Androgen receptor mutations for precision medicine in prostate cancer

in Endocrine-Related Cancer
Authors:
Masaki Shiota Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

Search for other papers by Masaki Shiota in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-3306-4858
,
Shusuke Akamatsu Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan

Search for other papers by Shusuke Akamatsu in
Current site
Google Scholar
PubMed
Close
,
Shigehiro Tsukahara Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

Search for other papers by Shigehiro Tsukahara in
Current site
Google Scholar
PubMed
Close
,
Shohei Nagakawa Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

Search for other papers by Shohei Nagakawa in
Current site
Google Scholar
PubMed
Close
,
Takashi Matsumoto Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

Search for other papers by Takashi Matsumoto in
Current site
Google Scholar
PubMed
Close
, and
Masatoshi Eto Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

Search for other papers by Masatoshi Eto in
Current site
Google Scholar
PubMed
Close

Correspondence should be addressed to M Shiota: shiota.masaki.101@m.kyushu-u.ac.jp
Free access

Sign up for journal news

Hormonal therapies including androgen deprivation therapy and androgen receptor (AR) pathway inhibitors such as abiraterone and enzalutamide have been widely used to treat advanced prostate cancer. However, treatment resistance emerges after hormonal manipulation in most prostate cancers, and it is attributable to a number of mechanisms, including AR amplification and overexpression, AR mutations, the expression of constitutively active AR variants, intra-tumor androgen synthesis, and promiscuous AR activation by other factors. Although various AR mutations have been reported in prostate cancer, specific AR mutations (L702H, W742L/C, H875Y, F877L, and T878A/S) were frequently identified after treatment resistance emerged. Intriguingly, these hot spot mutations were also revealed to change the binding affinity of ligands including steroids and antiandrogens and potentially result in altered responses to AR pathway inhibitors. Currently, precision medicine utilizing genetic and genomic data to choose suitable treatment for the patient is becoming to play an increasingly important role in clinical practice for prostate cancer management. Since clinical data between AR mutations and the efficacy of AR pathway inhibitors are accumulating, monitoring the AR mutation status is a promising approach for providing precision medicine in prostate cancer, which would be implemented through the development of clinically available testing modalities for AR mutations using liquid biopsy. However, there are few reviews on clinical significance of AR hot spot mutations in prostate cancer. Then, this review summarized the clinical landscape of AR mutations and discussed their potential implication for clinical utilization.

Abstract

Hormonal therapies including androgen deprivation therapy and androgen receptor (AR) pathway inhibitors such as abiraterone and enzalutamide have been widely used to treat advanced prostate cancer. However, treatment resistance emerges after hormonal manipulation in most prostate cancers, and it is attributable to a number of mechanisms, including AR amplification and overexpression, AR mutations, the expression of constitutively active AR variants, intra-tumor androgen synthesis, and promiscuous AR activation by other factors. Although various AR mutations have been reported in prostate cancer, specific AR mutations (L702H, W742L/C, H875Y, F877L, and T878A/S) were frequently identified after treatment resistance emerged. Intriguingly, these hot spot mutations were also revealed to change the binding affinity of ligands including steroids and antiandrogens and potentially result in altered responses to AR pathway inhibitors. Currently, precision medicine utilizing genetic and genomic data to choose suitable treatment for the patient is becoming to play an increasingly important role in clinical practice for prostate cancer management. Since clinical data between AR mutations and the efficacy of AR pathway inhibitors are accumulating, monitoring the AR mutation status is a promising approach for providing precision medicine in prostate cancer, which would be implemented through the development of clinically available testing modalities for AR mutations using liquid biopsy. However, there are few reviews on clinical significance of AR hot spot mutations in prostate cancer. Then, this review summarized the clinical landscape of AR mutations and discussed their potential implication for clinical utilization.

Introduction

Prostate cancer is one of the most frequently diagnosed cancers in developed countries. Characteristically, prostate cancer depends on androgen receptor (AR) signaling for its carcinogenesis, development, and progression (Basu & Tindall 2010). Meanwhile, androgen deprivation therapy has been a standard treatment for metastatic hormone-sensitive prostate cancer (mHSPC) (Shiota & Eto 2016). However, most prostate cancers eventually progress to castration-resistant prostate cancer (CRPC). Recently, novel AR pathway inhibitors (ARPIs) such as the cytochrome P17 (CYP17) inhibitor abiraterone acetate and second-generation antiandrogens such as enzalutamide, apalutamide, and darolutamide have proven to prolong survival in patients with mHSPC and CRPC (Shiota & Eto 2016, Harada et al. 2021).

Prostate cancer can acquire resistance to hormonal treatments including surgical or medical castration (leuprorelin, goserelin, and degarelix), antiandrogens (bicalutamide, flutamide, enzalutamide, apalutamide, and darolutamide), CYP17 inhibitors (abiraterone), and several steroidal agents (estrogen and glucocorticoid), and aberrant activation of AR signaling plays a crucial role in this process (Shiota et al. 2011a,, 2012). Aberrant activation of the AR signaling pathway in prostate cancer has been attributed to a number of mechanisms, including AR amplification and overexpression, AR mutations, expression of constitutively active AR variants, intra-tumor androgen synthesis, and promiscuous AR activation caused by other factors (Shiota et al. 2011a,b, 2012).

Among them, 10–20% of CRPCs are known to possess somatic AR gene mutations (Taplin et al. 2003). Actually, recent studies on whole genome sequencing by next-generation sequencing (NGS) in CRPC have revealed mutational landscape of prostate cancer and showed that AR gene mutations are one of most frequently observed mutated genes (Grasso et al. 2012). Interestingly, mutant AR can promiscuously be activated by adrenal androgens, non-androgenic steroids, and even antiandrogens (Waltering et al. 2012). In addition, clinical data between AR mutations and the efficacy of ARPIs have recently been accumulating (Huang et al. 2022). Thus, the clinical importance of AR mutations has been suggested, and information about the mutation status could contribute to the selection of effective treatment for individual patients. Currently, precision medicine utilizing genetic and genomic data is becoming to play an increasingly important role to choose suitable treatment for the patient in clinical practice for prostate cancer (Malik et al. 2019). However, there are few reviews on clinical significance of AR mutations in prostate cancer. In this review, we summarized the clinical landscape of AR mutations and discussed their potential implication for clinical utilization.

Research history on AR mutation in prostate cancer

In 1990, Veldscholte et al. at Erasmus University discovered a point mutation (T868A in codon 910 of AR cDNA; this mutation is equivalent to T878A in codon 920 of AR cDNA based on the human reference genome Hg19 and codon numbering was based on the human reference genome Hg19 thereafter) in the ligand-binding domain (LBD) of the AR gene in LNCaP prostate cancer cells and reported that this AR mutant was activated in vitro by androgens as well as progesterone, estrogen, and the antiandrogen cyproterone acetate (Table 1) (Veldscholte et al. 1990). Subsequently, AR mutations such as H875Y and L702H/T878A were found in CWR22 and MDA PCa 2a prostate cancer cell lines, respectively (Tan et al. 1997, Zhao et al. 1999).

Table 1

Timeline of research history on somatic mutation in prostate cancer, leading to alteration in the LBD of AR.

Year Institute (country) Discovered findings Reference
1990 Erasmus University (Netherland) T878A mutation in LNCaP prostate cancer cells

Promiscuous activation of mutated AR by steroids and antiandrogens
Veldscholte et al. 1990
1992 Johns Hopkins University (United States) AR mutation in patient’s sample from untreated organ-confined prostate cancer Newmark et al. 1992
1993 University of Innsbruck (Austria) AR mutation in patient’s sample from CRPC Culig et al. 1993
1993 Chiba University (Japan) L702H and T878A mutations in patient’ sample from CRPC Suzuki et al. 1993
1994 Walter Reed Army Medical Center (United States) T878A mutation in multiple cases Gaddipati et al. 1994
1995 University of Massachusetts Medical Center (United States) Multiple AR mutations in patient’ sample from metastatic CRPC Taplin et al. 1995
1996 Flinders University of South Australia (Australia) Association between AR mutation and rapid failure of subsequent hormonal therapies Tilley et al. 1996
1996 Chiba University (Japan) Association between T878A mutation and antiandrogen withdrawal syndrome Suzuki et al. 1996
1997 The University of North Carolina (United States) H875Y mutation in CWR22 prostate cancer cells Tan et al. 1997
1999 University of Massachusetts Cancer Center (United States) Selection for AR mutations in prostate cancers treated with antiandrogen Taplin et al. 1999
1999 Stanford University (United States) L702H and T878A mutations in MDA PCa 2a prostate cancer cells Zhao et al. 1999
2000 Baylor College of Medicine (United States) Association between AR mutations and metastatic disease Marcelli et al. 2000
2000 Stanford University (United States) Cancer cell growth by glucocorticoid through L702H and T878A mutations Zhao et al. 2000
2002 University of California (United States) In-frame tandem duplication of exon 3 in CWR22Rv1 prostate cancer cells, resultingin LBD-deleted AR Tepper et al. 2002
2003 Takeda Chemical Industries, Ltd. (Japan) Association between W742C/L mutations and antiandrogen withdrawal syndrome Hara et al. 2003
2003 CALGB 9663 (United States) Analysis on AR mutation with antiandrogen withdrawal syndrome in a clinical trial Taplin et al. 2003

AR, androgen receptor; CRPC, castration-resistant prostate cancer; LBD, ligand-binding domain.

In 1992, Newmark et al. at Johns Hopkins University reported the first case of a somatic mutation in the LBD of AR (V731M) among 26 specimens of untreated organ-confined prostate cancer (Table 1) (Newmark et al. 1992). Subsequently, Culig et al. at University of Innsbruck, Suzuki et al. at Chiba University, and others found somatic mutations in the tumor tissues from patient who showed refractory to endocrine treatment, indicating that AR mutations were associated with disease progression and resistant to endocrine therapy (Table 1) (Culig et al. 1993, Suzuki et al. 1993, Gaddipati et al. 1994, Taplin et al. 1995, 1999, Suzuki et al. 1996, Tilley et al. 1996, Marcelli et al. 2000, Tepper et al. 2002).

In addition to treatment resistance, AR mutations such as T878A and W742C/L were reported to be associated with antiandrogen withdrawal syndrome (AWS), suggesting the clinical importance of AR mutations in treatment navigation (Table 1) (Suzuki et al. 1996, Hara et al. 2003). Meanwhile, in the Cancer and Leukemia Group B Study 9663, AR mutations were detected in 5 of 48 CRPC tumors, but there was no association between AR mutations and antiandrogen withdrawal response or survival (Table 1) (Taplin et al. 2003). This null result may be because all AR mutations were analyzed together even though each AR mutation may have a different clinical impact, suggesting the importance of evaluating AR mutations individually. In addition, promiscuous activation of mutated AR by steroids and antiandrogens were shown to contribute cancer cell growth non-cognate ligand (Table 1) (Zhao et al. 2000). Thus, it was demonstrated that AR mutations were closely associated with tumor response to endocrine therapy in prostate cancer.

In the 2000s, NGS was developed, and it allowed the sequences of entire genomes or targeted regions of DNA or RNA to be determined. After the discoveries of AR mutations using traditional techniques such as Sanger sequencing, various studies were performed by NGS using tissues and blood from patients, and they revealed the landscape of gene mutations, in which AR mutations were detected reproducibly (Tolkach & Kristiansen 2019).

Mutation landscape of the AR gene in prostate cancer

In 2012, 159 AR mutations were found in prostate cancer tissues according to the AR gene mutations database, and almost all are single-base substitutions due to somatic mutations rather than germline mutations (Gottlieb et al. 2012). Although a substantial minority of mutations arise in N-terminal domain (NTD) and the fewest in the DNA-binding domain, the majority occur in the LBD (approximately 45%) (Gottlieb et al. 2012). Common mutations in the LBD broaden ligand specificity of the AR, which can be activated not only by androgens but also by non-androgenic steroids and antiandrogens (Gottlieb et al. 2012). Meanwhile, some of AR mutations in NTD such as E252G, E255K, and W435L were reported to increase AR activity through various mechanisms such as protein stability, nuclear translocation, intramolecular interaction, and recruitment of coactivator (Han et al. 2005, Steinkamp et al. 2009).

Among them, several mutations in the LBD were frequently and reproducibly detected in tissues and circulating tumor DNA (ctDNA) from patients with prostate cancer by NGS (Table 2). In a study using prostate cancer tissues, T878A (2.5%), L702H (2.3%), W742C (2.3%), H875Y (2.0%), W742L (0.5%), T878S (0.5%), and F877L (0.2%) were detected in 444 tumor tissues from 429 patients with metastatic CRPC (mCRPC) (Abida et al. 2019). Similar detection rates of AR mutations were reported in studies using ctDNA from patients with prostate cancer (Table 2). Among these studies, Ledet et al. performed the largest investigation of AR mutations using Guardant360 among 892 patients with advanced prostate cancer and detected L702H (24.6%), T878A (14.5%), H875Y (11.4%), W742C (8.4%), W742L (3.8%), F877L (2.1%), and T878S (1.6%) (Ledet et al. 2020). Consistently, a meta-analysis revealed that L702H (3.4%), H875Y (4.9%), and T878A (4.4%) were the most prevalent mutations across 1614 patients with CRPC from 21 studies (Snaterse et al. 2022). Similarly, recurrent mutations on these sites (L702H in 57 samples, W742C/L in 36 samples, H875Y in 45 samples, F877L in 5 samples, and T878A/S in 61 samples) were reported from 9087 patients/9377 samples in 23 studies, which was retrieved from cBioPortal (http://www.cbioportal.org/) (Fig. 1). In addition to recurrent mutations in hot spot residues, interestingly, other missense substitution in hot spot residues such as W742F, W742R, and T878G were reported (Armenia et al. 2018, Borgmann et al. 2018, Dong et al. 2021a ). Notably, each hot spot AR mutation was observed commonly among ethnics (Table 2).

Figure 1
Figure 1

The scheme for AR (androgen receptor) gene and protein structures. The human AR gene consists of eight exons, which are coding AR protein, including the N-terminal domain (NTD), DNA-binding domain (DBD), and the ligand-binding domain (LBD). Pathogenic mutations were indicated with their position and sample numbers, which was retrieved from cBioPortal (http://www.cbioportal.org/) at 30 April 2022. A full color version of this figure is available at https://doi.org/10.1530/ERC-22-0140.

Citation: Endocrine-Related Cancer 29, 10; 10.1530/ERC-22-0140

Table 2

Frequency of driver androgen receptor mutations in the ligand-binding domain.

Report Sequenced material Patients number Clinical stage Treatment Analysis method Countries AR mutations (corresponding base change by GRCh37/hg19)
L702H (chrX:66931463-AR-T>A) W742L (chrX:66937371-AR-G>T) W742C (chrX:66937372-AR-G>T; chrX:66937372-AR-G>C) H875Y (chrX:66943543-AR-C>T) F877L (chrX:66943549-AR-T>C; chrX:66943551-AR-C>A) T878A (chrX:66943552-AR-A>G) T878S (chrX:66943553-AR-C>G)
Grasso et al. 2012 Tissue at autopsy 50 mCRPC NA NGS United States 1 (2.0%) 0 (0%) 0 (0%) 2 (4.0%) 0 (0%) 3 (6.0%) 0 (0%)
Robinson et al. 2015 Metastatic tissue 150 mCRPC NA NGS United States 7 (4.7%) 0 (0%) 2 (1.3%) 4 (2.7%) 0 (0%) 7 (4.7%) 0 (0%)
Quigley et al. 2018 Tissue 101 mCRPC NA NGS United States 4 (4.0%) 1 (1.0%) 1 (1.0%) 0 (0%) 0 (0%) 3 (3.0%) 0 (0%)
Abida W et al. 2019 Tissue 429 mCRPC Abiraterone or enzalutamide (n = 128) NGS United States 10 (2.3%) 2 (0.5%) 10 (2.3%) 9 (2.0%) 1 (0.2%) 11 (2.5%) 2 (0.5%)
Mateo et al. 2020 Tissue 52 mCRPC NA NGS Europe 2 (3.8%) 0 (0%) 0 (0%) 0 (0%) 1 (1.9%) 1 (1.9%) 0 (0%)
Li et al. 2020 Metastatic tissue 41 mCRPC NA NGS United States 1 (2.4%) 0 (0%) 0 (0%) 1 (2.4%) 0 (0%) 2 (4.9%) 0 (0%)
McKay et al. 2021 Metastatic tissue 65 mCRPC Enzalutamide NGS United States 1 (1.5%) 1 (1.5%) 0 (0%) 1 (1.5%) 0 (0%) 1 (1.5%) 1 (1.5%)
Wyatt et al. 2017 Metastatic tissues(n = 45)

ctDNA(n = 45)
45 mCRPC NA NGS North America 4 (8.9%) 0 (0%) 0 (0%) 3 (6.7%) 0 (0%) 0 (0%) 0 (0%)
Gong et al. 2021 Metastatic tissue (n = 29)

ctDNA (n = 109)
129 mCRPC NA NGS China 4 (3.1%) 0 (0%) 0 (0%) 1 (0.8%) 0 (0%) 8 (6.2%) 0 (0%)
Jiang et al. 2010 CTC 35 CRPC NA Sanger United States 0 (0%) 2 (5.7%) 0 (0%) 2 (5.7%) 0 (0%) 1 (2.9%) 0 (0%)
Azad et al. 2015

Lallous et al. 2016
ctDNA 62 CRPC Abiraterone

Enzalutamide

Other agents
NGS Canada 0 (0%) 0 (0%) 0 (0%) 9 (14.5%) 1 (1.6%) 6 (9.7%) 0 (0%)
Romanel et al. 2015 ctDNA 97 CRPC Abiraterone NGS Europe 10 (4.6%) 0 (0%) 0 (0%) 20 (9.2%) 0 (0%) 7 (3.2%) 1 (0.5%)
Wyatt et al. 2016 ctDNA 65 mCRPC Enzalutamide NGS Canada 16 (13.4%) 2 (1.7%) 1 (0.8%) 12 (10.1%) 1 (0.8%) 10 (8.4%) 0 (0%)
Rathkopf et al. 2017 ctDNA 97 CRPC Apalutamide NGS United States 0 (0%) 0 (0%) 1 (1.1%) 1 (1.1%) 2 (2.2%) 3 (3.2%) 0 (0%)
De Laere et al. 2017 ctDNA 30 CRPC Abiraterone (n = 16)

Enzalutamide (n = 2)
NGS Europe 2 (6.7%) 0 (0%) 0 (0%) 2 (6.7%) 0 (0%) 0 (0%) 0 (0%)
Annala et al. 2018 ctDNA 202 mCRPC Abiraterone (n = 101)

Enzalutamide (n = 101)
NGS Canada 0 (0%) 0 (0%) 3 (1.5%) 0 (0%) 9 (4.5%) 0 (0%) 0 (0%)
Patsch et al. 2019 ctDNA 15 mCRPC Chemotherapy NGS United States 0 (0%) 0 (0%) 0 (0%) 2 (13.3%) 0 (0%) 5 (33.3%) 0 (0%)
Torquato et al. 2019 ctDNA 62 mCRPC Enzalutamide (n = 25)

Abiraterone (n = 35)

Enzalutamide and abiraterone (n = 2)
NGS United States 4 (6.5%) 0 (0%) 1 (1.6%) 3 (4.8%) 0 (0%) 3 (4.8%) 0 (0%)
Sumiyoshi et al. 2019 ctDNA 102 CRPC Abiraterone or enzalutamide (n = 38) ddPCR Japan 2 (2.0%) 4 (3.9%) 5 (4.9%) 9 (8.8%) 1 (1.0%) 9 (8.8%) 0 (0%)
Fettke et al. 2020 ctDNA 67 mCRPC ARPIs (n = 41)

Taxane (n = 26)
NGS Austraria 2 (3.0%) 0 (0%) 2 (3.0%) 9 (13.4%) 0 (0%) 3 (4.5%) 0 (0%)
Ledet et al. 2020 ctDNA 892 Advanced prostate cancer (predominantly CRPC) NA NGS (Guadant360) United States 219 (24.6%) 34 (3.8%) 75 (8.4%) 102 (11.4%) 19 (2.1%) 129 (14.5%) 14 (1.6%)
Moses et al. 2020 ctDNA 33 mCRPC Testosterone cypionate NGS (Guadant360) United States 1 (3.0%) 1 (3.0%) 2 (6.1%) 0 (0%) 0 (0%) 1 (3.0%) 0 (0%)
Jayaram et al. 2021 ctDNA 151 mCRPC Abiraterone NGS Europe 2 (1.3%) 0 (0%) 0 (0%) 0 (0%) 0 (0%) 6 (4.0%) 0 (0%)
Dong et al. 2021a ctDNA 292 mCRPC Abiraterone (n = 92)

Docetaxel (n = 40)
NGS China 11 (3.8%) 13 (4.5%) 19 (6.5%) 5 (1.7%) 0 (0%) 20 (6.8%) 1 (0.3%)
Mizuno et al. 2021 ctDNA 111 CRPC Abiraterone or enzalutamide NGS Japan 3 (3.0%) 3 (3.0%) 3 (3.0%) 2 (2.0%) 0 (0%) 4 (4.0%) 0 (0%)
Total 3386 306 (9.0%) 63 (1.9%) 125 (3.7%) 199 (5.9%) 35 (1.0%) 243 (7.2%) 19 (0.6%)

AR, androgen receptor; CRPC, castration-resistant prostate cancer; ctDNA, cirtulating tumor DNA, ddPCR, droplet digital PCR; NA, not available; NGS, next-generation sequencer.

Changes of binding affinity attributable to AR mutation

Missense mutations in the LBD of AR can alter the binding affinity of ligands and result in activation by ligands other than testosterone and dihydrotestosterone (DHT). As indicated in Fig. 2A, the residues of hot spot mutation are located close to the binding site of the cognate ligand testosterone (Lallous et al. 2016). Lallous et al. determined the in vitro effects of four antiandrogens (enzalutamide, hydroxyflutamide, bicalutamide, and apalutamide) and four steroids (DHT, progesterone, estradiol, and hydrocortisone) on AR variants with mutations in the LBD (Lallous et al. 2016). Intriguingly, as presented in Table 3, several mutants responded differentially to antiandrogens and steroids (Lallous et al. 2016). In addition, recent studies demonstrated an antagonistic effect of darolutamide on AR mutants (Moilanen et al. 2015, Sugawara et al. 2019, Lallous et al. 2021). Because abiraterone is administered with prednisone and it blocks the catalysis of progesterone into androstenedione, abiraterone may result in increased levels of glucocorticoid and progesterone (Attard et al. 2008). Therefore, the alterations of steroid levels following abiraterone treatment may affect tumor response through aberrant AR signaling caused by AR mutation.

Figure 2
Figure 2

Androgen receptor protein structures in complex with testosterone (TES) (A), bicalutamide (B), and enzalutamide (C). Reproduced, under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), from Lallous et al. (2016) (https://doi.org/10.1186/s13059-015-0864-1).A full color version of this figure is available at https://doi.org/10.1530/ERC-22-0140.

Citation: Endocrine-Related Cancer 29, 10; 10.1530/ERC-22-0140

Table 3

Pharmacological profiles of driver androgen receptor mutations to antiandrogens and steroids.

AR mutations Bicalutamide Flutamide Enzalutamide Apalutamide Darolutamide DHT (EC < 1 nM) Estradiol (EC < 100 nM) Progesterone (EC < 1 nM) Hydrocortisone (EC < 100 nM)
L702H Partial agonist Partial agonist Antagonist Antagonist Antagonist Less sensitive Less sensitive Less sensitive Sensitive
W742L Agonist Partial agonist Antagonist Antagonist Antagonist Less sensitive Less sensitive Less sensitive Less sensitive
W742C Agonist Partial agonist Antagonist Antagonist Antagonist Less sensitive Less sensitive Less sensitive Less sensitive
H875Y Partial agonist Agonist Partial agonist Partial agonist Antagonista Sensitive Sensitive Less sensitive Less sensitive
F877L Antagonist Partial agonist Partial agonist Partial agonist Antagonist Sensitive Less sensitive Less sensitive Less sensitive
T878A Partial agonist Partial agonist Partial agonist Partial agonist Antagonist Sensitive Less sensitive Sensitive Less sensitive
T878S Partial agonist Partial agonist Partial agonist Partial agonist Antagonista Sensitive Less sensitive Sensitive Less sensitive

aBased on data in H875Y/F877L and F877L/T878A (Lallous et al. 2021).

DHT, dihydrotestosterone.

Among the residues of hot spot mutation, L702 and W742 are located close to the binding site of testosterone, which may result in an altered activation by DHT (Table 3). As well, W742 and T878 are located close to the binding site of bicalutamide, and then mutations in these sites may lead to an altered function of bicalutamide from antagonist to partial agonist or agonist (Fig. 2B and Table 3). Similarly, H875, F877, and T878 are located close to the binding site of enzalutamide, and then mutations in these sites may lead to an altered function of enzalutamide from antagonist to partial agonist or agonist (Fig. 2C and Table 3). Interestingly, W742 is located close to the binding site of bicalutamide, but not enzalutamide, and then bicalutamide becomes agnostic to W742L/C mutations while enzalutamide remains antagonistic (Fig. 2B, C and Table 3). Similarly, F877 is located close to the binding site of enzalutamide, but not bicalutamide, and then enzalutamide becomes partial agnostic to F877L mutation while bicalutamide remains antagonistic (Fig. 2B, C and Table 3). In addition, W742L mutation was shown to change its formation into agonistic by bicalutamide but remains antagonistic by darolutamide (Sugawara et al. 2019). Thus, the structures of ligands such as steroids and antiandrogens is critical to their function to mutated AR. Actually, since enzalutamide and apalutamide have very similar molecular structure, they show similar pharmacological profiles to mutated AR (Table 3) (Tran et al. 2009, Clegg et al. 2012).

Clinical implications of AR mutation for precision medicine

Through altered ligand affinity, AR mutations of the LBD are believed to affect the clinical outcome of hormonal therapy. As presented in Table 4, the clinical implications of AR mutations regarding the effects of several hormonal treatments were estimated from in vitro data on antiandrogens and steroids (presented in Table 3). However, because AR L702H and W742L/C mutations were less sensitive to DHT, prostate cancer cells with these AR mutations may have intrinsic non-dependency on AR (Table 4).

Table 4

Expected response to androgen receptor pathway inhibitors by driver androgen receptor mutations.

AR mutations Bicalutamide Flutamide Enzalutamide Apalutamide Darolutamide Abiraterone Preferred treatment Non-preferred treatment
L702H Rb Rb Sa Sa Sa R Enzalutamide, apalutamide, darolutamide, dexamethasone Abiraterone plus prednisone/prednisolone
W742L/C R Rb Sa Sa Sa Sa Enzalutamide, apalutamide, darolutamide†, abiraterone Bicalutamide
H875Y Rb R R R S R Darolutamide Flutamide, enzalutamide, estrogen agent
F877L S R R R S S Bicalutamide, darolutamide, abiraterone Flutamide, enzalutamide
T878A/S Rb R Rb Rb S R Darolutamide, dexamethasone† Enzalutamide, abiraterone plus prednisone/prednisolone

Bold indicates clinically supported data.

aPossible intrinsic resistance due to low-dependency on DHT; bSeveral studies indicate sensitivity (Taplin et al. 1999, Urushibara et al. 2007, Tran et al. 2009, van de Wijngaart et al. 2010, Terada et al. 2010, Clegg et al. 2012, Liu et al. 2012); †Preferred agent (Romero-Laorden et al. 2018, Lallous et al. 2021).

R, resistance; S, sensitive.

Abiraterone

Several AR mutations including L702H, T878A, and T878S were demonstrated to emerge in ctDNA after treatment with abiraterone (Romanel et al. 2015). Consistently, detection of the AR L702H or T878A mutation in ctDNA before treatment was associated with poor responses to abiraterone, with zero of four patients exhibiting decreases of prostate-specific antigen (PSA) levels (Romanel et al. 2015). Similarly, another study demonstrated that only one of eight post-docetaxel patients carrying the AR L702H or T878A mutation experienced PSA decline during abiraterone treatment (Conteduca et al. 2017). Similarly, the H875Y mutation was reported to emerge after progression on abiraterone treatment (Azad et al. 2015). Consistently, the H875Y and T878A mutations were shown to be activated by progesterone in an experimental model (Hou et al. 2022). In contrary, two patients carrying the H875Y mutation without AR amplification displayed excellent PSA responses to abiraterone, raising controversy regarding the clinical significance of the H875Y mutation in abiraterone treatment and then requiring further investigation (Torquato et al. 2019). Thus, AR L702H, T878A, and T878S are clinically associated with abiraterone resistance, consistent with the in vitro data (Table 4).

Enzalutamide and apalutamide

Similarly, several AR mutations including H875Y and T878A emerged in ctDNA after treatment with enzalutamide (Romanel et al. 2015). Consistently, AR mutation (H875Y or T878A) was associated with a short duration of response to enzalutamide and an increased fraction of AR mutants during enzalutamide treatment (Wyatt et al. 2016). Meanwhile, the fraction change of AR mutants (L702H, W742L, or W742C) during enzalutamide treatment varied among patients, suggesting the AR L702H and W742L/C mutations did not reflect enzalutamide resistance (Wyatt et al. 2016). Intriguingly, enzalutamide and apalutamide are antagonistic to AR W742L/C mutants, but they had less potent suppressive effects on the activation of AR W742L/C mutants by androgen than darolutamide, suggesting darolutamide may be the preferred agent for patents carrying AR W742L/C mutations (Sugawara et al. 2019). In addition, AR F877L is another mutation associated with enzalutamide resistance. First, the AR F877L mutation was found to emerge after enzalutamide resistance in LNCaP prostate cancer cells (Joseph et al. 2013, Korpal et al. 2013). However, this mutation was rarely (0–2%) found among patients with CRPC (Table 2). Wyatt et al. detected the AR F877L mutation in only one patient after progression during enzalutamide treatment (Wyatt et al. 2016). Thus, AR H875Y, F877L, and T878A are clinically associated with enzalutamide resistance, consistent with the in vitro data (Table 4). However, darolutamide was shown to be more effective to these mutants than enzalutamide in an experimental model (Borgmann et al. 2018).

Glucocorticoid

The phase II SWITCH study indicated that switching from prednisone to dexamethasone during treatment with abiraterone resulted in PSA and radiographic responses in some patients with mCRPC (Romero-Laorden et al. 2018). Interestingly, this study evaluated AR copy number alteration and AR mutations including L702H and T878A in ctDNA, and the best treatment effect was observed in patients with the AR T878A mutation who received abiraterone plus dexamethasone. Conversely, poor responses were noted patients with AR amplification (Romero-Laorden et al. 2018). Consistently, a patient with multiple AR mutations (L702H and T878A) displayed PSA decline after abiraterone plus dexamethasone treatment, but no response to abiraterone plus prednisone was noted (Torquato et al. 2019). Because the AR T878A mutant is activated by progesterone and this effect is enhanced by abiraterone but suppressed by continuous low-dose dexamethasone, abiraterone plus dexamethasone may be effective against mutated AR (Attard et al. 2012). Intriguingly, the AR L702H mutant was stimulated by cortisol and prednisolone but not by dexamethasone, whereas the AR T878A mutant was not stimulated by cortisol, prednisolone, and dexamethasone in an in vitro assay (Snaterse et al. 2022). Thus, patients with AR mutations (L702H or T878A) may benefit from switching the steroid from prednisone/prednisolone to dexamethasone.

Estrogen

In addition, the H875Y mutation was indicated to be sensitive to estradiol in in vitro experiments (Lallous et al. 2016). In a case report, a patient carrying the H875Y mutation experienced rapid PSA elevation after treatment with the estrogen agent diethylstilbestrol (Vasudevamurthy et al. 2017). Therefore, the administration of estrogens should be avoided in patients carrying the H875Y mutation.

AWS

Furthermore, AWS may be observed after antiandrogen withdrawal if antiandrogens are agonistic to the AR mutation. After the seminal discovery by Suzuki et al. of chlormadinone acetate withdrawal syndrome in patients carrying the T878A mutation, several studies described the association between AR mutation and AWS (Suzuki et al. 1996, Lorente et al. 2015, Leone et al. 2018). The agonistic effects of flutamide on the H875Y or T878S mutant and those of bicalutamide on the W742L or W742C mutant were demonstrated by in vitro and in vivo experiments, suggesting these mutations may cause AWS after the withdrawal of each antiandrogen (Fenton et al. 1997, Taplin et al. 1999, Hara et al. 2003, Terada et al. 2010). Meanwhile, ASW after the withdrawal of enzalutamide and abiraterone has been reported at a frequency of 0–5%, and this finding may also be attributable to the agonistic effect of each agent on AR mutants (Leone et al. 2018). Therefore, information on AR mutations may be helpful for predicting the possibility of AWS in addition to the effectiveness of ARPIs.

Taken together, the clinical relevance of AR mutants including L702H, W742L, W742C, H875Y, F877L, T878A, and T878S has been demonstrated, as cBioPortal currently defines these mutants as driver missense mutations. However, since high-level evidence on clinical implications of AR mutation lacks currently, further investigations would be required in the future.

Spatiotemporal alterations of AR mutations in prostate cancer

In treatment-naïve prostate cancer, almost no AR mutation was detected in tissues and ctDNA (Cancer Genome Atlas Research Network 2015, Vandekerkhove et al. 2019). Meanwhile, comprehensive analyses in prostate cancer tissues robustly revealed increased genomic alterations including AR mutations after the progression to CRPC (Grasso et al. 2012, Beltran et al. 2013, Robinson et al. 2015, Mateo et al. 2020). Similarly, in the phase II PROPHECY trial, de novo AR somatic mutations were detected in 8 of 49 patients after abiraterone treatment (Gupta et al. 2021). In other phase II trials, the frequency of AR mutations detected in ctDNA was reported to change after abiraterone or enzalutamide exposure (Annala et al. 2021, McKay et al. 2021). Thus, AR mutations were detected more frequently after the emergence of resistance to hormonal therapy, but AR mutants vulnerable to treatment may regress in post-treatment samples.

In addition to temporal changes of AR mutations, spatial intra-tumor heterogeneity has been reported (Boutros et al. 2015, Su et al. 2018, VanderWeele et al. 2019). AR aberrations occurred on the off-trunk, indicating that AR genomic changes occur after tumorigenesis and vary spatially in an intra-patient manner, potentially leading to spatial heterogeneity in patients (Gundem et al. 2015). In fact, AR mutations were reported to be extremely rare in lung metastasis without liver involvement compared to the incidence of bone-only or liver metastasis, suggesting that the status of AR mutation appears to differ among tumor sites (Gong et al. 2021).

Clinical development of tests for detecting AR mutations

To date, various mechanisms other than AR mutations, including AR gene amplification, variant forms of AR, and non-AR pathways, have been described to induce resistance to hormonal treatments including novel ARPIs in prostate cancer (Shiota et al. 2012). Therefore, comprehensive assessment of the aberrations leading to treatment resistance is necessary for the accurate prediction of treatment response. Currently, clinical comprehensive assessments of genomic alterations can be performed via cancer genome profiling using NGS-based assays for target gene regions such as Foundation CDx, Foundation Liquid, and MSK-IMPACT. However, cancer genome profiling is expensive and time-consuming. Thus, its use is limited clinically, and performing multiple tests in individual patients is economically unfeasible.

Clinical information on the AR mutation status in addition to the AR copy number and AR variant status are critically important for selecting effective treatments for individual patients. Actually, recent analysis by liquid biopsy demonstrated that co-occurrence of AR mutation and amplification was observed in 5 (33.3%) among 15 clinical CRPC patients with AR mutation, in addition to co-occurrence with other genomic alterations (Kohli et al. 2020, Dong et al. 2021b ). As well, AR mutation in addition to APC and KIT mutations was shown to increase in mCRPC compared to mHSPC (Kohli et al. 2020). Because the AR mutation status and other alterations thus change throughout the clinical course, it is necessary to monitor AR alterations during the clinical course. Another clinical issue in detecting AR mutations is that obtaining tissues for sequencing is often difficult because of the invasiveness of the procedures and limited access to suitable tissues, especially when tissues are needed sequentially after treatment. In addition, spatial heterogeneity leads to difficulty in capturing all genomic alterations in the host. Therefore, liquid biopsy is a promising approach for monitoring the AR mutation status, and it is more feasible and comprehensive than tissue biopsy. In fact, Wyatt et al. demonstrated the high concordance between metastatic tissue and ctDNA and high detection of AR mutations in ctDNA. In particular, six AR mutations were detected in both metastatic tissue and ctDNA, whereas one AR mutation was detected in ctDNA but not in metastatic tissue (Wyatt et al. 2017). However, the amount of samples for analysis via liquid biopsy is limited. Therefore, multiplex assay or NGS for AR mutations using various technologies that enable the detection of multiple AR mutations simultaneously needs to be developed for clinical application. Droplet digital PCR technology has the advantages of lower costs and a better limit of detection than conventional NGS. Therefore, technologies based on droplet digital PCR may be more suitable for clinical application to permit multiple testing with high sensitivity for monitoring the AR mutation status.

Conclusion

Currently, clinical findings on the association between AR mutations and treatment response to various ARPIs in prostate cancer are limited as most of the studies cited in this review investigated low numbers of patients, and continuous accumulation of knowledge in this field would lead to improved prediction of treatment responses to hormonal manipulation associated with the AR mutation status. Monitoring AR mutation through the clinical course of advanced prostate cancer is clinically important for ensuring the accuracy of the selection and timing of treatments for individual patients. At present, there is no clinically available test for detecting AR mutations excluding cancer genome profiling. Thus, clinically available tests for AR mutation represent an unmet need in advanced prostate cancer treatment.

Declaration of interest

Masaki Shiota received honoraria from Janssen Pharmaceutical, AstraZeneca, and Astellas Pharma and research funding support from Daiichi Sankyo. Shusuke Akamatsu received honoraria from Janssen, AstraZeneca, Astellas, Sanofi, Takeda, and Chugai, and research grants from AstraZeneca, Astellas, and Tosoh. Takashi Matsumoto is a faculty member of the collaborative research division with Denka. Masatoshi Eto received honoraria from Ono Pharmaceutical, Takeda Pharmaceutical, Novartis Pharma, Pfizer, Bristol-Myers Squibb, Janssen Pharmaceutical, MSD, and Merck Biopharma and research funding support from Sanofi, Bayer Yakuhin, Astellas Pharma, Ono Pharmaceutical, and Takeda Pharmaceutical.

Funding

This work was supported in part by a Kakenhi Grant (21K09347) from the Ministry of Education, Culture, Sports, Science and Technology of Japan (MEXT), Japan.

Acknowledgements

The authors apologize to authors whose works were inadvertently overlooked or could not be cited because of space constraints. They thank Joe Barber Jr., PhD, from Edanz (https://jp.edanz.com/ac) for editing a draft of this manuscript.

References

  • Abida W, Cyrta J, Heller G, Prandi D, Armenia J, Coleman I, Cieslik M, Benelli M, Robinson D & Van Allen EM et al.2019 Genomic correlates of clinical outcome in advanced prostate cancer. PNAS 116 1142811436. (https://doi.org/10.1073/pnas.1902651116)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Annala M, Vandekerkhove G, Khalaf D, Taavitsainen S, Beja K, Warner EW, Sunderland K, Kollmannsberger C, Eigl BJ & Finch D et al.2018 Circulating tumor DNA genomics correlate with resistance to abiraterone and enzalutamide in prostate cancer. Cancer Discovery 8 444457. (https://doi.org/10.1158/2159-8290.CD-17-0937)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Annala M, Taavitsainen S, Khalaf DJ, Vandekerkhove G, Beja K, Sipola J, Warner EW, Herberts C, Wong A & Fu S et al.2021 Evolution of castration-resistant prostate cancer in ctDNA during sequential androgen receptor pathway inhibition. Clinical Cancer Research 27 46104623. (https://doi.org/10.1158/1078-0432.CCR-21-1625)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Armenia J, Wankowicz SAM, Liu D, Gao J, Kundra R, Reznik E, Chatila WK, Chakravarty D, Han GC & Coleman I et al.2018 The long tail of oncogenic drivers in prostate cancer. Nature Genetics 50 645651. (https://doi.org/10.1038/s41588-018-0078-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Attard G, Reid AH, Yap TA, Raynaud F, Dowsett M, Settatree S, Barrett M, Parker C, Martins V & Folkerd E et al.2008 Phase I clinical trial of a selective inhibitor of CYP17, abiraterone acetate, confirms that castration-resistant prostate cancer commonly remains hormone driven. Journal of Clinical Oncology 26 45634571. (https://doi.org/10.1200/JCO.2007.15.9749)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Attard G, Reid AH, Auchus RJ, Hughes BA, Cassidy AM, Thompson E, Oommen NB, Folkerd E, Dowsett M & Arlt W et al.2012 Clinical and biochemical consequences of CYP17A1 inhibition with abiraterone given with and without exogenous glucocorticoids in castrate men with advanced prostate cancer. Journal of Clinical Endocrinology and Metabolism 97 507516. (https://doi.org/10.1210/jc.2011-2189)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Azad AA, Volik SV, Wyatt AW, Haegert A, Le Bihan S, Bell RH, Anderson SA, McConeghy B, Shukin R & Bazov J et al.2015 Androgen receptor gene aberrations in circulating cell-free DNA: biomarkers of therapeutic resistance in castration-resistant prostate cancer. Clinical Cancer Research 21 23152324. (https://doi.org/10.1158/1078-0432.CCR-14-2666)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Basu S & Tindall DJ 2010 Androgen action in prostate cancer. Hormones and Cancer 1 223228. (https://doi.org/10.1007/s12672-010-0044-4)

  • Beltran H, Yelensky R, Frampton GM, Park K, Downing SR, MacDonald TY, Jarosz M, Lipson D, Tagawa ST & Nanus DM et al.2013 Targeted next-generation sequencing of advanced prostate cancer identifies potential therapeutic targets and disease heterogeneity. European Urology 63 920926. (https://doi.org/10.1016/j.eururo.2012.08.053)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Borgmann H, Lallous N, Ozistanbullu D, Beraldi E, Paul N, Dalal K, Fazli L, Haferkamp A, Lejeune P & Cherkasov A et al.2018 Moving towards precision urologic oncology: targeting enzalutamide-resistant prostate cancer and mutated forms of the androgen receptor using the novel inhibitor darolutamide (ODM-201). European Urology 73 48. (https://doi.org/10.1016/j.eururo.2017.08.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Boutros PC, Fraser M, Harding NJ, de Borja R, Trudel D, Lalonde E, Meng A, Hennings-Yeomans PH, McPherson A & Sabelnykova VY et al.2015 Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nature Genetics 47 736745. (https://doi.org/10.1038/ng.3315)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cancer Genome Atlas Research Network 2015 The molecular taxonomy of primary prostate. Cancer Cell 163 10111025. (https://doi.org/10.1016/j.cell.2015.10.025)

  • Clegg NJ, Wongvipat J, Joseph JD, Tran C, Ouk S, Dilhas A, Chen Y, Grillot K, Bischoff ED & Cai L et al.2012 ARN-509: a novel antiandrogen for prostate cancer treatment. Cancer Research 72 14941503. (https://doi.org/10.1158/0008-5472.CAN-11-3948)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Conteduca V, Wetterskog D, Sharabiani MTA, Grande E, Fernandez-Perez MP, Jayaram A, Salvi S, Castellano D, Romanel A & Lolli C et al.2017 Androgen receptor gene status in plasma DNA associates with worse outcome on enzalutamide or abiraterone for castration-resistant prostate cancer: a multi-institution correlative biomarker study. Annals of Oncology 28 15081516. (https://doi.org/10.1093/annonc/mdx155)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Culig Z, Hobisch A, Cronauer MV, Cato AC, Hittmair A, Radmayr C, Eberle J, Bartsch G & Klocker H 1993 Mutant androgen receptor detected in an advanced-stage prostatic carcinoma is activated by adrenal androgens and progesterone. Molecular Endocrinology 7 15411550. (https://doi.org/10.1210/mend.7.12.8145761)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • De Laere B, van Dam PJ, Whitington T, Mayrhofer M, Diaz EH, Van den Eynden G, Vandebroek J, Del-Favero J, Van Laere S & Dirix L et al.2017 Comprehensive profiling of the androgen receptor in liquid biopsies from castration-resistant prostate cancer reveals novel intra-AR structural variation and splice variant expression patterns. European Urology 72 192200. (https://doi.org/10.1016/j.eururo.2017.01.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dong B, Fan L, Yang B, Chen W, Li Y, Wu K, Zhang F, Dong H, Cheng H & Pan J et al.2021a Use of circulating tumor DNA for the clinical management of metastatic castration-resistant prostate cancer: a multicenter, real-world study. Journal of the National Comprehensive Cancer Network 19 905914. (https://doi.org/10.6004/jnccn.2020.7663)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dong X, Zheng T, Zhang M, Dai C, Wang L, Wang L, Zhang R, Long Y, Wen D & Xie F et al.2021b Circulating cell-free DNA-based detection of tumor suppressor gene copy number loss and its clinical implication in metastatic prostate cancer. Frontiers in Oncology 11 720727. (https://doi.org/10.3389/fonc.2021.720727)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fenton MA, Shuster TD, Fertig AM, Taplin ME, Kolvenbag G, Bubley GJ & Balk SP 1997 Functional characterization of mutant androgen receptors from androgen-independent prostate cancer. Clinical Cancer Research 3 13831388.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fettke H, Kwan EM, Docanto MM, Bukczynska P, Ng N, Graham LK, Mahon K, Hauser C, Tan W & Wang XH et al.2020 Combined cell-free DNA and RNA profiling of the androgen receptor: clinical utility of a novel multianalyte liquid biopsy assay for metastatic prostate cancer. European Urology 78 173180. (https://doi.org/10.1016/j.eururo.2020.03.044)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gaddipati JP, McLeod DG, Heidenberg HB, Sesterhenn IA, Finger MJ, Moul JW & Srivastava S 1994 Frequent detection of codon 877 mutation in the androgen receptor gene in advanced prostate cancers. Cancer Research 54 28612864.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gong Y, Fan L, Fei X, Zhu Y, Du X, He Y, Pan J, Dong B & Xue W 2021 Targeted next-generation sequencing reveals heterogenous genomic features in viscerally metastatic prostate cancer. Journal of Urology 206 279288. (https://doi.org/10.1097/JU.0000000000001731)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gottlieb B, Beitel LK, Nadarajah A, Paliouras M & Trifiro M 2012 The androgen receptor gene mutations database: 2012 update. Human Mutation 33 887894. (https://doi.org/10.1002/humu.22046)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, Quist MJ, Jing X, Lonigro RJ & Brenner JC et al.2012 The mutational landscape of lethal castration-resistant prostate cancer. Nature 487 239243. (https://doi.org/10.1038/nature11125)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gundem G, Van Loo P, Kremeyer B, Alexandrov LB, Tubio JMC, Papaemmanuil E, Brewer DS, Kallio HML, Högnäs G & Annala M et al.2015 The evolutionary history of lethal metastatic prostate cancer. Nature 520 353357. (https://doi.org/10.1038/nature14347)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gupta S, Halabi S, Kemeny G, Anand M, Giannakakou P, Nanus DM, George DJ, Gregory SG & Armstrong AJ 2021 Circulating tumor cell genomic evolution and hormone therapy outcomes in men with metastatic castration-resistant prostate cancer. Molecular Cancer Research 19 10401050. (https://doi.org/10.1158/1541-7786.MCR-20-0975)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Han G, Buchanan G, Ittmann M, Harris JM, Yu X, Demayo FJ, Tilley W & Greenberg NM 2005 Mutation of the androgen receptor causes oncogenic transformation of the prostate. PNAS 102 11511156. (https://doi.org/10.1073/pnas.0408925102)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hara T, Miyazaki J, Araki H, Yamaoka M, Kanzaki N, Kusaka M & Miyamoto M 2003 Novel mutations of androgen receptor: a possible mechanism of bicalutamide withdrawal syndrome. Cancer Research 63 149153.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Harada K, Shiota M, Minato A, Matsumoto M, Tomisaki I, Fujisawa M & Fujimoto N 2021 Treatment strategies for metastatic castration-sensitive prostate cancer: from ‘all-comers’ to ‘personalized’ approach. OncoTargets and Therapy 14 29672974. (https://doi.org/10.2147/OTT.S306345)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hou Z, Huang S, Mei Z, Chen L, Guo J, Gao Y, Zhuang Q, Zhang X, Tan Q & Yang T et al.2022 Inhibiting 3βHSD1 to eliminate the oncogenic effects of progesterone in prostate cancer. Cell Reports. Medicine 3 100561. (https://doi.org/10.1016/j.xcrm.2022.100561)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang J, Lin B & Li B 2022 Anti-androgen receptor therapies in prostate cancer: A brief update and perspective. Frontiers in Oncology 12 865350. (https://doi.org/10.3389/fonc.2022.865350)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jayaram A, Wingate A, Wetterskog D, Wheeler G, Sternberg CN, Jones R, Berruti A, Lefresne F, Lahaye M & Thomas S et al.2021 Plasma tumor gene conversions after one cycle abiraterone acetate for metastatic castration-resistant prostate cancer: a biomarker analysis of a multicenter international trial. Annals of Oncology 32 726735. (https://doi.org/10.1016/j.annonc.2021.03.196)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jiang Y, Palma JF, Agus DB, Wang Y & Gross ME 2010 Detection of androgen receptor mutations in circulating tumor cells in castration-resistant prostate cancer. Clinical Chemistry 56 14921495. (https://doi.org/10.1373/clinchem.2010.143297)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Joseph JD, Lu N, Qian J, Sensintaffar J, Shao G, Brigham D, Moon M, Maneval EC, Chen I & Darimont B et al.2013 A clinically relevant androgen receptor mutation confers resistance to second-generation antiandrogens enzalutamide and ARN-509. Cancer Discovery 3 10201029. (https://doi.org/10.1158/2159-8290.CD-13-0226)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kohli M, Tan W, Zheng T, Wang A, Montesinos C, Wong C, Du P, Jia S, Yadav S & Horvath LG et al.2020 Clinical and genomic insights into circulating tumor DNA-based alterations across the spectrum of metastatic hormone-sensitive and castrate-resistant prostate cancer. EBiomedicine 54 102728. (https://doi.org/10.1016/j.ebiom.2020.102728)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Korpal M, Korn JM, Gao X, Rakiec DP, Ruddy DA, Doshi S, Yuan J, Kovats SG, Kim S & Cooke VG et al.2013 An F876L mutation in androgen receptor confers genetic and phenotypic resistance to MDV3100 (enzalutamide). Cancer Discovery 3 10301043. (https://doi.org/10.1158/2159-8290.CD-13-0142)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lallous N, Volik SV, Awrey S, Leblanc E, Tse R, Murillo J, Singh K, Azad AA, Wyatt AW & LeBihan S et al.2016 Functional analysis of androgen receptor mutations that confer anti-androgen resistance identified in circulating cell-free DNA from prostate cancer patients. Genome Biology 17 10. (https://doi.org/10.1186/s13059-015-0864-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lallous N, Snow O, Sanchez C, Parra Nuñez AK, Sun B, Hussain A, Lee J, Morin H, Leblanc E & Gleave ME et al.2021 Evaluation of darolutamide (ODM201) efficiency on androgen receptor mutants reported to date in prostate cancer patients. Cancers 13 2939. (https://doi.org/10.3390/cancers13122939)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ledet EM, Lilly MB, Sonpavde G, Lin E, Nussenzveig RH, Barata PC, Yandell M, Nagy RJ, Kiedrowski L & Agarwal N et al.2020 Comprehensive analysis of AR alterations in circulating tumor DNA from patients with advanced prostate cancer. Oncologist 25 327333. (https://doi.org/10.1634/theoncologist.2019-0115)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leone G, Tucci M, Buttigliero C, Zichi C, Pignataro D, Bironzo P, Vignani F, Scagliotti GV & Di Maio M 2018 Antiandrogen withdrawal syndrome (AAWS) in the treatment of patients with prostate cancer. Endocrine-Related Cancer 25 R1R9. (https://doi.org/10.1530/ERC-17-0355)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li Y, Yang R, Henzler CM, Ho Y, Passow C, Auch B, Carreira S, Nava Rodrigues D, Bertan C & Hwang TH et al.2020 Diverse AR gene rearrangements mediate resistance to androgen receptor inhibitors in metastatic prostate cancer. Clinical Cancer Research 26 19651976. (https://doi.org/10.1158/1078-0432.CCR-19-3023)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu B, Geng G, Lin R, Ren C & Wu JH 2012 Learning from estrogen receptor antagonism: structure-based identification of novel antiandrogens effective against multiple clinically relevant androgen receptor mutants. Chemical Biology and Drug Design 79 300312. (https://doi.org/10.1111/j.1747-0285.2011.01290.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lorente D, Mateo J, Zafeiriou Z, Smith AD, Sandhu S, Ferraldeschi R & de Bono JS 2015 Switching and withdrawing hormonal agents for castration-resistant prostate cancer. Nature Reviews. Urology 12 3747. (https://doi.org/10.1038/nrurol.2014.345)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Malik A, Srinivasan S & Batra J 2019 A New Era of prostate cancer precision medicine. Frontiers in Oncology 9 1263. (https://doi.org/10.3389/fonc.2019.01263)

  • Marcelli M, Ittmann M, Mariani S, Sutherland R, Nigam R, Murthy L, Zhao Y, DiConcini D, Puxeddu E & Esen A et al.2000 Androgen receptor mutations in prostate cancer. Cancer Research 60 944949.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mateo J, Seed G, Bertan C, Rescigno P, Dolling D, Figueiredo I, Miranda S, Nava Rodrigues D, Gurel B & Clarke M et al.2020 Genomics of lethal prostate cancer at diagnosis and castration resistance. Journal of Clinical Investigation 130 17431751. (https://doi.org/10.1172/JCI132031)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McKay RR, Kwak L, Crowdis JP, Sperger JM, Zhao SG, Xie W, Werner L, Lis RT, Zhang Z & Wei XX et al.2021 Phase II multicenter study of enzalutamide in metastatic castration-resistant prostate cancer to identify mechanisms driving resistance. Clinical Cancer Research 27 36103619. (https://doi.org/10.1158/1078-0432.CCR-20-4616)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mizuno K, Sumiyoshi T, Okegawa T, Terada N, Ishitoya S, Miyazaki Y, Kojima T, Katayama H, Fujimoto N & Hatakeyama S et al.2021 Clinical impact of detecting low-frequency variants in cell-free DNA on treatment of castration-resistant prostate cancer. Clinical Cancer Research 27 61646173. (https://doi.org/10.1158/1078-0432.CCR-21-2328)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moilanen AM, Riikonen R, Oksala R, Ravanti L, Aho E, Wohlfahrt G, Nykänen PS, Törmäkangas OP, Palvimo JJ & Kallio PJ 2015 Discovery of ODM-201, a new-generation androgen receptor inhibitor targeting resistance mechanisms to androgen signaling-directed prostate cancer therapies. Scientific Reports 5 12007. (https://doi.org/10.1038/srep12007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moses M, Koksal U, Ledet E, Manogue C, Cotogno P, Lewis B, Layton J, Sartor AO & Barata P 2020 Evaluation of the genomic alterations in the androgen receptor gene during treatment with high-dose testosterone for metastatic castrate-resistant prostate cancer. Oncotarget 11 1521. (https://doi.org/10.18632/oncotarget.27408)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Newmark JR, Hardy DO, Tonb DC, Carter BS, Epstein JI, Isaacs WB, Brown TR & Barrack ER 1992 Androgen receptor gene mutations in human prostate cancer. PNAS 89 63196323. (https://doi.org/10.1073/pnas.89.14.6319)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Patsch K, Matasci N, Soundararajan A, Diaz P, Agus DB, Ruderman D & Gross ME 2019 Monitoring dynamic cytotoxic chemotherapy response in castration-resistant prostate cancer using plasma cell-free DNA (cfDNA). BMC Research Notes 12 275. (https://doi.org/10.1186/s13104-019-4312-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Quigley DA, Dang HX, Zhao SG, Lloyd P, Aggarwal R, Alumkal JJ, Foye A, Kothari V, Perry MD & Bailey AM et al.2018 Genomic hallmarks and structural variation in metastatic prostate cancer. Cell 174 758.e9769.e9. (https://doi.org/10.1016/j.cell.2018.06.039)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rathkopf DE, Smith MR, Ryan CJ, Berry WR, Shore ND, Liu G, Higano CS, Alumkal JJ, Hauke R & Tutrone RF et al.2017 Androgen receptor mutations in patients with castration-resistant prostate cancer treated with apalutamide. Annals of Oncology 28 22642271. (https://doi.org/10.1093/annonc/mdx283)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, Mosquera JM, Montgomery B, Taplin ME, Pritchard CC & Attard G et al.2015 Integrative clinical genomics of advanced prostate cancer. Cell 161 12151228. (https://doi.org/10.1016/j.cell.2015.05.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Romanel A, Gasi Tandefelt D, Conteduca V, Jayaram A, Casiraghi N, Wetterskog D, Salvi S, Amadori D, Zafeiriou Z & Rescigno P et al.2015 Plasma AR and abiraterone-resistant prostate cancer. Science Translational Medicine 7 312re10. (https://doi.org/10.1126/scitranslmed.aac9511)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Romero-Laorden N, Lozano R, Jayaram A, López-Campos F, Saez MI, Montesa A, Gutierrez-Pecharoman A, Villatoro R, Herrera B & Correa R et al.2018 Phase II pilot study of the prednisone to dexamethasone switch in metastatic castration-resistant prostate cancer (mCRPC) patients with limited progression on abiraterone plus prednisone (SWITCH study). British Journal of Cancer 119 10521059. (https://doi.org/10.1038/s41416-018-0123-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shiota M & Eto M 2016 Current status of primary pharmacotherapy and future perspectives toward upfront therapy for metastatic hormone-sensitive prostate cancer. International Journal of Urology 23 360369. (https://doi.org/10.1111/iju.13091)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shiota M, Yokomizo A & Naito S 2011a Increased androgen receptor transcription: a cause of castration-resistant prostate cancer and a possible therapeutic target. Journal of Molecular Endocrinology 47 R25R41. (https://doi.org/10.1530/JME-11-0018)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shiota M, Yokomizo A & Naito S 2011b Oxidative stress and androgen receptor signaling in the development and progression of castration-resistant prostate cancer. Free Radical Biology and Medicine 51 13201328. (https://doi.org/10.1016/j.freeradbiomed.2011.07.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shiota M, Yokomizo A & Naito S 2012 Pro-survival and anti-apoptotic properties of androgen receptor signaling by oxidative stress promote treatment resistance in prostate cancer. Endocrine-Related Cancer 19 R243R253. (https://doi.org/10.1530/ERC-12-0232)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Snaterse G, Mies R, van Weerden WM, French PJ, Jonker JW, Houtsmuller AB, van Royen ME, Visser JA & Hofland J 2022 Androgen receptor mutations modulate activation by 11-oxygenated androgens and glucocorticoids. Prostate Cancer and Prostatic Diseases [epub]. (https://doi.org/10.1038/s41391-022-00491-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Steinkamp MP, O’Mahony OA, Brogley M, Rehman H, Lapensee EW, Dhanasekaran S, Hofer MD, Kuefer R, Chinnaiyan A & Rubin MA et al.2009 Treatment-dependent androgen receptor mutations in prostate cancer exploit multiple mechanisms to evade therapy. Cancer Research 69 44344442. (https://doi.org/10.1158/0008-5472.CAN-08-3605)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Su F, Zhang W, Zhang D, Zhang Y, Pang C, Huang Y, Wang M, Cui L, He L & Zhang J et al.2018 Spatial intratumor genomic heterogeneity within localized prostate cancer revealed by single-nucleus sequencing. European Urology 74 551559. (https://doi.org/10.1016/j.eururo.2018.06.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sugawara T, Baumgart SJ, Nevedomskaya E, Reichert K, Steuber H, Lejeune P, Mumberg D & Haendler B 2019 Darolutamide is a potent androgen receptor antagonist with strong efficacy in prostate cancer models. International Journal of Cancer 145 13821394. (https://doi.org/10.1002/ijc.32242)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sumiyoshi T, Mizuno K, Yamasaki T, Miyazaki Y, Makino Y, Okasho K, Li X, Utsunomiya N, Goto T & Kobayashi T et al.2019 Clinical utility of androgen receptor gene aberrations in circulating cell-free DNA as a biomarker for treatment of castration-resistant prostate cancer. Scientific Reports 9 4030. (https://doi.org/10.1038/s41598-019-40719-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Suzuki H, Sato N, Watabe Y, Masai M, Seino S & Shimazaki J 1993 Androgen receptor gene mutations in human prostate cancer. Journal of Steroid Biochemistry and Molecular Biology 46 759765. (https://doi.org/10.1016/0960-0760(9390316-o)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Suzuki H, Akakura K, Komiya A, Aida S, Akimoto S & Shimazaki J 1996 Codon 877 mutation in the androgen receptor gene in advanced prostate cancer: relation to antiandrogen withdrawal syndrome. Prostate 29 153158. (https://doi.org/10.1002/1097-0045(199609)29:3<153::aid-pros2990290303>3.0.co;2-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tan J, Sharief Y, Hamil KG, Gregory CW, Zang DY, Sar M, Gumerlock PH, deVere White RW, Pretlow TG & Harris SE et al.1997 Dehydroepiandrosterone activates mutant androgen receptors expressed in the androgen-dependent human prostate cancer xenograft CWR22 and LNCaP cells. Molecular Endocrinology 11 450459. (https://doi.org/10.1210/mend.11.4.9906)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Taplin ME, Bubley GJ, Shuster TD, Frantz ME, Spooner AE, Ogata GK, Keer HN & Balk SP 1995 Mutation of the androgen-receptor gene in metastatic androgen-independent prostate cancer. New England Journal of Medicine 332 13931398. (https://doi.org/10.1056/NEJM199505253322101)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Taplin ME, Bubley GJ, Ko YJ, Small EJ, Upton M, Rajeshkumar B & Balk SP 1999 Selection for androgen receptor mutations in prostate cancers treated with androgen antagonist. Cancer Research 59 25112515.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Taplin ME, Rajeshkumar B, Halabi S, Werner CP, Woda BA, Picus J, Stadler W, Hayes DF, Kantoff PW & Vogelzang NJ et al.2003 Androgen receptor mutations in androgen-independent prostate cancer: Cancer and Leukemia Group B Study 9663. Journal of Clinical Oncology 21 26732678. (https://doi.org/10.1200/JCO.2003.11.102)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tepper CG, Boucher DL, Ryan PE, Ma AH, Xia L, Lee LF, Pretlow TG & Kung HJ 2002 Characterization of a novel androgen receptor mutation in a relapsed CWR22 prostate cancer xenograft and cell line. Cancer Research 62 66066614.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Terada N, Shimizu Y, Yoshida T, Maeno A, Kamba T, Inoue T, Nakamura E, Kamoto T & Ogawa O 2010 Antiandrogen withdrawal syndrome and alternative antiandrogen therapy associated with the W741C mutant androgen receptor in a novel prostate cancer xenograft. Prostate 70 252261. (https://doi.org/10.1002/pros.21058)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tilley WD, Buchanan G, Hickey TE & Bentel JM 1996 Mutations in the androgen receptor gene are associated with progression of human prostate cancer to androgen independence. Clinical Cancer Research 2 277285.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tolkach Y & Kristiansen G 2019 An overview of translational prostate cancer cohorts for prognostic and predictive studies. Histopathology 74 161170. (https://doi.org/10.1111/his.13770)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Torquato S, Pallavajjala A, Goldstein A, Toro PV, Silberstein JL, Lee J, Nakazawa M, Waters I, Chu D & Shinn D et al.2019 Genetic alterations detected in cell-free DNA are associated with enzalutamide and abiraterone resistance in castration-resistant prostate cancer. JCO Precision Oncology 3 PO.18.00227. (https://doi.org/10.1200/PO.18.00227)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tran C, Ouk S, Clegg NJ, Chen Y, Watson PA, Arora V, Wongvipat J, Smith-Jones PM, Yoo D & Kwon A et al.2009 Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science 324 787790. (https://doi.org/10.1126/science.1168175)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Urushibara M, Ishioka J, Hyochi N, Kihara K, Hara S, Singh P, Isaacs JT & Kageyama Y 2007 Effects of steroidal and non-steroidal antiandrogens on wild-type and mutant androgen receptors. Prostate 67 799807. (https://doi.org/10.1002/pros.20542)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • van de Wijngaart DJ, Molier M, Lusher SJ, Hersmus R, Jenster G, Trapman J & Dubbink HJ 2010 Systematic structure-function analysis of androgen receptor Leu701 mutants explains the properties of the prostate cancer mutant L701H. Journal of Biological Chemistry 285 50975105. (https://doi.org/10.1074/jbc.M109.039958)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vandekerkhove G, Struss WJ, Annala M, Kallio HML, Khalaf D, Warner EW, Herberts C, Ritch E, Beja K & Loktionova Y et al.2019 Circulating tumor DNA abundance and potential utility in de novo metastatic prostate cancer. European Urology 75 667675. (https://doi.org/10.1016/j.eururo.2018.12.042)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • VanderWeele DJ, Finney R, Katayama K, Gillard M, Paner G, Imoto S, Yamaguchi R, Wheeler D, Lack J & Cam M et al.2019 Genomic heterogeneity within individual prostate cancer foci impacts predictive biomarkers of targeted therapy. European Urology Focus 5 416424. (https://doi.org/10.1016/j.euf.2018.01.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vasudevamurthy AK, Ledet E, Garvey C, Lewis BE & Sartor O 2017 Estrogen-mediated activation of H875Y androgen receptor mutation in a prostate cancer patient. Clinical Genitourinary Cancer 15 e111e113. (https://doi.org/10.1016/j.clgc.2016.07.015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Veldscholte J, Ris-Stalpers C, Kuiper GG, Jenster G, Berrevoets C, Claassen E, van Rooij HC, Trapman J, Brinkmann AO & Mulder E 1990 A mutation in the ligand binding domain of the androgen receptor of human LNCaP cells affects steroid binding characteristics and response to anti-androgens. Biochemical and Biophysical Research Communications 173 534540. (https://doi.org/10.1016/s0006-291x(0580067-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Waltering KK, Urbanucci A & Visakorpi T 2012 Androgen receptor (AR) aberrations in castration-resistant prostate cancer. Molecular and Cellular Endocrinology 360 3843. (https://doi.org/10.1016/j.mce.2011.12.019)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wyatt AW, Azad AA, Volik SV, Annala M, Beja K, McConeghy B, Haegert A, Warner EW, Mo F & Brahmbhatt S et al.2016 Genomic alterations in cell-free DNA and enzalutamide resistance in castration-resistant prostate cancer. JAMA Oncology 2 15981606. (https://doi.org/10.1001/jamaoncol.2016.0494)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wyatt AW, Annala M, Aggarwal R, Beja K, Feng F, Youngren J, Foye A, Lloyd P, Nykter M & Beer TM et al.2017 Concordance of circulating tumor DNA and matched metastatic tissue biopsy in prostate cancer. Journal of the National Cancer Institute 109 djx118. (https://doi.org/10.1093/jnci/djx118)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhao XY, Boyle B, Krishnan AV, Navone NM, Peehl DM & Feldman D 1999 Two mutations identified in the androgen receptor of the new human prostate cancer cell line MDA PCa 2a. Journal of Urology 162 21922199. (https://doi.org/10.1016/s0022-5347(0568158-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhao XY, Malloy PJ, Krishnan AV, Swami S, Navone NM, Peehl DM & Feldman D 2000 Glucocorticoids can promote androgen-independent growth of prostate cancer cells through a mutated androgen receptor. Nature Medicine 6 703706. (https://doi.org/10.1038/76287)

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
  • Abida W, Cyrta J, Heller G, Prandi D, Armenia J, Coleman I, Cieslik M, Benelli M, Robinson D & Van Allen EM et al.2019 Genomic correlates of clinical outcome in advanced prostate cancer. PNAS 116 1142811436. (https://doi.org/10.1073/pnas.1902651116)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Annala M, Vandekerkhove G, Khalaf D, Taavitsainen S, Beja K, Warner EW, Sunderland K, Kollmannsberger C, Eigl BJ & Finch D et al.2018 Circulating tumor DNA genomics correlate with resistance to abiraterone and enzalutamide in prostate cancer. Cancer Discovery 8 444457. (https://doi.org/10.1158/2159-8290.CD-17-0937)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Annala M, Taavitsainen S, Khalaf DJ, Vandekerkhove G, Beja K, Sipola J, Warner EW, Herberts C, Wong A & Fu S et al.2021 Evolution of castration-resistant prostate cancer in ctDNA during sequential androgen receptor pathway inhibition. Clinical Cancer Research 27 46104623. (https://doi.org/10.1158/1078-0432.CCR-21-1625)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Armenia J, Wankowicz SAM, Liu D, Gao J, Kundra R, Reznik E, Chatila WK, Chakravarty D, Han GC & Coleman I et al.2018 The long tail of oncogenic drivers in prostate cancer. Nature Genetics 50 645651. (https://doi.org/10.1038/s41588-018-0078-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Attard G, Reid AH, Yap TA, Raynaud F, Dowsett M, Settatree S, Barrett M, Parker C, Martins V & Folkerd E et al.2008 Phase I clinical trial of a selective inhibitor of CYP17, abiraterone acetate, confirms that castration-resistant prostate cancer commonly remains hormone driven. Journal of Clinical Oncology 26 45634571. (https://doi.org/10.1200/JCO.2007.15.9749)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Attard G, Reid AH, Auchus RJ, Hughes BA, Cassidy AM, Thompson E, Oommen NB, Folkerd E, Dowsett M & Arlt W et al.2012 Clinical and biochemical consequences of CYP17A1 inhibition with abiraterone given with and without exogenous glucocorticoids in castrate men with advanced prostate cancer. Journal of Clinical Endocrinology and Metabolism 97 507516. (https://doi.org/10.1210/jc.2011-2189)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Azad AA, Volik SV, Wyatt AW, Haegert A, Le Bihan S, Bell RH, Anderson SA, McConeghy B, Shukin R & Bazov J et al.2015 Androgen receptor gene aberrations in circulating cell-free DNA: biomarkers of therapeutic resistance in castration-resistant prostate cancer. Clinical Cancer Research 21 23152324. (https://doi.org/10.1158/1078-0432.CCR-14-2666)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Basu S & Tindall DJ 2010 Androgen action in prostate cancer. Hormones and Cancer 1 223228. (https://doi.org/10.1007/s12672-010-0044-4)

  • Beltran H, Yelensky R, Frampton GM, Park K, Downing SR, MacDonald TY, Jarosz M, Lipson D, Tagawa ST & Nanus DM et al.2013 Targeted next-generation sequencing of advanced prostate cancer identifies potential therapeutic targets and disease heterogeneity. European Urology 63 920926. (https://doi.org/10.1016/j.eururo.2012.08.053)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Borgmann H, Lallous N, Ozistanbullu D, Beraldi E, Paul N, Dalal K, Fazli L, Haferkamp A, Lejeune P & Cherkasov A et al.2018 Moving towards precision urologic oncology: targeting enzalutamide-resistant prostate cancer and mutated forms of the androgen receptor using the novel inhibitor darolutamide (ODM-201). European Urology 73 48. (https://doi.org/10.1016/j.eururo.2017.08.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Boutros PC, Fraser M, Harding NJ, de Borja R, Trudel D, Lalonde E, Meng A, Hennings-Yeomans PH, McPherson A & Sabelnykova VY et al.2015 Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nature Genetics 47 736745. (https://doi.org/10.1038/ng.3315)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cancer Genome Atlas Research Network 2015 The molecular taxonomy of primary prostate. Cancer Cell 163 10111025. (https://doi.org/10.1016/j.cell.2015.10.025)

  • Clegg NJ, Wongvipat J, Joseph JD, Tran C, Ouk S, Dilhas A, Chen Y, Grillot K, Bischoff ED & Cai L et al.2012 ARN-509: a novel antiandrogen for prostate cancer treatment. Cancer Research 72 14941503. (https://doi.org/10.1158/0008-5472.CAN-11-3948)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Conteduca V, Wetterskog D, Sharabiani MTA, Grande E, Fernandez-Perez MP, Jayaram A, Salvi S, Castellano D, Romanel A & Lolli C et al.2017 Androgen receptor gene status in plasma DNA associates with worse outcome on enzalutamide or abiraterone for castration-resistant prostate cancer: a multi-institution correlative biomarker study. Annals of Oncology 28 15081516. (https://doi.org/10.1093/annonc/mdx155)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Culig Z, Hobisch A, Cronauer MV, Cato AC, Hittmair A, Radmayr C, Eberle J, Bartsch G & Klocker H 1993 Mutant androgen receptor detected in an advanced-stage prostatic carcinoma is activated by adrenal androgens and progesterone. Molecular Endocrinology 7 15411550. (https://doi.org/10.1210/mend.7.12.8145761)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • De Laere B, van Dam PJ, Whitington T, Mayrhofer M, Diaz EH, Van den Eynden G, Vandebroek J, Del-Favero J, Van Laere S & Dirix L et al.2017 Comprehensive profiling of the androgen receptor in liquid biopsies from castration-resistant prostate cancer reveals novel intra-AR structural variation and splice variant expression patterns. European Urology 72 192200. (https://doi.org/10.1016/j.eururo.2017.01.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dong B, Fan L, Yang B, Chen W, Li Y, Wu K, Zhang F, Dong H, Cheng H & Pan J et al.2021a Use of circulating tumor DNA for the clinical management of metastatic castration-resistant prostate cancer: a multicenter, real-world study. Journal of the National Comprehensive Cancer Network 19 905914. (https://doi.org/10.6004/jnccn.2020.7663)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dong X, Zheng T, Zhang M, Dai C, Wang L, Wang L, Zhang R, Long Y, Wen D & Xie F et al.2021b Circulating cell-free DNA-based detection of tumor suppressor gene copy number loss and its clinical implication in metastatic prostate cancer. Frontiers in Oncology 11 720727. (https://doi.org/10.3389/fonc.2021.720727)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fenton MA, Shuster TD, Fertig AM, Taplin ME, Kolvenbag G, Bubley GJ & Balk SP 1997 Functional characterization of mutant androgen receptors from androgen-independent prostate cancer. Clinical Cancer Research 3 13831388.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fettke H, Kwan EM, Docanto MM, Bukczynska P, Ng N, Graham LK, Mahon K, Hauser C, Tan W & Wang XH et al.2020 Combined cell-free DNA and RNA profiling of the androgen receptor: clinical utility of a novel multianalyte liquid biopsy assay for metastatic prostate cancer. European Urology 78 173180. (https://doi.org/10.1016/j.eururo.2020.03.044)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gaddipati JP, McLeod DG, Heidenberg HB, Sesterhenn IA, Finger MJ, Moul JW & Srivastava S 1994 Frequent detection of codon 877 mutation in the androgen receptor gene in advanced prostate cancers. Cancer Research 54 28612864.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gong Y, Fan L, Fei X, Zhu Y, Du X, He Y, Pan J, Dong B & Xue W 2021 Targeted next-generation sequencing reveals heterogenous genomic features in viscerally metastatic prostate cancer. Journal of Urology 206 279288. (https://doi.org/10.1097/JU.0000000000001731)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gottlieb B, Beitel LK, Nadarajah A, Paliouras M & Trifiro M 2012 The androgen receptor gene mutations database: 2012 update. Human Mutation 33 887894. (https://doi.org/10.1002/humu.22046)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, Quist MJ, Jing X, Lonigro RJ & Brenner JC et al.2012 The mutational landscape of lethal castration-resistant prostate cancer. Nature 487 239243. (https://doi.org/10.1038/nature11125)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gundem G, Van Loo P, Kremeyer B, Alexandrov LB, Tubio JMC, Papaemmanuil E, Brewer DS, Kallio HML, Högnäs G & Annala M et al.2015 The evolutionary history of lethal metastatic prostate cancer. Nature 520 353357. (https://doi.org/10.1038/nature14347)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gupta S, Halabi S, Kemeny G, Anand M, Giannakakou P, Nanus DM, George DJ, Gregory SG & Armstrong AJ 2021 Circulating tumor cell genomic evolution and hormone therapy outcomes in men with metastatic castration-resistant prostate cancer. Molecular Cancer Research 19 10401050. (https://doi.org/10.1158/1541-7786.MCR-20-0975)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Han G, Buchanan G, Ittmann M, Harris JM, Yu X, Demayo FJ, Tilley W & Greenberg NM 2005 Mutation of the androgen receptor causes oncogenic transformation of the prostate. PNAS 102 11511156. (https://doi.org/10.1073/pnas.0408925102)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hara T, Miyazaki J, Araki H, Yamaoka M, Kanzaki N, Kusaka M & Miyamoto M 2003 Novel mutations of androgen receptor: a possible mechanism of bicalutamide withdrawal syndrome. Cancer Research 63 149153.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Harada K, Shiota M, Minato A, Matsumoto M, Tomisaki I, Fujisawa M & Fujimoto N 2021 Treatment strategies for metastatic castration-sensitive prostate cancer: from ‘all-comers’ to ‘personalized’ approach. OncoTargets and Therapy 14 29672974. (https://doi.org/10.2147/OTT.S306345)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hou Z, Huang S, Mei Z, Chen L, Guo J, Gao Y, Zhuang Q, Zhang X, Tan Q & Yang T et al.2022 Inhibiting 3βHSD1 to eliminate the oncogenic effects of progesterone in prostate cancer. Cell Reports. Medicine 3 100561. (https://doi.org/10.1016/j.xcrm.2022.100561)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang J, Lin B & Li B 2022 Anti-androgen receptor therapies in prostate cancer: A brief update and perspective. Frontiers in Oncology 12 865350. (https://doi.org/10.3389/fonc.2022.865350)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jayaram A, Wingate A, Wetterskog D, Wheeler G, Sternberg CN, Jones R, Berruti A, Lefresne F, Lahaye M & Thomas S et al.2021 Plasma tumor gene conversions after one cycle abiraterone acetate for metastatic castration-resistant prostate cancer: a biomarker analysis of a multicenter international trial. Annals of Oncology 32 726735. (https://doi.org/10.1016/j.annonc.2021.03.196)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jiang Y, Palma JF, Agus DB, Wang Y & Gross ME 2010 Detection of androgen receptor mutations in circulating tumor cells in castration-resistant prostate cancer. Clinical Chemistry 56 14921495. (https://doi.org/10.1373/clinchem.2010.143297)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Joseph JD, Lu N, Qian J, Sensintaffar J, Shao G, Brigham D, Moon M, Maneval EC, Chen I & Darimont B et al.2013 A clinically relevant androgen receptor mutation confers resistance to second-generation antiandrogens enzalutamide and ARN-509. Cancer Discovery 3 10201029. (https://doi.org/10.1158/2159-8290.CD-13-0226)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kohli M, Tan W, Zheng T, Wang A, Montesinos C, Wong C, Du P, Jia S, Yadav S & Horvath LG et al.2020 Clinical and genomic insights into circulating tumor DNA-based alterations across the spectrum of metastatic hormone-sensitive and castrate-resistant prostate cancer. EBiomedicine 54 102728. (https://doi.org/10.1016/j.ebiom.2020.102728)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Korpal M, Korn JM, Gao X, Rakiec DP, Ruddy DA, Doshi S, Yuan J, Kovats SG, Kim S & Cooke VG et al.2013 An F876L mutation in androgen receptor confers genetic and phenotypic resistance to MDV3100 (enzalutamide). Cancer Discovery 3 10301043. (https://doi.org/10.1158/2159-8290.CD-13-0142)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lallous N, Volik SV, Awrey S, Leblanc E, Tse R, Murillo J, Singh K, Azad AA, Wyatt AW & LeBihan S et al.2016 Functional analysis of androgen receptor mutations that confer anti-androgen resistance identified in circulating cell-free DNA from prostate cancer patients. Genome Biology 17 10. (https://doi.org/10.1186/s13059-015-0864-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lallous N, Snow O, Sanchez C, Parra Nuñez AK, Sun B, Hussain A, Lee J, Morin H, Leblanc E & Gleave ME et al.2021 Evaluation of darolutamide (ODM201) efficiency on androgen receptor mutants reported to date in prostate cancer patients. Cancers 13 2939. (https://doi.org/10.3390/cancers13122939)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ledet EM, Lilly MB, Sonpavde G, Lin E, Nussenzveig RH, Barata PC, Yandell M, Nagy RJ, Kiedrowski L & Agarwal N et al.2020 Comprehensive analysis of AR alterations in circulating tumor DNA from patients with advanced prostate cancer. Oncologist 25 327333. (https://doi.org/10.1634/theoncologist.2019-0115)

    • PubMed