Elsevier

European Urology

Volume 67, Issue 3, March 2015, Pages 470-479
European Urology

Platinum Priority – Prostate Cancer
Editorial by Jens Adam Ceder on pp. 480–481 of this issue
Understanding the Mechanisms of Androgen Deprivation Resistance in Prostate Cancer at the Molecular Level

https://doi.org/10.1016/j.eururo.2014.09.049Get rights and content

Abstract

Context

Various molecular mechanisms play a role in the development of resistance to androgen deprivation therapy in castration-resistant prostate cancer (CRPC).

Objective

To understand the mechanisms and biological pathways associated with the progression of prostate cancer (PCa) under systemic androgen depletion or administration of the novel antiandrogens abiraterone, enzalutamide, and ARN-509. This review also examines the introduction of novel combinational approaches for patients with CRPC.

Evidence acquisition

PubMed was the data source. Keywords for the search were castrate resistant prostate cancer, abiraterone, enzalutamide resistance mechanisms, resistance to androgen deprivation, AR mutations, amplifications, splice variants, and AR alterations. Papers published before 1990 were excluded from the review, and only English-language papers were included.

Evidence synthesis

This review summarizes the current literature regarding the mechanisms implicated in the development of CRPC and the acquisition of resistance to novel antiandrogen axis agents. The review focuses on androgen biosynthesis in the tumor microenvironment, androgen receptor (AR) alterations and post-transcriptional modifications, the role of glucocorticoid receptor, and alternative oncogenic signaling that is derepressed on maximum AR inhibition and thus promotes cancer survival and progression.

Conclusions

The mechanisms implicated in the development of resistance to AR inhibition in PCa are multiple and complex, involving virtually all classes of genomic alteration and leading to a host of selective/adaptive responses. Combinational therapeutic approaches targeting both AR signaling and alternative oncogenic pathways may be reasonable for patients with CRPC.

Patient summary

We looked for mechanisms related to the progression of PCa in patients undergoing hormonal therapy and treatment with novel drugs targeting the AR. Based on recent data, combining maximal AR inhibition with novel agents targeting other tumor-compensatory, non–AR-related pathways may improve the survival and quality of life of patients with castration-resistant PCa.

Introduction

Prostate cancer (PCa) remains the second leading cause of death due to cancer in Western societies [1]. Usually PCa is diagnosed as localized disease, and its management includes surveillance or radical prostatectomy, radiation therapy, or even combination approaches such as hormonal therapy prior to prostatectomy. Few patients undergoing PCa screening present with metastatic disease, which is also present in ≥10% of unscreened populations at first presentation—usually in bone, the predominant site of advanced and lethal PCa [2]—highlighting critical differences between screened and unscreened populations.

Despite the obvious increase in overall survival of patients with PCa over the past decade, recent data indicate that improvement of survival among patients with metastatic PCa has not significantly contributed to this decline in mortality [3]. Patients with metastatic disease usually receive hormonal therapy that decreases the production of testosterone by the testes. However, after an initial response, which varies significantly among patients, the disease eventually progresses despite the low levels of testosterone in the systemic circulation (<20 ng/dl) [4]. This stage of disease is known as metastatic castration-resistant PCa (mCRPC); the average overall survival is 1.5 yr, with significant variability among patients with lymph node metastasis, bone metastasis, and metastasis in both lymph nodes and bone [3]. Docetaxel and cabazitaxel are the only chemotherapy regimens approved for this stage of the disease [5], [6]. Radium Ra 233 (Xofigo) injection was recently approved for the treatment of patients with castration-resistant PCa (CRPC), symptomatic bone metastases, and no known visceral metastatic disease [7].

Numerous studies during the last decade have highlighted the role of androgen receptor (AR) in the development of mCRPC, showing that despite systemic androgen depletion, AR signaling remains active and supports the survival and growth of PCa cells. Based on these results, two novel agents have been recently evaluated in clinical trials: abiraterone acetate (AA), an inhibitor of androgen synthesis, and enzalutamide, a potent antiandrogen.

AA is a CYP17A1 inhibitor blocking the production of androgens in the testes, adrenal glands, and tumor microenvironment by inhibiting both 17α-hydroxylase and 17,20 lyase activities of the CYP17A1 enzyme [8]. AA has been recently approved for chemotherapy-naive patients with mCRPC, improving overall survival by 4 mo [9].

Enzalutamide is a novel antagonist of AR, inhibiting nuclear translocation, chromatin binding, and interactions with AR coregulators [10]. Enzalutamide prolongs the survival of patients who failed chemotherapy [11], and more recent data suggest that in chemotherapy-naive mCRPC patients, enzalutamide increases overall and progression-free survival and delays the need for chemotherapy [12].

ARN-509, a next-generation antiandrogen, was found to be more effective than enzalutamide in CRPC preclinical models in terms of tumor growth [13]. According to a recent phase 1 clinical trial, ARN-509 is safe and well tolerated and displays dose-proportional pharmacokinetics demonstrating pharmacodynamic and antitumor activity across all dose levels examined [14].

Despite the significant advances in the targeting of AR that have been translated into survival improvement for patients with mCRPC, this stage of disease remains incurable and is associated with significant morbidity and mortality [15]. While the introduction of novel antiandrogens has provided survival benefits through tumor growth inhibition, two critical clinical concerns arise: (1) Which patients really benefit from these agents, and which biomarkers can be used to identify these patients? (2) What alternative approaches can be used if the disease progresses during treatment with these agents?

The aim of this review is to summarize the recent advances in the evaluation of the multiple levels of development of resistance to androgen deprivation and AR inhibition that occur before and after the introduction of novel antiandrogen axis agents. In PCa, the heterogeneity of the diversity of pathways involved demands a critical consideration of numerous possible explanations of these biological events.

Section snippets

Evidence acquisition

PubMed was the data source. Keywords for the search were castrate resistant prostate cancer, abiraterone, enzalutamide resistance mechanisms, resistance to androgen deprivation, AR mutations, amplifications, splice variants, and AR alterations. Papers published before 1990 were excluded from the review, and only English-language papers were included.

Paracrine/autocrine androgen synthesis as a mechanism of resistance to systemic hormonal therapy and novel inhibitors of androgen biosynthesis

It is well documented that in normal prostate tissue and low-grade PCa, the prostate stroma secretes active androgens (eg, dihydrotestosterone [DHT]) and other growth factors supporting the survival and proliferation of overlying epithelium by a paracrine loop [16]. During PCa progression, this paracrine dependence is lost and converted to an autocrine phase in which cancer cells produce numerous factors, including androgens, supporting their own growth and survival [16]. Although circulating

Conclusions

Despite the introduction of novel agents targeting androgen action, mCRPC remains an incurable disease. Increased androgen biosynthesis in the tumor microenvironment and alterations of AR signaling (including AR mutations mainly in the LBD, AR variants, and AR gene amplifications) all play roles in the development of resistance to systemic hormonal therapy. Especially after the introduction of AA and enzalutamide, which maximally decrease AR activity, these and other mechanisms related to

References (96)

  • V.K. Arora et al.

    Glucocorticoid receptor confers resistance to antiandrogens by bypassing androgen receptor blockade

    Cell

    (2013)
  • C. Cai et al.

    Androgen receptor gene expression in prostate cancer is directly suppressed by the androgen receptor through recruitment of lysine-specific demethylase 1

    Cancer Cell

    (2011)
  • D.J. Mulholland et al.

    Cell autonomous role of PTEN in regulating castration-resistant prostate cancer growth

    Cancer Cell

    (2011)
  • T. Muranen et al.

    Inhibition of PI3K/mTOR leads to adaptive resistance in matrix-attached cancer cells

    Cancer Cell

    (2012)
  • J. Abdulghani et al.

    Stat3 promotes metastatic progression of prostate cancer

    Am J Pathol

    (2008)
  • B.S. Knudsen et al.

    Prostate cancer and the met hepatocyte growth factor receptor

    Adv Cancer Res

    (2004)
  • S. Kasai et al.

    Hepatocyte growth factor is a paracrine regulator of rat prostate epithelial growth

    Biochem Biophys Res Commun

    (1996)
  • Y. Loriot et al.

    Targeted therapies in metastatic castration-resistant prostate cancer: beyond the androgen receptor

    Urol Clin North Am

    (2012)
  • R. Siegel et al.

    Cancer statistics, 2013

    CA Cancer J Clin

    (2013)
  • J. Sturge et al.

    Bone metastasis in prostate cancer: emerging therapeutic strategies

    Nat Rev Clin Oncol

    (2011)
  • J.N. Wu et al.

    No improvement noted in overall or cause-specific survival for men presenting with metastatic prostate cancer over a 20-year period

    Cancer

    (2014)
  • W.P. Harris et al.

    Androgen deprivation therapy: progress in understanding mechanisms of resistance and optimizing androgen depletion

    Nat Clin Pract Urol

    (2009)
  • I.F. Tannock et al.

    Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer

    N Engl J Med

    (2004)
  • P.G. Kluetz et al.

    Radium Ra 223 dichloride injection: U.S. Food and Drug Administration drug approval summary

    Clin Cancer Res

    (2014)
  • J.S. de Bono et al.

    Abiraterone and increased survival in metastatic prostate cancer

    N Engl J Med

    (2011)
  • C.J. Ryan et al.

    Abiraterone in metastatic prostate cancer without previous chemotherapy

    N Engl J Med

    (2013)
  • C. Tran et al.

    Development of a second-generation antiandrogen for treatment of advanced prostate cancer

    Science

    (2009)
  • H.I. Scher et al.

    Increased survival with enzalutamide in prostate cancer after chemotherapy

    N Engl J Med

    (2012)
  • T.M. Beer et al.

    Enzalutamide in men with chemotherapy-naive metastatic prostate cancer (mCRPC): results of phase III PREVAIL study [abstract LBA1]

    J Clin Oncol

    (2014)
  • N.J. Clegg et al.

    ARN-509: a novel antiandrogen for prostate cancer treatment

    Cancer Res

    (2012)
  • D.E. Rathkopf et al.

    Phase I study of ARN-509, a novel antiandrogen, in the treatment of castration-resistant prostate cancer

    J Clin Oncol

    (2013)
  • P.J. Toren et al.

    Evolving landscape and novel treatments in metastatic castrate-resistant prostate cancer

    Asian J Androl

    (2013)
  • C.J. Logothetis et al.

    Molecular classification of prostate cancer progression: foundation for marker-driven treatment of prostate cancer

    Cancer Discov

    (2013)
  • R.B. Montgomery et al.

    Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth

    Cancer Res

    (2008)
  • K.H. Chang et al.

    Dihydrotestosterone synthesis bypasses testosterone to drive castration-resistant prostate cancer

    Proc Natl Acad Sci U S A

    (2011)
  • E. Efstathiou et al.

    Effects of abiraterone acetate on androgen signaling in castrate-resistant prostate cancer in bone

    J Clin Oncol

    (2012)
  • T. Saloniemi et al.

    The diversity of sex steroid action: novel functions of hydroxysteroid (17beta) dehydrogenases as revealed by genetically modified mouse models

    J Endocrinol

    (2012)
  • F. Ishizaki et al.

    Androgen deprivation promotes intratumoral synthesis of dihydrotestosterone from androgen metabolites in prostate cancer

    Sci Rep

    (2013)
  • B.H. Lee et al.

    Dysregulation of cholesterol homeostasis in human prostate cancer through loss of ABCA1

    Cancer Res

    (2013)
  • E.A. Mostaghel et al.

    Resistance to CYP17A1 inhibition with abiraterone in castration-resistant prostate cancer: induction of steroidogenesis and androgen receptor splice variants

    Clin Cancer Res

    (2011)
  • Z. Culig et al.

    Mutant androgen receptor detected in an advanced-stage prostatic carcinoma is activated by adrenal androgens and progesterone

    Mol Endocrinol

    (1993)
  • R. De Wit et al.

    Results from a phase 3, randomized, double-blind, multicenter, placebo-controlled trial of orteronel (TAK-700) plus prednisone in patients with metastatic castration-resistant prostate cancer (mCRPC) that has progressed during or following docetaxel-based therapy (ELM-PC 5 trial)

    Clin Adv Hematol Oncol

    (2014)
  • M.E. Taplin et al.

    Androgen receptor mutations in androgen-independent prostate cancer: Cancer and Leukemia Group B study 9663

    J Clin Oncol

    (2003)
  • M.E. Taplin et al.

    Mutation of the androgen-receptor gene in metastatic androgen-independent prostate cancer

    N Engl J Med

    (1995)
  • C.E. Barbieri et al.

    Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer

    Nat Genet

    (2012)
  • C.S. Grasso et al.

    The mutational landscape of lethal castration resistant prostate cancer

    Nature

    (2012)
  • M.E. Taplin et al.

    Selection for androgen receptor mutations in prostate cancers treated with androgen antagonist

    Cancer Res

    (1999)
  • Z. Culig et al.

    Mutant androgen receptor detected in an advanced-stage prostatic carcinoma is activated by adrenal androgens and progesterone

    Mol Endocrinol

    (1993)
  • Cited by (219)

    View all citing articles on Scopus
    View full text