|
An androgen response element driven reporter assay for the detection of androgen receptor activity in prostate cells. PLoS One 2017; 12:e0177861. [PMID: 28570625 PMCID: PMC5453475 DOI: 10.1371/journal.pone.0177861] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/20/2017] [Indexed: 01/20/2023] Open
Abstract
The androgen receptor (AR) transcription factor plays a key role in the development and progression of prostate cancer, as is evident from the efficacy of androgen-deprivation therapy, AR is also the most frequently mutated gene, in castration resistant prostate cancer (CRPC). AR has therefore become an even more attractive therapeutic target in aggressive and disseminated prostate cancer. To investigate mechanisms of AR and AR target gene activation in different subpopulations of prostate cancer cells, a toolkit of AR expressor and androgen response element (ARE) reporter vectors were developed. Three ARE reporter vectors were constructed with different ARE consensus sequences in promoters linked to either fluorescence or luciferase reporter genes in lentiviral vector backbones. Cell lines transduced with the different vectors expressed the reporters in an androgen-dependent way according to fluorescence microscopy, flow cytometry and multi-well fluorescent and luminescence assays. Interestingly, the background reporter activity in androgen-depleted medium was significantly higher in LNCaP cells compared to the prostate transit amplifying epithelial cell lines, EP156T-AR and 957E/hTERT-AR with exogenous AR. The androgen-induced signal to background was much higher in the latter benign prostate cells than in LNCaP cells. Androgen-independent nuclear localization of AR was seen in LNCaP cells and reduced ARE-signaling was seen following treatment with abiraterone, an androgen synthesis inhibitor. The ARE reporter activity was significantly stronger when stimulated by androgens than by β-estradiol, progesterone and dexamethasone in all tested cell types. Finally, no androgen-induced ARE reporter activity was observed in tumorigenic mesenchymal progeny cells of EP156T cells following epithelial to mesenchymal transition. This underscores the observation that expression of the classical luminal differentiation transcriptome is restricted in mesenchymal type cells with or without AR expression, and presence of androgen.
Collapse
|