Development of a Functional Assay to Test for Homologous Recombination

Total Page:16

File Type:pdf, Size:1020Kb

Development of a Functional Assay to Test for Homologous Recombination Use of the androgen signalling pathway to identify ovarian cancer patients suitable for hormonal therapy Thesis submitted for the degree of Doctor of Philosophy Northern Institute for Cancer Research Angelika Kaufmann Clinical Research Fellow Northern Gynaecological Cancer Centre, Queen Elizabeth Hospital, Gateshead Supervisors Professor Richard J Edmondson Professor of Gynaecological Oncology, Faculty Institute for Cancer Sciences, University of Manchester Professor Craig N Robson Professor of Molecular Urology, NICR, Newcastle University Luke Gaughan University Lecturer, NICR, Newcastle University March 2020 Statement of originality The work presented in this thesis is original except where acknowledged in the references and was carried out between February 2012 until January 2015 at the at the Northern Institute for Cancer Research, Newcastle University and the Northern Gynaecological Oncology Centre, Gateshead. It has not been, or will not be, submitted for any other qualification at any other university. i ii Abstract Background Despite confirmed AR expression in epithelial ovarian cancer (EOC), clinical response to anti- androgen treatment is poor. Stratification of susceptible individuals with a specific biomarker, such as the previously described Rab35, might enable more effective treatment strategies. Abiraterone, a steroid synthesis inhibitor, might be of therapeutic benefit in specific EOC subgroups. Methods Primary cell cultures (PCO) generated from ascites were used as a representative model for the heterogeneity of EOC. PCOs were examined for AR and Rab35 expression at mRNA and protein level and were stimulated with androgens to evaluate subsequent Rab35 expression. CYP17 expression was measured in ovarian cancer cell lines and PCOs and the effect of abiraterone on proliferation was assessed in two ovarian cancer cell lines. Results The AR expression was widely different when examined with qRT-PCR, Western blotting and immunohistochemistry. No correlations were found between the modalities for either AR or Rab35 expression. In contrast, AR and Rab35 expression showed a positive correlation at the protein and mRNA level. However, androgen treatment of PCOs showed >50% increase in Rab35 mRNA expression in only 40% of PCOs. CYP17 expression was confirmed in all examined cell cultures and PCOs at both, the protein and mRNA level. Abiraterone treatment of the ovarian cancer cell lines led to significant inhibitory effects on proliferation. On protein level however, abiraterone exposure resulted in increased expression of AR and CYP17. Conclusion Although AR expression was confirmed in POCs, it remains unclear which technique would be most suitable to stratify for androgen expressing tumours. iii Rab35 in PCOs appeared to be androgen-related and hence may not be a suitable biomarker in EOC for AR. The inhibitory effect of abiraterone on proliferation that was observed in ovarian cultures is suggestive of a dual action of the compound. Response to abiraterone exposure in PCOs might help to determine potential treatment effects. iv Acknowledgements I would like to thank my supervisors Prof Richard Edmondson and Prof Craig Robson for their guidance, support and trouble-shooting during this project and Luke Gaughan and Peter Donoghue for enduring my many questions. The immense help, teaching, advice and support from all the members from the Solid Tumour Target Discovery Group and my “ovarian partners in crime” Rachel O’Donnell and Aiste McCormick has greatly influenced this project. I am grateful for the funding received from and the lessons learnt at the Northern Gynaecology Oncology Centre (NGOC) during my time in Newcastle and Gateshead. But most of all, I would like to thank the women donating ascites to enable this project in the hope to selflessly help other ovarian cancer sufferers. Meeting these women and their families has touched me deeply. Family and friends near and afar: for your love and support before, throughout and after this journey- thank you. v vi Table of Contents Statement of originality ............................................................................................................. i Abstract ....................................................................................................................................iii Acknowledgements ................................................................................................................... v Table of Contents .................................................................................................................... vii List of Abbreviations ............................................................................................................... xi List of Figures ........................................................................................................................ xiv List of Tables ........................................................................................................................xviii 1 Introduction ...................................................................................................................... 1 1.1 Ovarian cancer ............................................................................................................. 1 1.1.1 Epidemiology of ovarian cancer ........................................................................... 1 1.2 Classification of ovarian cancer ................................................................................... 3 1.2.1 Historical classification of ovarian cancer ............................................................ 3 1.2.2 New classification of epithelial ovarian cancer .................................................... 3 1.3 Aetiology of ovarian cancer ......................................................................................... 4 1.4 Risk factors for ovarian cancer .................................................................................... 7 1.4.1 Hormonal factors .................................................................................................. 8 1.5 Protective factors for ovarian cancer ......................................................................... 10 1.6 Screening in ovarian cancer ....................................................................................... 11 1.7 Treatment in ovarian cancer ....................................................................................... 13 1.7.1 Treatment in primary disease ............................................................................. 13 1.7.2 Treatment of recurrent disease ........................................................................... 14 1.7.3 Targeted therapy ................................................................................................. 14 1.8 Androgens and ovarian cancer ................................................................................... 19 1.8.1 Androgens in the healthy female ........................................................................ 19 1.8.2 Androgen receptor (AR) ..................................................................................... 21 1.8.3 Androgen receptor signalling in ovarian cancer ................................................. 22 1.8.4 Androgen Receptor expression in ovarian cancer .............................................. 25 1.8.5 Androgen receptor expression in ovarian cancer cell lines ................................ 26 1.8.6 Androgen receptor expression data in primary cell cultures .............................. 26 1.8.7 Epidemiologic evidence ..................................................................................... 27 1.8.8 Anti-Androgen therapy in ovarian cancer .......................................................... 28 1.9 GTPases ..................................................................................................................... 31 1.9.1 Rab subfamily ..................................................................................................... 32 1.10 Cytochrome P450 17α (CYP17) ............................................................................ 35 1.10.1 Congenital 17-hydroxylase/ 17,20 deficiency .................................................... 36 1.10.2 CYP17 and ovarian cancer ................................................................................. 36 1.10.3 Abiraterone (CB7598) ........................................................................................ 37 2 Materials and Methods................................................................................................... 42 2.1 General Laboratory Practice ...................................................................................... 42 vii 2.2 Tissue culture ............................................................................................................ 42 2.2.1 Cell lines ............................................................................................................ 42 2.2.2 Cell culture maintenance .................................................................................... 43 2.2.3 Primary cultures (PCO) ...................................................................................... 44 2.2.4 Cell culture storage ............................................................................................ 44 2.2.5 Thawing of cell cultures ..................................................................................... 45 2.3 Immunofluorescence ................................................................................................
Recommended publications
  • Therapies Targeted to Androgen Receptor Signaling Axis in Prostate Cancer: Progress, Challenges, and Hope
    cancers Review Therapies Targeted to Androgen Receptor Signaling Axis in Prostate Cancer: Progress, Challenges, and Hope Sirin Saranyutanon 1,2, Sanjeev Kumar Srivastava 1,2,*, Sachin Pai 3, Seema Singh 1,2,4 and Ajay Pratap Singh 1,2,4,* 1 Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL 36617, USA; [email protected] (S.S.); [email protected] (S.S.) 2 Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA 3 Department of Medical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA; [email protected] 4 Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA * Correspondence: [email protected] (S.K.S.); [email protected] (A.P.S.); Tel.: +1-251-445-9874 (S.K.S.); +1-251-445-9843 (A.P.S.) Received: 4 November 2019; Accepted: 18 December 2019; Published: 23 December 2019 Abstract: Prostate cancer is the mostly commonly diagnosed non-cutaneous malignancy and the second leading cause of cancer-related death affecting men in the United States. Moreover, it disproportionately affects the men of African origin, who exhibit significantly greater incidence and mortality as compared to the men of European origin. Since androgens play an important role in the growth of normal prostate and prostate tumors, targeting of androgen signaling has remained a mainstay for the treatment of aggressive prostate cancer. Over the years, multiple approaches have been evaluated to effectively target the androgen signaling pathway that include direct targeting of the androgens, androgen receptor (AR), AR co-regulators or other alternate mechanisms that impact the outcome of androgen signaling.
    [Show full text]
  • HSD3B1 and Response to a Nonsteroidal CYP17A1 Inhibitor in Castration-Resistant Prostate Cancer
    Research JAMA Oncology | Brief Report HSD3B1 and Response to a Nonsteroidal CYP17A1 Inhibitor in Castration-Resistant Prostate Cancer Nima Almassi, MD; Chad Reichard, MD; Jianbo Li, PhD; Carly Russell, MS; Jaselle Perry, MS; Charles J. Ryan, MD; Terence Friedlander, MD; Nima Sharifi, MD Invited Commentary page 562 IMPORTANCE The HSD3B1 (1245C) germline variant encodes for a gain-of-function missense Related article page 558 in 3β-hydroxysteroid dehydrogenase isoenzyme 1 (3βHSD1) that results in increased dihydrotestosterone synthesis from extragonadal precursors and is predictive of more rapid progression to castration-resistant prostate cancer (CRPC). OBJECTIVE To determine whether the HSD3B1 (1245C) genotype is predictive of clinical response to extragonadal androgen ablation with nonsteroidal 17α-hydroxylase/17,20-lyase (CYP17A1) inhibition in men with metastatic CRPC. DESIGN, SETTING, AND PARTICIPANTS An observational study of men with metastatic CRPC treated with ketoconazole between June 1998 and December 2012 was conducted at the University of California, San Francisco. EXPOSURES Extragonadal androgen ablation with the nonsteroidal CYP17A1 inhibitor ketoconazole among men with metastatic CRPC. MAIN OUTCOMES AND MEASURES The primary end points of analysis were duration of ketoconazole therapy and time to disease progression stratified by HSD3B1 genotype. Disease progression was defined as either biochemical or radiographic progression, using the Prostate Cancer Working Group 3 and Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 definitions, respectively. Kaplan-Meier analysis was used to estimate time on therapy and time to disease progression. A log-rank test for trend was used to compare outcomes by HSD3B1 genotype. RESULTS A total of 90 men (median [interquartile range] age, 61.5 [55.3-67.0] years) with metastatic CRPC were included in the analysis, with sufficient data to determine duration of ketoconazole therapy and time to disease progression in 88 and 81 patients, respectively.
    [Show full text]
  • Slow Tight Binding Inhibition of CYP17A1 by Abiraterone Redefines Its Kinetic Selectivity and Dosing Regimen
    JPET Fast Forward. Published on June 17, 2020 as DOI: 10.1124/jpet.120.265868 This article has not been copyedited and formatted. The final version may differ from this version. JPET # 265868 Slow tight binding inhibition of CYP17A1 by abiraterone redefines its kinetic selectivity and dosing regimen Eleanor Jing Yi Cheong1, Pramod C Nair2, Rebecca Wan Yi Neo1, Ho Thanh Tu1, Fu Lin3, Edmund Chiong4,5, Kesavan Esuvaranathan4,5, Hao Fan6, Russell Z. Szmulewitz7, Cody J. Peer8, William D. Figg8, Christina Li Lin Chai1, John O Miners2, Eric Chun Yong Chan1,9 1Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore; 2Department of Clinical Pharmacology and Flinders Centre for Downloaded from Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide, Australia; 3Bioinformatics Institute, Biotransformation Innovation Platform (BioTrans), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, Singapore 138671; 4Department of Surgery, jpet.aspetjournals.org National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore; 5Department of Urology, National University Hospital, 5 Lower Kent Ridge Road, Singapore 119074; 6Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, at ASPET Journals on September 27, 2021 Singapore 138671; Department of Biological Sciences, National University of Singapore, 14 Science 23 Drive 4, Singapore 117543; Centre for Computational Biology, DUKE-NUS Medical School, 8 College Road, Singapore 169857; 7The University of Chicago, Chicago, 8National Cancer Institute, Rockville, MD, 9National University Cancer Institute, Singapore (NCIS), NUH Medical Centre (NUHMC), 5 Lower Kent Ridge Road, Singapore 119074 1 | Page JPET Fast Forward.
    [Show full text]
  • Resistance to Second Generation Antiandrogens in Prostate Cancer: Pathways and Mechanisms
    Verma et al. Cancer Drug Resist 2020;3:742-61 Cancer DOI: 10.20517/cdr.2020.45 Drug Resistance Review Open Access Resistance to second generation antiandrogens in prostate cancer: pathways and mechanisms Shiv Verma1,2, Kumari Sunita Prajapati3, Prem Prakash Kushwaha3, Mohd Shuaib3, Atul Kumar Singh3, Shashank Kumar3, Sanjay Gupta1,2,4,5,6 1Department of Urology, Case Western Reserve University, Cleveland, OH 44106, USA. 2The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA. 3School of Basic and Applied Sciences, Department of Biochemistry and Microbial Sciences, Central University of Punjab, Bathinda 151001, India. 4Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA. 5Divison of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA. 6Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA. Correspondence to: Prof. Sanjay Gupta, The James and Eilleen Dicke Laboratory, Department of Urology, Case Western Reserve University, 2109 Adelbert Road, Wood Research Tower-RTG01, Cleveland, OH 44106, USA. E-mail: [email protected] How to cite this article: Verma S, Prajapati KS, Kushwaha PP, Shuaib M, Singh AK, Kumar S, Gupta S. Resistance to second generation antiandrogens in prostate cancer: pathways and mechanisms. Cancer Drug Resist 2020;3:742-61. http://dx.doi.org/10.20517/cdr.2020.45 Received: 24 Jun 2020 First Decision: 31 Jul 2020 Revised: 3 Aug 2020 Accepted: 6 Aug 2020 Available online: 17 Sep 2020 Academic Editor: Vincent C. O. Njar Copy Editor: Cai-Hong Wang Production Editor: Jing Yu Abstract Androgen deprivation therapy targeting the androgens/androgen receptor (AR) signaling continues to be the mainstay treatment of advanced-stage prostate cancer.
    [Show full text]
  • What Predicts Minimal Response to Abiraterone in Metastatic Castrate-Resistant Prostate Cancer?
    ANTICANCER RESEARCH 35: 5615-5622 (2015) What Predicts Minimal Response to Abiraterone in Metastatic Castrate-resistant Prostate Cancer? RHIAN SIÂN DAVIES1, CHRISTIAN SMITH1, MICHAEL ROBERT BUTTON1, JACOB TANGUAY1, JIM BARBER1, NACHIAPPAN PALANIAPPAN1, JOHN STAFFURTH1,2 and JASON FRANCIS LESTER1 1Velindre Cancer Centre, Cardiff, Wales, U.K.; 2Institute of Cancer Genetics, Cardiff University, Cardiff, Wales, U.K. Abstract. Background: Several treatments have been shown mitoxantrone-plus-prednisolone in patients with mCRPC (3, to prolong survival in metastatic castrate-resistant prostate 4). Since 2010, several new agents have been shown to cancer (mCRPC); the optimum sequencing of these is not prolong overall survival in men with mCRPC after docetaxel established. We investigated methods of identifying patients treatment (Table I) (5-8). These new agents have with mCRPC unlikely to respond to abiraterone. Patients and significantly improved the outcomes of men with mCRPC, Methods: A retrospective analysis was carried-out in 47 however, there are a proportion of patients who do not consecutive patients with mCRPC treated sequentially with respond to a particular therapy. For example, in the Cou-301 androgen deprivation (LHRHa), bicalutamide, docetaxel then trial, only 38% of patients in the abiraterone arm had a PSA abiraterone. Results: The median progression-free survival and/or radiological response (5). It is reasonable to assume in patients treated with abiraterone was shorter in those with there are a significant number of patients on abiraterone who ≤18 months’ response to LHRHa (118 vs. 279 days; do not respond to treatment, develop worsening disease- p=0.018), bicalutamide non-responders (91 vs. 196 days; related symptoms, suffer a fall in performance status and as p=0.003) and patients with ≤6 months' response to docetaxel a consequence are not fit enough for other potentially (102 vs.
    [Show full text]
  • Androgen Receptor Functions in Castration-Resistant Prostate Cancer and Mechanisms of Resistance to New Agents Targeting the Androgen Axis
    Oncogene (2014) 33, 2815–2825 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc REVIEW Androgen receptor functions in castration-resistant prostate cancer and mechanisms of resistance to new agents targeting the androgen axis X Yuan, C Cai, S Chen, S Chen, Z Yu and SP Balk The metabolic functions of androgen receptor (AR) in normal prostate are circumvented in prostate cancer (PCa) to drive tumor growth, and the AR also can acquire new growth-promoting functions during PCa development and progression through genetic and epigenetic mechanisms. Androgen deprivation therapy (ADT, surgical or medical castration) is the standard treatment for metastatic PCa, but patients invariably relapse despite castrate androgen levels (castration-resistant PCa, CRPC). Early studies from many groups had shown that AR was highly expressed and transcriptionally active in CRPC, and indicated that steroids from the adrenal glands were contributing to this AR activity. More recent studies showed that CRPC cells had increased expression of enzymes mediating androgen synthesis from adrenal steroids, and could synthesize androgens de novo from cholesterol. Phase III clinical trials showing a survival advantage in CRPC for treatment with abiraterone (inhibitor of the enzyme CYP17A1 required for androgen synthesis that markedly reduces androgens and precursor steroids) and for enzalutamide (new AR antagonist) have now confirmed that AR activity driven by residual androgens makes a major contribution to CRPC, and led to the recent Food and Drug Administration approval of both agents. Unfortunately, patients treated with these agents for advanced CRPC generally relapse within a year and AR appears to be active in the relapsed tumors, but the molecular mechanisms mediating intrinsic or acquired resistance to these AR-targeted therapies remain to be defined.
    [Show full text]
  • Sex Steriods
    Sex Steroids Endo, Energy and Repro 2017-2018 SEX STEROIDS David Mangelsdorf, Ph.D., Office: ND9.120, Phone: 55957 Email: [email protected] LEARNING OBJECTIVES: This section covers the mechanism of action and therapeutic uses of sex steroids. At the end of this lecture you should be able to: • Summarize the biosynthesis and mechanism of actions of androgens, estrogens, and progestins. • Identify the different therapeutic uses of sex steroid-based drugs (including HRT and birth control) and their mechanism of action. • Interpret the parallel and contrasting uses of these drugs in men vs. women; contrast the use of these drugs in cancer versus non-cancer indications. • Identify the basic structural features and the general pharmacological differences (including mechanisms of action, side effects, etc.) between estrogenic, progestinic, and androgenic drugs. • Describe the different treatment options for endocrine-related disorders of sex steroids. • In a given postmenopausal patient, identify risk factors for the use of HRT, list the types of drugs available and identify when they should be used. • Interpret the phase relationships between different OCP preparations and why they exist. • Recognize the use of non-pharmacological options for contraception. I. INTRODUCTION A. The sex steroids are the effector arm of the hypothalamic-pituitary-gonadal axis (see lecture on “Hypothalamic Pituitary Axes”). B. Sex steroids are some of the most widely used drugs pharmaceutically, particularly in birth control. 1. This is an oddity in pharmacology and medicine, in that the drug is given to healthy normal individuals. 2. Because the drug is used on a healthy person, it has to be under more stringent control to avoid potential side effects.
    [Show full text]
  • Inhibition of the Androgen Receptor by Mineralocorticoids at Levels Physiologically Achieved in Serum in Patients Treated with Abiraterone Acetate
    Prostate Cancer and Prostatic Disease (2014) 17, 292–299 & 2014 Macmillan Publishers Limited All rights reserved 1365-7852/14 www.nature.com/pcan ORIGINAL ARTICLE Inhibition of the androgen receptor by mineralocorticoids at levels physiologically achieved in serum in patients treated with abiraterone acetate W Kim1, JO Jones2, M Diamond3, C Haqq4, A Molina5, EJ Small1 and CJ Ryan1 BACKGROUND: Abiraterone acetate (AA), a highly potent CYP17A1 inhibitor, has demonstrated marked clinical benefit in patients with metastatic castration-resistant prostate cancer (CRPC). Phase I trials of AA without prednisone showed significant elevation of serum mineralocorticoid concentrations. The aim of this study was to elucidate the biological significance of elevated mineralocorticoid levels on androgen receptor (AR) activity in prostate cancer (PC) cells. METHODS: Fluorescence resonance energy transfer (FRET) assay was used to assess the effect of mineralocorticoids on androgen- induced conformational change of the AR. LAPC4, LNCaP and LN-AR cells that were cultured and treated with androgens were exposed to mineralocorticoids at varying concentrations, including levels measured in the serum of AA-treated patients in a phase I trial. AR-dependent transcriptional activity and cell growth were measured in these cell lines to determine the biological impact of mineralocorticoids on PC cells. RESULTS: Corticosterone (CS) and deoxycorticosterone (DOC) inhibited androgen-induced conformational change of the AR in the FRET assay. CS inhibited AR-dependent transcriptional activity and cell growth at concentrations comparable to those measured in the serum of AA-treated patients. DOC inhibited AR transcriptional activity and cell growth at 10-fold greater concentrations than measured in the serum of AA-treated patients.
    [Show full text]
  • De Novo Androgen Synthesis As a Mechanism Contributing to the Progression of Prostate Cancer to Castration Resistance
    DE NOVO ANDROGEN SYNTHESIS AS A MECHANISM CONTRIBUTING TO THE PROGRESSION OF PROSTATE CANCER TO CASTRATION RESISTANCE by JENNIFER ANN LOCKE B.Sc., The University of British Columbia, 2005 A THESIS SUBMITTED IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY in THE FACULTY OF GRADUATE STUDIES (Experimental Medicine) THE UNIVERSITY OF BRITISH COLUMBIA (Vancouver) June 2009 © Jennifer Ann Locke, 2009 Abstract Prostate cancer (CaP) is the leading cause of cancer in men affecting 24,700 Canadians each year and the third leading cause of cancer mortality with 4,300 deaths each year. CaP cells are derived from the prostate secretory epithelium and depend on androgen ligand activation of androgen receptor (AR) for survival, growth and proliferation. Androgen deprivation therapy (ADT) through pharmacological methods has been the leading form of CaP therapy since Huggin‟s discovery that castration induced the regression of CaP tumors in 1941. Unfortunately, the cancer often recurs within 2-4 years in what has classically been considered “androgen-independent” (AI) disease. Growing evidence implicates androgens and AR activation in this disease recurrence despite castration, suggesting that this terminology should be more appropriately called “castration-resistant” prostate cancer (CRPC). Firstly, AR is found amplified, overexpressed or mutated in a majority of recurrent cancers as compared to primary cancers and secondly, intratumoral testosterone levels remain the same pre- and post-ADT. Additionally, the measured intratumoral DHT levels are sufficient to activate AR in recurrent CaP cells despite low serum androgen levels suggesting that intratumoral androgens remain important mediators of AR-mediated CaP progression. Previously, we and others discovered that recurrent tumor cells have elevated levels of enzymes in the pathways necessary for androgen synthesis from cholesterol.
    [Show full text]
  • Pharmacokinetics of Abiraterone Acetate Released from a Tablet Based on Lipid Matrix in Beagle Dogs
    ORIGINAL ARTICLES Department of Pharmaceutics1, School of Pharmacy, Qingdao University; Department of Thoracic Surgery2, Qingdao Central Hospital, Qingdao, China Pharmacokinetics of abiraterone acetate released from a tablet based on lipid matrix in beagle dogs H. ZHOU1, P. XU2, Y. SUN1,* Received November 23, 2020, accepted April 17, 2021 *Corresponding author: Yong Sun, Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, China [email protected] Pharmazie 76: 308-312 (2021) doi: 10.1691/ph.2021.0171 Objective: To compare the pharmacokinetic behavior of abiraterone acetate after oral administration of a lipid- based formulation tablet and a reference preparation, and to study the relative bioavailability of abiraterone acetate released from the lipid matrix-based tablet. Methods: Beagle dogs received a single dose orally. The experimental dosage was 75 mg/tablet, and the reference dosage was 250 mg/tablet. Six beagle dogs in each group were investigated with the method of 3 × 3 cross-administration. Blood plasma was collected and centri- fuged before administration and 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 4, 6, 8, 12, and 24 h after administration, and the plasma samples were analyzed by liquid chromatography–tandem mass spectrometry (LC–MS/MS). Results: The half-life (T1/2) values of lipid-based formulation (LBF) abiraterone acetate tablet samples (75 mg/tablet) and reference tablets (250 mg/tablet) after oral administration were 3.42 and 4.27 h, respectively, while the area under the concentration time curve (AUC0-t) (h·ng/mL) values were 107.71 and 52.83, respectively. The F value of the relative bioavailability was 704.80%.
    [Show full text]
  • Discovery of Novel Non-Steroidal Cytochrome P450 17A1 Inhibitors As Potential Prostate Cancer Agents
    International Journal of Molecular Sciences Article Discovery of Novel Non-Steroidal Cytochrome P450 17A1 Inhibitors as Potential Prostate Cancer Agents Tomasz M. Wróbel 1,2,* , Oksana Rogova 1, Kasper L. Andersen 3 , Rahul Yadav 4, Simone Brixius-Anderko 4, Emily E. Scott 4,5, Lars Olsen 1,6, Flemming Steen Jørgensen 1 and Fredrik Björkling 1 1 Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; [email protected] (O.R.); [email protected] (L.O.); [email protected] (F.S.J.); [email protected] (F.B.) 2 Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Medical University of Lublin, Chod´zki4a, 20093 Lublin, Poland 3 Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, DK-2200 Copenhagen, Denmark; [email protected] 4 Department of Medicinal Chemistry, University of Michigan, 428 Church Street, Ann Arbor, MI 48109-1065, USA; [email protected] (R.Y.); [email protected] (S.B.-A.); [email protected] (E.E.S.) 5 Department of Pharmacology, University of Michigan, 428 Church Street, Ann Arbor, MI 48109-1065, USA 6 Protein Engineering, Novozymes A/S, Krogshøjvej 36, DK-2880 Bagsvaerd, Denmark * Correspondence: [email protected]; Tel.: +48-814-487-273 Received: 1 June 2020; Accepted: 7 July 2020; Published: 9 July 2020 Abstract: The current study presents the design, synthesis, and evaluation of novel cytochrome P450 17A1 (CYP17A1) ligands. CYP17A1 is a key enzyme in the steroidogenic pathway that produces androgens among other steroids, and it is implicated in prostate cancer.
    [Show full text]
  • Changing Paradigms in Management of Metastatic Castration Resistant Prostate Cancer (Mcrpc) Eva Gupta1*, Troy Guthrie2 and Winston Tan1
    Gupta et al. BMC Urology 2014, 14:55 http://www.biomedcentral.com/1471-2490/14/55 REVIEW Open Access Changing paradigms in management of metastatic Castration Resistant Prostate Cancer (mCRPC) Eva Gupta1*, Troy Guthrie2 and Winston Tan1 Abstract Recently, the standard of care for metastatic Castration Resistant Prostate Cancer (mCRPC) has changed considerably. Persistent androgen receptor (AR) signaling has been identified as a target for novel therapies and reengages the fact that AR continues to be the primary target responsible for metastatic prostate cancer. Androgen receptor gene amplification and over expression have been found to result in a higher concentration of androgen receptors on tumor cells, making them extremely sensitive to low levels of circulating androgens. Additionally, prostate cancer cells are able to maintain dihydrotestosterone (DHT) concentration in excess of serum concentrations to support tumor growth. For many years ketoconazole was the only CYP17 inhibitor that was used to treat mCRPC. However, significant toxicities limit its use. Newly approved chemotherapeutic agents such as Abiraterone (an oral selective inhibitor of CYP17A), which blocks androgen biosynthesis both within and outside the prostate cancer cells), and enzalutamide (blocks AR signaling) have improved overall survival. There are also ongoing phase III trials for Orteronel (TAK- 700), ARN- 509 and Galeterone (TOK-001), which targets androgen signaling. In this review, we will present the rationale for the newly approved hormonal treatments, their indications and complications, and we will discuss ongoing trials that are being done to improve the efficacy of the approved agents. Finally, we will talk about the potential upcoming hormonal treatments for mCRPC.
    [Show full text]