Skip to main content
Cold Spring Harbor Perspectives in Medicine logoLink to Cold Spring Harbor Perspectives in Medicine
. 2017 Sep;7(9):a030452. doi: 10.1101/cshperspect.a030452

Androgen Signaling in Prostate Cancer

Charles Dai 1,2, Hannelore Heemers 1,2,3,4, Nima Sharifi 1,2,3,4
PMCID: PMC5580512  PMID: 28389515

Abstract

The androgen-signaling axis plays a pivotal role in the pathogenesis of prostate cancer. Since the landmark discovery by Huggins and Hodges, gonadal depletion of androgens has remained a mainstay of therapy for advanced disease. However, progression to castration-resistant prostate cancer (CRPC) typically follows and is largely the result of restored androgen signaling. Efforts to understand the mechanisms behind CRPC have revealed new insights into dysregulated androgen signaling and intratumoral androgen synthesis, which has ultimately led to the development of several novel androgen receptor (AR)-directed therapies for CRPC. However, emergence of resistance to these newer agents has also galvanized new directions in investigations of prereceptor and postreceptor AR regulation. Here, we review our current understanding of AR signaling as it pertains to the biology and natural history of prostate cancer.


Castration-resistant prostate cancer is largely the result of restored androgen signaling after androgen-deprivation therapy. Recent work offers insights into the complexities of dysregulated androgen signaling in prostate cancer.


It has now been more than 70 years since Huggins and Hodges (1941) first exposed the central role of androgen signaling in prostate cancer by showing that orchiectomy induces considerable tumor regression. Their seminal discovery was recognized with the Nobel Prize in Medicine in 1966 and, to this day, gonadal testosterone depletion remains a mainstay of therapy for advanced disease (Mohler et al. 2012). It is now evident that the majority of prostate cancers express the androgen receptor (AR) throughout the course of the disease (Sadi et al. 1991; Ruizeveld de Winter et al. 1994; Attard et al. 2009), and, in recent years, deeper interrogation into the molecular basis of androgen signaling has offered a better understanding of how AR specifically directs cancer cell behavior. Taken together, these findings have solidified the importance of androgen signaling in prostate cancer pathogenesis.

Nevertheless, androgen-deprivation therapy (ADT) by chemical or surgical castration is invariably followed by the recurrence of castration-resistant prostate cancer (CRPC) within a median of 14–20 months (Sharifi et al. 2005). Once thought to be an androgen-independent state, it is now recognized that this is generally not the case (Mohler 2008). Progression to CRPC is typically heralded by a rising prostate-specific antigen (PSA) despite castrate concentrations of testosterone, suggesting that inappropriate restoration of the AR signaling axis remains pivotal to this progressive and lethal form of disease (Scher and Sawyers 2005; Ryan and Tindall 2011). Efforts to identify the mechanisms underlying CRPC have revealed new insights into dysregulated androgen signaling, including how AR may incur gain-of-function through mutations, splice variants, and aberrant coregulation (postreceptor regulation), as well as how intracrine steroidogenesis (prereceptor regulation) critically contributes to tumor progression. This has ultimately led to the development of several novel AR-directed therapies, which have since clinically validated many of these concepts (Sharifi 2010; Chang and Sharifi 2012). In this work, we review our current understanding of the androgen signaling axis as it directly pertains to the biology of prostate cancer in its various stages, highlighting aspects of prereceptor and postreceptor regulation (Ryan and Tindall 2011; Heemers 2014), as well as emerging AR-directed therapeutic strategies and ongoing areas of research.

ANDROGEN BIOSYNTHESIS IN NORMAL MALE PHYSIOLOGY

Androgens play an essential role in the development and maintenance of normal male physiology (Griffin 1992). The biosynthesis of all steroid hormones begins with 27-carbon cholesterol, which undergoes stepwise modification by a small complement of enzymes first to 21-carbon steroids (progestins) and subsequently to 19-carbon androgens (Fig. 1). In normal male physiology, early steps in steroidogenesis occur efficiently in two tissues—the adrenal cortex and the testes—so that these tissues together play a major role in the synthesis of circulating steroids (Sharifi and Auchus 2012). Further downstream reactions in the steroidogenic pathways are then refined by specific isoenzymes in target tissues to meet site-specific requirements.

Figure 1.

Figure 1.

Pathways of androgen biosynthesis in normal physiology and prostate cancer. Key enzymes are denoted next to arrows for each reaction. Specific isoenzymes responsible for particular reactions are discussed in the main text. DHEA, dehydroepiandrosterone; AD, androstenedione; T, testosterone; DHT, dihydrotestosterone.

The testes are responsible for the biosynthesis of the majority of testosterone in circulation, with comparatively minor input from the adrenal glands (Nakamura et al. 2009). In both the zona reticularis of the adrenal cortex and Leydig cells of the testes, steroidogenesis starts with the side-chain cleavage of cholesterol by CYP11A1 (cholesterol side-chain cleavage enzyme, P450scc) to generate pregnenolone. Pregnenolone is then converted by CYP17A1 (17-hydroxylase/17,20-lyase, P450c17) to 17-OH-pregnenolone and subsequently to dehydroepiandrosterone (DHEA). Although much of the nascent DHEA in the adrenal cortex is readily sulfonated by sulfotransferase (SULT2A1) for eventual secretion into circulation, testicular Leydig cells lack SULT2A1 and abundantly express 3β-hydroxysteroid dehydrogenase 2 (3β-HSD2), which enables further downstream metabolism of DHEA to testosterone (Sharifi and Auchus 2012). Two final steps are required for the generation of testosterone in the testes, primarily mediated by 3β-HSD2 and 17β-hydroxysteroid dehydrogenase 3 (17β-HSD3). A requirement for the latter enzyme is shown by loss-of-function mutations that lead to pseudohermaphroditism (Geissler et al. 1994). Following synthesis, testosterone is secreted into serum, in which it is mostly bound to sex hormone–binding globulin (SHBG) and albumin (Dunn et al. 1981; Rosner et al. 1991). The degree of bound and unbound testosterone probably exists at equilibrium, with free testosterone thought to readily undergo cellular uptake through passive diffusion into peripheral tissues (Dunn et al. 1981). Intriguingly, some studies have shown that exogenous administration of testosterone and dihydrotestosterone (DHT) leads to increased levels of serum but not necessarily intraprostatic androgens (Page et al. 2011; Thirumalai et al. 2016), indicating that currently underappreciated mechanisms may be at play to tightly regulate intracellular androgens within a narrow concentration range.

In prostate cells, testosterone may act directly on AR or be irreversibly converted to DHT by 5α-reductase, of which there are two isoenzymes (SRD5A1, SRD5A2) (Russell and Wilson 1994; Zhu and Imperato-McGinley 2009). In particular, SRD5A2 is the predominant enzyme present in benign prostatic tissue that mediates the testosterone→DHT reaction and is necessary for proper development of the male phenotype (Wilson 2001). A loss-of-function mutation in SRD5A2 causes 5α-reductase deficiency, manifesting in pseudohermaphroditism and failure to develop a normal prostate (Imperato-McGinley et al. 1974; Andersson et al. 1991). The requirement for DHT in prostatic growth has also been confirmed through the development of 5α-reductase inhibitors as an effective treatment for benign prostatic hyperplasia (BPH) (Rittmaster 1997; Steers 2001; Marks 2004). Recognizing the potential complement of enzymes that can participate in androgen biosynthesis is essential, because prostate cancers may frequently commandeer this enzymatic machinery to sustain steroidogenesis and fuel tumor growth, particularly following ADT (Stanbrough et al. 2006; Montgomery et al. 2008; Knudsen 2014).

PRERECEPTOR MODULATION OF AR SIGNALING

Androgen synthesis is tightly governed by the hypothalamic–pituitary–gonadal axis. Pulsatile release of hypothalamic gonadotropin-releasing hormone (GnRH) stimulates luteinizing hormone (LH) secretion from the anterior pituitary gland, which signals for the production of testosterone in the testes. Testosterone subsequently exerts negative feedback on the hypothalamus and pituitary gland. The pulsatile nature of GnRH is necessary to sustain continued LH secretion; persistent GnRH stimulation leads to ensuing desensitization, which is the rationale behind administering long-acting GnRH agonists for ADT. Following an initial flare, serum testosterone concentrations are effectively suppressed by GnRH agonists to medically castrate levels of <50 ng/dL (Nishiyama 2014).

Although testosterone is a physiologic AR ligand sharing a similarly high equilibrium affinity as DHT (Wilson and French 1976), DHT is the principal androgen found within the prostatic cell nucleus (Bruchovsky and Wilson 1968) and is approximately 10-fold more potent in the stimulation of AR target genes (Deslypere et al. 1992). This difference is thought to be attributed to the greater hydrophobicity of DHT, which stabilizes the ligand–receptor state through intermolecular interactions and decreases the ligand dissociation rate (Zhou et al. 1995; Askew et al. 2007). Therefore, the principal effect achieved through testosterone depletion is likely attributed to the intraprostatic reduction in DHT. However, despite castrate concentrations of testosterone and an observed tumor response in 80%–90% of patients, incomplete depletion of prostate cancer tissue androgens occurs following ADT. Residual concentrations of intratumoral DHT can remain at 10%–40% of pretreatment levels (Forti et al. 1989; Labrie et al. 1993; Page et al. 2006), even before the development of CRPC (Nishiyama et al. 2004). This is substantial because this concentration range of typically 1 nm remains sufficient to permit AR signaling, AR target gene expression, and tumor growth both in vitro and in vivo (Gregory et al. 1998, 2001; Mohler et al. 2004; Mostaghel et al. 2007). Multiple studies have now corroborated the presence of residual androgens in recurrent tumors after castration (Geller et al. 1978; Titus et al. 2005; Montgomery et al. 2008), together signifying that the persistence of AR signaling likely promotes the emergence of CRPC. This is perhaps unsurprising, given that the onset of CRPC is predictably and near universally accompanied by an increase in PSA, a widely used clinical biomarker expressed by an AR-responsive gene (Ryan et al. 2006). Furthermore, recent therapeutic advances in the Food and Drug Administration (FDA) approval of novel, life-prolonging AR-directed therapies, such as the potent second-generation competitive AR antagonist enzalutamide (Scher et al. 2012; Beer et al. 2014) and the androgen synthesis inhibitor abiraterone acetate (de Bono et al. 2011; Ryan et al. 2015), have provided the highest level of clinical evidence for this evolving paradigm. A number of similar agents are currently under clinical investigation, which could soon add to a growing arsenal of therapeutic options for men with metastatic CRPC (Dellis and Papatsoris 2016).

INTRACRINE ANDROGEN BIOSYNTHESIS IN PROSTATE CANCER

A variety of mechanisms may explain the restoration of competent AR signaling in CRPC. These include AR overexpression and amplification, intracrine androgen synthesis, acquisition of constitutively active AR splice variants, and gain-of-function mutations, deregulated AR coactivators/corepressors that sensitize AR in response to ligand binding, and ligand-independent signaling and redundant downstream cross talk (Sharifi 2013; Ferraldeschi et al. 2015). Of note, these postulated mechanisms are not necessarily mutually exclusive and may arise together under the selective pressure of ADT. Importantly, the persistence of physiologically significant intratumoral androgens despite castration indicates that prereceptor regulation remains central to many of these mechanisms to further fuel tumor growth (Zhang et al. 2016). To support this are observations that castrate tumors often up-regulate key steroidogenic enzymes to utilize alternative sources of androgen synthesis (Holzbeierlein et al. 2004; Stanbrough et al. 2006; Montgomery et al. 2008).

Several possibilities exist for the origin of these intratumoral androgens. The first is the de novo pathway, which begins with cholesterol and requires multiple steps in the synthesis of DHT. This may occur either via the canonical route as described in normal physiology (Sharifi and Auchus 2012), or alternatively via a “backdoor” pathway, which involves intratumoral CYP17A1 activity to convert pregnanes to androgens that are then 5α- and 3-keto-reduced, with eventual terminal conversion to DHT (Fig. 1) (Fiandalo et al. 2014). Whether tumors express the complete repertoire of steroidogenic enzymes required to generate androgens from cholesterol remains to be fully elucidated (Hofland et al. 2010). On the other hand, circulating adrenal androgens, which are abundant in the form of DHEA and a larger depot of sulfated DHEA-S, are readily interconverted to DHT via an abbreviated series of steps (Mostaghel 2013). DHT concentrations in prostatic tissues of castrate men positively correlate with serum DHEA/DHEA-S levels (Page et al. 2006) and treatment with abiraterone acetate markedly reduces serum DHEA concentrations (Attard et al. 2012; Taplin et al. 2014; Mostaghel 2014a), while aptly suppressing intraprostatic androgen levels (Mostaghel et al. 2014). To generate downstream testosterone and DHT, DHEA must first undergo oxidation of its 3β-hydroxyl group and Δ5 to Δ4 isomerization to form androstenedione (AD). This rate-limiting step is catalyzed by 3β-HSD, for which there are two human isoenzymes: 3β-HSD1 and 3β-HSD2. In peripheral tissues, including the prostate, 3β-HSD1 predominates, whereas 3β-HSD2 is expressed preferentially in the adrenal glands and gonads (Simard et al. 2005). Given its unique position within the steroidogenic pathway, 3β-HSD1 is likely a critical enzymatic gatekeeper that confers on tumors the ability to harness adrenal androgens (Evaul et al. 2010). In fact, a gain-of-function missense in 3β-HSD1 has recently been described, which remarkably augments the capacity of this enzyme to drive conversion of DHEA→AD, thereby permitting more efficient DHT synthesis (Chang et al. 2013). This missense arises from a single nucleotide polymorphism (SNP) at position 1245 (A→C), substituting an asparagine for threonine at amino acid position 367. The functional consequence of this alteration, which can occur as either a somatic mutation or germline variant, is an enzyme protein product that is rendered resistant to ubiquitin-mediated degradation, resulting in intracellular accumulation. Notably, it appears that ADT may select for this particular mutation; CRPC tumors from patients who are germline heterozygous variants will not infrequently show loss of heterozygosity or acquire a second variant allele by way of a somatic mutation (Chang et al. 2013). This leads to markedly stable enzyme expression, detailing yet another adaptive mechanism through which tumors may subvert androgen deprivation. Furthermore, inheritance of the gain-of-function HSD3B1(1245C) SNP is associated with rapid resistance and poorer survival after ADT in patients with prostate cancer (Hearn et al. 2016).

The subsequent conversion from AD to DHT requires two additional reactions. In the canonical pathway, AD first forms testosterone through reduction of its 17-keto moiety mediated by 17β-HSD, before 5α-reduction to DHT by SRD5A (Fig. 1). In contrast, an alternative pathway has been described, in which AD can bypass testosterone as an obligate precursor, instead undergoing 5α-reduction to an intermediate 5α-androstanedione (5α-dione), followed by 17-keto reduction to DHT (Chang et al. 2011). In fact, this “5α-dione pathway” appears to be the favored directionality of adrenal androgen flux in virtually all prostate cancer cell lines as well as in sampled metastatic CRPC biopsies from patients (Chang et al. 2011). Furthermore, in contrast to the robust flux of AD→5α-dione, the comparable reaction of testosterone→DHT is relatively inefficient. This paradoxical shift in the preferred precursor for 5α-reduction from testosterone to AD in CRPC tissues may be explained by the differential expression of 5α-reductase isoenzymes in tumors. Expression studies have repeatedly revealed that the transition from benign tissue to high-grade prostate cancers and CRPC is associated with stepwise up-regulation of SRD5A1 and subtotal loss SRD5A2 (Thomas et al. 2008). Given that the optimal substrate for SRD5A1 is AD rather than testosterone (Thigpen et al. 1993), this genotypic switch may specifically herald an acquired ability of tumors to efficiently harness adrenal androgens to circumvent testosterone depletion. Genetic silencing of SRD5A1 in cell lines effectively abolishes the conversion of adrenal androgens to DHT (Chang et al. 2011).

Because 3β-HSD1, 17β-HSD, and SRD5A are all required for the generation of DHT from adrenal androgens, pharmacologic inhibition of these enzyme targets has been an active area of clinical interest. Two 5α-reductase inhibitors are currently available: finasteride, primarily an SRD5A2 inhibitor, and dutasteride, a dual SRD5A1/SRD5A2 inhibitor (Schmidt and Tindall 2011). These agents have been tested in a variety of settings, including in the prevention of prostate cancer (Azzouni and Mohler 2012; Fleshner et al. 2012; Schröder et al. 2013) and as an adjuvant therapy to additionally suppress residual androgens following ADT (Xu et al. 2006; Shah et al. 2009). One challenge to SRD5A inhibition is a concomitant rise in upstream testosterone following blockade, which may rescue AR activity and obscure potential therapeutic efficacy (Rittmaster et al. 2008; Chang et al. 2011).

Inhibition of the 17β-HSD family enzymes instead may potentially overcome this issue. 17β-HSD5 (aldo-keto reductase 1C3 [AKR1C3]) is one particular member of this family, which shows a reductive preference for the conversion of AD→testosterone and is broadly implicated in prostate cancer (Adeniji et al. 2013). Expression levels of AKR1C3 are associated with the highest increase in CRPC relative to primary cancer among profiled steroidogenic enzymes; in one study, 58% of CRPC samples were positively stained for AKR1C3 compared with only 5.6% of primary cancers (Stanbrough et al. 2006). The design of effective AKR1C3 inhibitors is an ongoing area of investigation. Importantly, inhibitors must show enzyme specificity given multiple closely related aldo-keto reductase isoforms, some of which drive other reactions (Byrns et al. 2011; Adeniji et al. 2013).

Further upstream inhibition of 3β-HSD isoenzymes presents as another potentially viable opportunity for additional androgen suppression. In preclinical models, treatment with abiraterone acetate notably reduces activity of not only CYP17A1 but also 3β-HSD (Evaul et al. 2010). Interestingly, the Δ5, 3β-hydroxyl steroidal structure of abiraterone is amenable to direct enzymatic conversion by 3β-HSD to a Δ4, 3-keto congener (D4A), which is an active inhibitor of multiple steroidogenic enzymes, including 3β-HSD, CYP17A1, and SRD5A (Li et al. 2015). Furthermore, D4A antagonizes AR at levels comparable to enzalutamide (Li et al. 2015). A subsequent metabolite of D4A also shows AR agonist activity; the contributory effect of these derivative compounds therefore suggests that pharmacologic blockade of particular metabolic pathways could be a feasible method to limit the production of AR-promoting metabolites, thereby refining the antitumor properties of abiraterone (Li et al. 2016). Given the appreciable role of residual androgen production in driving progression to CRPC, identifying opportunities for intensive and directed suppression of intracrine androgen synthesis remain paramount.

ANDROGEN RECEPTOR STRUCTURE/FUNCTION

The AR is a ligand-dependent nuclear transcription factor (TF) and member of the steroid hormone receptor superfamily (Nuclear Receptors Nomenclature Committee 1999). The gene for AR is located on the X chromosome (q11-12) and expresses a 110-kDa protein that is 919 amino acids in length, encoded by eight exons (Chang et al. 1988; Lubahn et al. 1989; Tilley et al. 1989). Common in resemblance to other nuclear hormone receptors, the structure of AR is comprised of four separate functionally distinct domains: an amino-terminal domain (NTD), a carboxy-terminal ligand-binding domain (LBD), a DNA-binding domain (DBD), and a flexible hinge region, which joins the LBD and the DBD (Fig. 2) (Gelmann 2002; Claessens et al. 2008).

Figure 2.

Figure 2.

The androgen receptor (AR) gene locus and structure of the AR (full-length and AR-V7). The transcript for wild-type AR full-length (FL) includes eight exons, which correspond to the four respective domains of the AR protein (as depicted by color scheme). AR-V7 includes a cryptic exon region (CE3b) at the carboxyl terminus. NTD, Amino-terminal domain; DBD, DNA-binding domain; LBD, ligand-binding domain; NLS, nuclear localization signal.

The main native agonists for AR under normal physiologic conditions are testosterone and DHT. When unoccupied by ligand, AR resides primarily in the cytoplasm, anchored to cytoskeletal elements, and associated in a complex with heat shock proteins (HSP-90, HSP-70, HSP-56) and other chaperone proteins to protect the receptor against degradation (Smith and Toft 2008). Binding of ligand to the cognate receptor causes dissociation from this complex and initiates a sequence of molecular events that eventually leads to AR nuclear translocation and activation of AR target genes (Fig. 3). The LBD is vital to directing this response; this is well illustrated by the fact that a deletion of the LBD renders AR completely unresponsive to androgens (Jenster et al. 1991). Furthermore, the LBD is the target for the most competitive AR antagonists, including enzalutamide (Knudsen and Scher 2009), and is the most frequent site of gain-of-function point mutations (Buchanan et al. 2001). Although AR mutations are relatively infrequent in early-stage hormone-naïve prostate cancers, they are detected in approximately 10%–30% of patients previously treated with first-generation competitive AR antagonists (Taplin et al. 1995, 2003; Wallén et al. 1999). Acquisition of AR mutations can enhance receptor promiscuity, broadening the range of potential endogenous steroid ligands (Culig et al. 1993; Mostaghel 2014b) or imparting the reversal of AR antagonists to agonists (Veldscholte et al. 1992; Culig et al. 1999; Taplin et al. 1999). The latter is the presumed mechanism by which tumors may regress following the withdrawal of AR antagonist therapy (Scher and Kelly 1993; Hara et al. 2003). Furthermore, this mechanism may explain why tumors refractory to select AR antagonists can show continued susceptibility to alternative agents (Tran et al. 2009; Balbas et al. 2013).

Figure 3.

Figure 3.

Prereceptor and receptor-level modulation of androgen receptor (AR) action within the prostate cancer cell. Examples of transcriptional coregulators discussed within the text are depicted but are a limited representation of all potential participating proteins. DHEA, dehydroepiandrosterone; SHBG, sex hormone–binding globulin; T, testosterone; DHT, dihydrotestosterone; HSP, heat shock protein; TF, transcription factor; ARE, androgen response element.

Ligand binding causes a critical conformational change in AR, which not only facilitates the nuclear targeting of AR but also exposes transcriptional activation function 2 (AF-2), a functionally significant hydrophobic binding surface-spanning helices 3, 4, and 12 within the LBD. Through recognition of FxxLF motifs embedded in the NTD (He et al. 2000), AF-2 mediates protein–protein interactions between the carboxyl and amino termini that are necessary for receptor homodimerization, stabilization of the ligand within the ligand-binding pocket, and optimization of AR activity (Doesburg et al. 1997; Berrevoets et al. 1998). AF-2 also enables the recruitment of specific AR cofactors, which bear FxxLF and LxxLL motifs to modulate receptor function (Heery et al. 1997; He et al. 2002). Nuclear translocation is mediated by a bipartite nuclear localization signal (NLS) located within the hinge region, which interacts with cytoskeletal proteins (Ozanne et al. 2000; Thadani-Mulero et al. 2012) to orchestrate the transport of AR via importin-α across the nuclear membrane (Kaku et al. 2008; Ni et al. 2013). Once in the nucleus, AR generally persists in a homodimer localizing to specific recognition sequences designated as androgen response elements (AREs) found within the promoter and enhancer regions of AR target genes (Claessens et al. 2001). Following localization, coregulators, general TFs, and RNA polymerase II are successively recruited to AR to direct the organization of the preinitiation transcriptional complex (Heemers and Tindall 2009).

The DBD of AR is highly conserved and contains two zinc finger domains, through which specificity for DNA binding is determined (Umesono and Evans 1989; Shaffer et al. 2004). The first zinc finger is responsible for interacting with nucleotides within the major groove of DNA, thereby tethering the receptor for the assembly of a transcriptional complex around AR, whereas the second zinc finger coordinates homodimer formation (Shaffer et al. 2004). Notably, a specific sequence of three amino acid residues (Gly-Ser-Val) within the first zinc finger, known as the P(roximal)-box, is conserved across other steroid receptors, including glucocorticoid receptor (GR), progesterone receptor (PR), and mineralocorticoid receptor (MR) (Umesono and Evans 1989). This homology enables other steroid receptors to recognize response elements in common with AR, which bears potentially significant clinical implications. Recent investigation into postenzalutamide resistance in CRPC has revealed that GR up-regulation may reinstate oncogenic programming through the expression of overlapping, albeit not identical, AR-regulated genes (Arora et al. 2013; Sahu et al. 2013). Around 30% of prostate cancers express GR, with this proportion increased under androgen-deprived conditions (Szmulewitz et al. 2012). In preclinical models, treatment with enzalutamide up-regulates GR expression, which is increased considerably more so following the emergence of enzalutamide resistance. Furthermore, dexamethasone can induce enzalutamide resistance in prostate cancer cell lines, which is subsequently reversed by a glucocorticoid antagonist or genetic silencing of GR expression. This newfound reliance on GR, however, presents an inherent challenge for any additional signaling inhibition because, unlike AR, GR signaling is essential for life (Nicolaides et al. 2010). A satisfactory approach to GR pathway blockade may therefore necessitate the identification of suitable downstream targets for inhibition that will not elicit intolerable or life-threatening toxicities (Sharifi 2014; Li et al. 2017).

The NTD contains transcriptional activation function-1 (AF-1), which commands transcriptional activity and is basally suppressed by the LBD (Jenster et al. 1991; Simental et al. 1991). In recent years, a number of truncated AR splice variants (AR-Vs) have been identified and implicated in CRPC (Dehm et al. 2008; Guo et al. 2009; Hu et al. 2009; Sun et al. 2010); these variants all harbor an intact NTD and DBD but reveal notable loss of the carboxy-terminal LBD, leading to the uncoupling of transcriptional control from ligand-dependent induction. It is thought that AR-Vs may emerge through aberrant alternative splicing (Liu et al. 2014) or AR gene rearrangements (Li et al. 2011, 2012) to escape antiandrogen therapies that target the LBD. Although more than 20 AR-Vs have now been confirmed in prostate cancer specimens (Robinson et al. 2015), which show different levels of transcriptional activity and expression (Ware et al. 2014; Lu et al. 2015), AR-V7 is the most commonly detected variant in CRPC (Ware et al. 2014). Truncation of AR-V7 occurs after exon 3 and includes a cryptic exon 3b from an intron into the expressed protein (Fig. 2). AR-V7 is constitutively active, and mRNA levels in circulating tumor cells (CTCs) have been recently found to correlate strikingly with resistance to enzalutamide and abiraterone, suggesting that AR-Vs may serve as a promising biomarker for therapeutic response (Antonarakis et al. 2014). Several preclinical models in which AR-V7 is either expressed endogenously with full-length AR (AR-FL) or exogenously in AR-FL-negative cells show an abrogated androgen requirement and resistance to antiandrogens in the presence of AR-V7 (Hu et al. 2009; Mostaghel et al. 2011; Li et al. 2013; Cao et al. 2014). Furthermore, exposure to ADT and AR-directed therapies may reciprocally induce AR-V7 expression (Watson et al. 2010; Mostaghel et al. 2011). Although it was originally suggested that AR-V7 primarily heterodimerizes with AR-FL to mediate target gene transcription (Watson et al. 2010; Cao et al. 2014), AR-V7 may also alternatively homodimerize to drive AR signaling independently of AR-FL (Chan et al. 2015; Xu et al. 2015). However, in comparison to AR-FL, AR-V levels are generally low (Watson et al. 2010), particularly in tumors treated with new generation hormonal therapies, and expression of AR-FL nearly always co-occurs with the presence of AR-Vs (Lu et al. 2015). Thus, whether AR-Vs are a self-sufficient substitute for AR-FL and whether differential changes in oncogenic transcriptional programming can occur in the presence of AR-Vs remains a topic of interest for further investigation (Lu et al. 2015).

Advances in our knowledge on AR-Vs in prostate cancer progression and the dynamic structure–function relationships of the different AR domains have unveiled alternative approaches to achieve therapeutic inhibition of AR signaling. Among these are AR-directed agents that do not target the LBD. EPI-506 is an NTD inhibitor that can bind both AR-Vs and AR-FL and is currently under evaluation in phase I clinical trials (NCT02606123) (Maughan and Antonarakis 2015). Other potentially attractive therapeutic targets include the DBD (Dalal et al. 2014) and sites of AR cofactor interaction (Ravindranathan et al. 2013). In summary, our progressive understanding of the potential molecular mechanisms through which AR may drive transcriptional programming continues to guide the development of novel strategies to disrupt AR signaling.

ANDROGEN RECEPTOR COREGULATORS

Approximately 300 AR coregulators have now been identified (Heemers and Tindall 2007; DePriest et al. 2016), which can coactivate or corepress AR transactivation and are increasingly recognized to do so in a target-gene-specific manner (Marshall et al. 2003; Agoulnik and Weigel 2009; Heemers et al. 2009; Ianculescu et al. 2012). Within a large class of proteins with diverse cellular functions and characteristics, these coregulators commonly associate with AR to ensure effective transcription of target genes (Fig. 3) (Heemers and Tindall 2007). Coregulators can alter transcriptional activity through modulation of a variety of processes, including (1) AR stabilization, homodimerization, and nuclear translocation, (2) chromatin remodeling and DNA occupancy, (3) recruitment of general TFs, and (4) priming and assembly of the preinitiation transcriptional complex (Heemers and Tindall 2007; Shiota et al. 2011). Among the prototypical and most well-studied coregulators is the p160 coactivator family, comprised of three protein members: SRC1, SRC2 (TIF2), and SRC3. These proteins specifically bind to the AR NTD, influencing transactivation through direct histone acetyltransferase activity, as well as through indirect recruitment of secondary coactivators to induce chromatin remodeling (Chakravarti et al. 1996). A common attribute among many coregulators is the ability to enzymatically modify AR and other components within the local molecular environment, such as histones, transcriptional proteins, and other coregulators, through acetylation, methylation, phosphorylation, SUMOylation, and ubiquitination (Heemers and Tindall 2007, 2009). This, in turn, initiates cellular processes such as proliferation and invasion, driving tumor progression. An example of this relationship is underscored by speckle-type POZ protein (SPOP) missense mutations in prostate cancer (Berger et al. 2011; Barbieri et al. 2012; Grasso et al. 2012). SPOP, which is an E3 ubiquitin ligase normally involved in the degradation and turnover of AR as well as SRC3, may incur mutations that lead to increased AR protein levels and liberation of AR-mediated gene transcription (An et al. 2014; Geng et al. 2014). Interestingly, AR-Vs that lack the hinge region required for interaction with SPOP are resistant to degradation (An et al. 2014). SPOP mutations are common, occurring in up to 11%–13% of primary prostate cancers, and represent a distinct molecular subtype of disease (The Cancer Genome Atlas Research Network 2015).

Androgens have been shown to regulate the expression of ∼30% of coregulators (Heemers et al. 2009, 2010). This response is variable across coregulators and is highly specific to particular AR target genes (Heemers et al. 2009). Furthermore, overexpression of coactivators is associated with increased clinical aggressiveness (Gnanapragasam et al. 2001; Debes et al. 2003; Zhou et al. 2005). The recent development of peptidomimetics (Ravindranathan et al. 2013) and small molecule inhibitors (Wang et al. 2011b, 2014; Asangani et al. 2014), which target these various coregulators offers a promising approach that may yield a new class of therapeutic agents for CRPC. Prototypical examples include SRC-3 and SRC-1 inhibitors (Wang et al. 2011b, 2014), as well as bromodomain and extraterminal (BET) inhibitors, which disrupt target gene activation by preventing the binding of BET subfamily proteins to acetylated chromatin (Asangani et al. 2014, 2016). In addition, the use of innovative molecular screening approaches such as “Chem-seq”—in which biotin-tagged small molecules are captured by ChIP to link candidate compounds to regulated target genes—may increasingly reveal suitable agents to disrupt the transcriptional program of prostate cancer. Overall, efforts to elucidate key AR coregulators have shown an impressive number of potentially actionable proteins involved in the intricate, selective, and dynamic interplay with AR to promote AR signaling (DePriest et al. 2016).

ANDROGEN RECEPTOR ACTION

The classical model of genomic AR signaling involves the recruitment of the ligand-bound steroid receptor to AR-binding sites to activate the AR transcriptome (Nelson et al. 2002; Dehm and Tindall 2006). A compelling link that underpins AR signaling to prostate tumorigenesis is well illustrated through the occurrence of chromosomal rearrangements that generate novel fusions between the androgen-regulatory elements of TMPRSS2 and ETS family of oncogenes (ERG, ETV1) (Tomlins et al. 2005). TMPRSS2-ERG fusions are the most common molecular alteration in prostate cancer, occurring in 40%–50% of tumors (Tomlins et al. 2009; The Cancer Genome Atlas Research Network 2015). These fusions are also recognized in isolated high-grade prostatic intraepithelial neoplasia (HGPIN) lesions (Park et al. 2014), lesions associated with cancer (Perner et al. 2007), as well as benign prostatic epithelial cells after extended exposure to DHT (Berger et al. 2011), suggesting that the acquisition of TMPRSS2-ETS fusions is likely an early carcinogenic event. Moreover, some evidence suggests that androgens themselves can provoke nonrandom fusion events (Lin et al. 2009; Mani et al. 2009). However, other instigators such as activation of the PI3K/Akt pathway may be required in the presence of fusions to fully induce malignant transformation (Carver et al. 2009; King et al. 2009).

The collective AR cistrome appears to undergo extensive reprogramming with malignant transformation and disease progression (Wang et al. 2009; Sharma et al. 2013; Pomerantz et al. 2015a). Large-scale bioinformatics and systems-based initiatives to characterize the genomic regions of global AR occupancy have revealed an incredible degree of complexity and variation to AR-responsive gene regulation (Sharma et al. 2013; Mills 2014). In fact, the interfacing of TF networks may critically dictate a particular AR-binding profile, which is distinctly different between normal and tumor tissue (Pomerantz et al. 2015b) and may be perturbed by the presence of external signaling factors such as inflammatory cytokines (Sharma et al. 2013). Considerable differences also exist between the AR-binding profile of cell lines and that of primary tissue, indicating that a set of genes might be selectively activated through in vivo signaling (Sharma et al. 2013). Among TFs most enriched at AR-binding sites is forkhead box A1 (FOXA1), a pioneer factor that globally facilitates AR action through interaction with AR at the DBD. FOXA1-binding sites are typically found in close proximity to AR-binding sites, with a large amount of overlap between their respective cistromes (Zhao et al. 2014). In experiments, FOXA1 may either augment or antagonize AR signaling depending on the setting (Wang et al. 2011a). Homeobox B13 (HOXB13), a highly lineage-specific factor, which is itself regulated by FOXA1 (McMullin et al. 2010), has also emerged through recognition of its role in hereditable prostate cancer disposition and disease progression (Ewing et al. 2012; Decker and Ostrander 2014). Together, FOXA1 and HOXB13 have been shown to be sufficient in reprogramming the AR cistrome in an immortalized prostate cell line to resemble that of malignancy (Pomerantz et al. 2015a). These findings have highlighted the dynamic and contextually dependent nature of AR binding (Heemers and Tindall 2009).

AR PATHWAY CROSS TALK AND LIGAND-INDEPENDENT ACTIVATION

Evidence also indicates that various growth factor, cytokine, and nonreceptor tyrosine kinase pathways are activated in prostate cancer (Lamont and Tindall 2011). A number of cell surface receptors including epidermal growth factor receptor (EGFR), interleukin (IL)-6 and IL-8 receptors, insulin-like growth factor 1 (IGF-1) receptor, and Her2/neu have been implicated in cross talk with AR to drive ligand-independent signaling or to sensitize AR to subphysiologic androgen concentrations (Mellinghoff et al. 2004; Guo et al. 2006; Ponguta et al. 2008; Dutt and Gao 2009). Intracellular kinases such as mitogen-activated protein kinase (MAPK), as well as its effectors Src and ERK1/2, and PI3K/Akt have also been shown to drive prostate cancer progression (Guo et al. 2006). Many of these proteins are downstream elements of nongenomic AR signaling, which can mediate a proliferation response typically within minutes of ligand stimulation (Lösel and Wehling 2003; Liao et al. 2013) via cytoplasmic and lipid raft–associated AR (Pedram et al. 2007). Although sizable preclinical data exist to suggest a therapeutic benefit with pharmacologically inhibiting these pathways, clinical results have been mostly disappointing to date (Ziada et al. 2004; de Bono et al. 2007; Araujo et al. 2013). Overall, these signaling molecules may represent a larger coordinated and possibly redundant network of signal transduction pathways that act in concert with AR signaling to promote key neoplastic processes.

CONCLUDING REMARKS

Since the work of Huggins and Hodges, major advances have contributed to our understanding of the AR signaling axis in the pathogenesis of prostate cancer. With this also comes a greater appreciation for the complexity of prereceptor and postreceptor AR regulation. Major milestones were achieved with the introduction of abiraterone and enzalutamide in the treatment of CRPC, which has resulted in a significant paradigm shift and renewed interest in intratumoral androgen suppression. However, onset of resistance to these second-generation agents has also galvanized new directions to investigate the mechanisms that may promote this escape. Evolving molecular approaches have revealed key insights into the structural basis of AR function and the dynamic, context-dependent nature of AR transcriptional control. The hope is that these ongoing efforts will translate into greater precision in AR targeting and novel therapeutic options in the near future for men with prostate cancer.

ACKNOWLEDGMENTS

This work is supported by funding from the Howard Hughes Medical Institute Medical Fellows Program (C.D.), National Cancer Institute (CA166440 to H.H. and R01CA168899, R01CA172382, R01CA190289 to N.S.), Prostate Cancer Foundation (Young Investigator Award to H.H. and Challenge Award to N.S.), Howard Hughes Medical Institute Physician-Scientist Early Career Award (N.S.), American Cancer Society Research Scholar Award (12-038-01-CCE to N.S), and Department of Defense PCRP award W81XWH-16-1-0404 (H.H.).

Footnotes

Editors: Michael M. Shen and Mark A. Rubin

Additional Perspectives on Prostate Cancer available at www.perspectivesinmedicine.org

REFERENCES

  1. Adeniji AO, Chen M, Penning TM. 2013. AKR1C3 as a target in castrate resistant prostate cancer. J Steroid Biochem Mol Biol 137: 136–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Agoulnik IU, Weigel NL. 2009. Coactivator selective regulation of androgen receptor activity. Steroids 74: 669–674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. An J, Wang C, Deng Y, Yu L, Huang H. 2014. Destruction of full-length androgen receptor by wild-type SPOP, but not prostate-cancer-associated mutants. Cell Rep 6: 657–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Andersson S, Berman DM, Jenkins EP, Russell DW. 1991. Deletion of steroid 5 α-reductase 2 gene in male pseudohermaphroditism. Nature 354: 159–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, Chen Y, Mohammad TA, Chen Y, Fedor HL, et al. 2014. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med 371: 1028–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Araujo JC, Trudel GC, Saad F, Armstrong AJ, Yu EY, Bellmunt J, Wilding G, McCaffrey J, Serrano SV, Matveev VB, et al. 2013. Docetaxel and dasatinib or placebo in men with metastatic castration-resistant prostate cancer (READY): A randomised, double-blind phase 3 trial. Lancet Oncol 14: 1307–1316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Arora VK, Schenkein E, Murali R, Subudhi SK, Wongvipat J, Balbas MD, Shah N, Cai L, Efstathiou E, Logothetis C, et al. 2013. Glucocorticoid receptor confers resistance to antiandrogens by bypassing androgen receptor blockade. Cell 155: 1309–1322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Asangani IA, Dommeti VL, Wang X, Malik R, Cieslik M, Yang R, Escara-Wilke J, Wilder-Romans K, Dhanireddy S, Engelke C, et al. 2014. Therapeutic targeting of BET bromodomain proteins in castration-resistant prostate cancer. Nature 510: 278–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Asangani IA, Wilder-Romans K, Dommeti VL, Krishnamurthy PM, Apel IJ, Escara-Wilke J, Plymate SR, Navone NM, Wang S, Feng FY, et al. 2016. BET bromodomain inhibitors enhance efficacy and disrupt resistance to AR antagonists in the treatment of prostate cancer. Mol Cancer Res 14: 324–331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Askew EB, Gampe RT, Stanley TB, Faggart JL, Wilson EM. 2007. Modulation of androgen receptor activation function 2 by testosterone and dihydrotestosterone. J Biol Chem 282: 25801–25816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Attard G, Swennenhuis JF, Olmos D, Reid AHM, Vickers E, A’Hern R, Levink R, Coumans F, Moreira J, Riisnaes R, et al. 2009. Characterization of ERG, AR and PTEN gene status in circulating tumor cells from patients with castration-resistant prostate cancer. Cancer Res 69: 2912–2918. [DOI] [PubMed] [Google Scholar]
  12. Attard G, Reid AHM, 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. J Clin Endocrinol Metab 97: 507–516. [DOI] [PubMed] [Google Scholar]
  13. Azzouni F, Mohler J. 2012. Role of 5α-reductase inhibitors in prostate cancer prevention and treatment. Urology 79: 1197–1205. [DOI] [PubMed] [Google Scholar]
  14. Balbas MD, Evans MJ, Hosfield DJ, Wongvipat J, Arora VK, Watson PA, Chen Y, Greene GL, Shen Y, Sawyers CL. 2013. Overcoming mutation-based resistance to antiandrogens with rational drug design. eLife 2: e00499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Barbieri CE, Baca SC, Lawrence MS, Demichelis F, Blattner M, Theurillat JP, White TA, Stojanov P, Van Allen E, Stransky N, et al. 2012. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet 44: 685–689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, Iversen P, Bhattacharya S, Carles J, Chowdhury S, et al. 2014. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med 371: 424–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Berger MF, Lawrence MS, Demichelis F, Drier Y, Cibulskis K, Sivachenko AY, Sboner A, Esgueva R, Pflueger D, Sougnez C, et al. 2011. The genomic complexity of primary human prostate cancer. Nature 470: 214–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Berrevoets CA, Doesburg P, Steketee K, Trapman J, Brinkmann AO. 1998. Functional interactions of the AF-2 activation domain core region of the human androgen receptor with the amino-terminal domain and with the transcriptional coactivator TIF2 (transcriptional intermediary factor2). Mol Endocrinol 12: 1172–1183. [DOI] [PubMed] [Google Scholar]
  19. Bruchovsky N, Wilson JD. 1968. The intranuclear binding of testosterone and 5-α-androstan-17-β-ol-3-one by rat prostate. J Biol Chem 243: 5953–5960. [PubMed] [Google Scholar]
  20. Buchanan G, Greenberg NM, Scher HI, Harris JM, Marshall VR, Tilley WD. 2001. Collocation of androgen receptor gene mutations in prostate cancer. Clin Cancer Res 7: 1273–1281. [PubMed] [Google Scholar]
  21. Byrns MC, Jin Y, Penning TM. 2011. Inhibitors of type 5 17β-hydroxysteroid dehydrogenase (AKR1C3): Overview and structural insights. J Steroid Biochem Mol Biol 125: 95–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Cao B, Qi Y, Zhang G, Xu D, Zhan Y, Alvarez X, Guo Z, Fu X, Plymate SR, Sartor O, et al. 2014. Androgen receptor splice variants activating the full-length receptor in mediating resistance to androgen-directed therapy. Oncotarget 5: 1646–1656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Carver BS, Tran J, Gopalan A, Chen Z, Shaikh S, Carracedo A, Alimonti A, Nardella C, Varmeh S, Scardino PT, et al. 2009. Aberrant ERG expression cooperates with loss of PTEN to promote cancer progression in the prostate. Nat Genet 41: 619–624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Chakravarti D, LaMorte VJ, Nelson MC, Nakajima T, Schulman IG, Juguilon H, Montminy M, Evans RM. 1996. Role of CBP/P300 in nuclear receptor signalling. Nature 383: 99–103. [DOI] [PubMed] [Google Scholar]
  25. Chan SC, Selth LA, Li Y, Nyquist MD, Miao L, Bradner JE, Raj GV, Tilley WD, Dehm SM. 2015. Targeting chromatin binding regulation of constitutively active AR variants to overcome prostate cancer resistance to endocrine-based therapies. Nucleic Acids Res 43: 5880–5897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Chang KH, Sharifi N. 2012. Prostate cancer-from steroid transformations to clinical translation. Nat Rev Urol 9: 721–724. [DOI] [PubMed] [Google Scholar]
  27. Chang CS, Kokontis J, Liao ST. 1988. Molecular cloning of human and rat complementary DNA encoding androgen receptors. Science 240: 324–326. [DOI] [PubMed] [Google Scholar]
  28. Chang K-H, Li R, Papari-Zareei M, Watumull L, Zhao YD, Auchus RJ, Sharifi N. 2011. Dihydrotestosterone synthesis bypasses testosterone to drive castration-resistant prostate cancer. Proc Natl Acad Sci 108: 13728–13733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Chang KH, Li R, Kuri B, Lotan Y, Roehrborn CG, Liu J, Vessella R, Nelson PS, Kapur P, Guo X, et al. 2013. A gain-of-function mutation in DHT synthesis in castration-resistant prostate cancer. Cell 154: 1074–1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Claessens F, Verrijdt G, Schoenmakers E, Haelens A, Peeters B, Verhoeven G, Rombauts W. 2001. Selective DNA binding by the androgen receptor as a mechanism for hormone-specific gene regulation. J Steroid Biochem Mol Biol 76: 23–30. [DOI] [PubMed] [Google Scholar]
  31. Claessens F, Denayer S, Van Tilborgh N, Kerkhofs S, Helsen C, Haelens A. 2008. Diverse roles of androgen receptor (AR) domains in AR-mediated signaling. Nucl Recept Signal 6: e008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. 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. Mol Endocrinol 7: 1541–1550. [DOI] [PubMed] [Google Scholar]
  33. Culig Z, Hoffmann J, Erdel M, Eder IE, Hobisch A, Hittmair A, Bartsch G, Utermann G, Schneider MR, Parczyk K, et al. 1999. Switch from antagonist to agonist of the androgen receptor bicalutamide is associated with prostate tumour progression in a new model system. Br J Cancer 81: 242–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Dalal K, Roshan-Moniri M, Sharma A, Li H, Ban F, Hassona MD, Hessein M, Hsing M, Singh K, LeBlanc E, et al. 2014. Selectively targeting the DNA-binding domain of the androgen receptor as a prospective therapy for prostate cancer. J Biol Chem 289: 26417–26429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Debes JD, Sebo TJ, Lohse CM, Murphy LM, Haugen DAL, Tindall DJ. 2003. p300 in prostate cancer proliferation and progression. Cancer Res 63: 7638–7640. [PubMed] [Google Scholar]
  36. de Bono JS, Bellmunt J, Attard G, Droz JP, Miller K, Flechon A, Sternberg C, Parker C, Zugmaier G, Hersberger-Gimenez V, et al. 2007. Open-label phase II study evaluating the efficacy and safety of two doses of pertuzumab in castrate chemotherapy-naïve patients with hormone-refractory prostate cancer. J Clin Oncol 25: 257–262. [DOI] [PubMed] [Google Scholar]
  37. de Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, Chi KN, Jones RJ, Goodman OB, Saad F, et al. 2011. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med 364: 1995–2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Decker B, Ostrander EA. 2014. Dysregulation of the homeobox transcription factor gene HOXB13: Role in prostate cancer. Pharmgenomics Pers Med 7: 193–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Dehm SM, Tindall DJ. 2006. Molecular regulation of androgen action in prostate cancer. J Cell Biochem 99: 333–344. [DOI] [PubMed] [Google Scholar]
  40. Dehm SM, Schmidt LJ, Heemers HV, Vessella RL, Tindall DJ. 2008. Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Res 68: 5469–5477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Dellis A, Papatsoris AG. 2016. Phase I and II therapies targeting the androgen receptor for the treatment of castration resistant prostate cancer. Expert Opin Investig Drugs 1–11. [DOI] [PubMed] [Google Scholar]
  42. DePriest AD, Fiandalo MV, Schlanger S, Heemers F, Mohler JL, Liu S, Heemers HV. 2016. Regulators of androgen action resource: A one-stop shop for the comprehensive study of androgen receptor action. Database (Oxford) 10.1093/database/bav125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Deslypere JP, Young M, Wilson JD, McPhaul MJ. 1992. Testosterone and 5 α-dihydrotestosterone interact differently with the androgen receptor to enhance transcription of the MMTV-CAT reporter gene. Mol Cell Endocrinol 88: 15–22. [DOI] [PubMed] [Google Scholar]
  44. Doesburg P, Kuil CW, Berrevoets CA, Steketee K, Faber PW, Mulder E, Brinkmann AO, Trapman J. 1997. Functional in vivo interaction between the amino-terminal, transactivation domain and the ligand binding domain of the androgen receptor. Biochemistry 36: 1052–1064. [DOI] [PubMed] [Google Scholar]
  45. Dunn JF, Nisula BC, Rodbard D. 1981. Transport of steroid hormones: Binding of 21 endogenous steroids to both testosterone-binding globulin and corticosteroid-binding globulin in human plasma. J Clin Endocrinol Metab 53: 58–68. [DOI] [PubMed] [Google Scholar]
  46. Dutt SS, Gao AC. 2009. Molecular mechanisms of castration-resistant prostate cancer progression. Future Oncol 5: 1403–1413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Evaul K, Li R, Papari-Zareei M, Auchus RJ, Sharifi N. 2010. 3β-hydroxysteroid dehydrogenase is a possible pharmacological target in the treatment of castration-resistant prostate cancer. Endocrinology 151: 3514–3520. [DOI] [PubMed] [Google Scholar]
  48. Ewing CM, Ray AM, Lange EM, Zuhlke KA, Robbins CM, Tembe WD, Wiley KE, Isaacs SD, Johng D, Wang Y, et al. 2012. Germline mutations in HOXB13 and prostate-cancer risk. N Engl J Med 366: 141–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Ferraldeschi R, Welti J, Luo J, Attard G, de Bono JS. 2015. Targeting the androgen receptor pathway in castration-resistant prostate cancer: Progresses and prospects. Oncogene 34: 1745–1757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Fiandalo MV, Wilton J, Mohler JL. 2014. Roles for the backdoor pathway of androgen metabolism in prostate cancer response to castration and drug treatment. Int J Biol Sci 10: 596–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Fleshner NE, Lucia MS, Egerdie B, Aaron L, Eure G, Nandy I, Black L, Rittmaster RS. 2012. Dutasteride in localised prostate cancer management: The REDEEM randomised, double-blind, placebo-controlled trial. Lancet 379: 1103–1111. [DOI] [PubMed] [Google Scholar]
  52. Forti G, Salerno R, Moneti G, Zoppi S, Fiorelli G, Marinoni T, Natali A, Costantini A, Serio M, Martini L. 1989. Three-month treatment with a long-acting gonadotropin-releasing hormone agonist of patients with benign prostatic hyperplasia: Effects on tissue androgen concentration, 5α-reductase activity and androgen receptor content. J Clin Endocrinol Metab 68: 461–468. [DOI] [PubMed] [Google Scholar]
  53. Geissler WM, Davis DL, Wu L, Bradshaw KD, Patel S, Mendonca BB, Elliston KO, Wilson JD, Russell DW, Andersson S. 1994. Male pseudohermaphroditism caused by mutations of testicular 17β-hydroxysteroid dehydrogenase 3. Nat Genet 7: 34–39. [DOI] [PubMed] [Google Scholar]
  54. Geller J, Albert J, Loza D, Geller S, Stoeltzing W, de la Vega D. 1978. DHT concentrations in human prostate cancer tissue. J Clin Endocrinol Metab 46: 440–444. [DOI] [PubMed] [Google Scholar]
  55. Gelmann EP. 2002. Molecular biology of the androgen receptor. J Clin Oncol 20: 3001–3015. [DOI] [PubMed] [Google Scholar]
  56. Geng C, Rajapakshe K, Shah SS, Shou J, Eedunuri VK, Foley C, Fiskus W, Rajendran M, Chew SA, Zimmermann M, et al. 2014. Androgen receptor is the key transcriptional mediator of the tumor suppressor SPOP in prostate cancer. Cancer Res 74: 5631–5643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Gnanapragasam VJ, Leung HY, Pulimood AS, Neal DE, Robson CN. 2001. Expression of RAC 3, a steroid hormone receptor co-activator in prostate cancer. Br J Cancer 85: 1928–1936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Grasso CS, Wu Y-M, 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: 239–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Gregory CW, Hamil KG, Kim D, Hall SH, Pretlow TG, Mohler JL, French FS. 1998. Androgen receptor expression in androgen-independent prostate cancer is associated with increased expression of androgen-regulated genes. Cancer Res 58: 5718–5724. [PubMed] [Google Scholar]
  60. Gregory CW, Johnson RT, Mohler JL, French FS, Wilson EM. 2001. Androgen receptor stabilization in recurrent prostate cancer is associated with hypersensitivity to low androgen. Cancer Res 61: 2892–2898. [PubMed] [Google Scholar]
  61. Griffin JE. 1992. Androgen resistance—The clinical and molecular spectrum. N Engl J Med 326: 611–618. [DOI] [PubMed] [Google Scholar]
  62. Guo Z, Dai B, Jiang T, Xu K, Xie Y, Kim O, Nesheiwat I, Kong X, Melamed J, Handratta VD, et al. 2006. Regulation of androgen receptor activity by tyrosine phosphorylation. Cancer Cell 10: 309–319. [DOI] [PubMed] [Google Scholar]
  63. Guo Z, Yang X, Sun F, Jiang R, Linn DE, Chen H, Chen H, Kong X, Melamed J, Tepper CG, et al. 2009. A novel androgen receptor splice variant is up-regulated during prostate cancer progression and promotes androgen depletion-resistant growth. Cancer Res 69: 2305–2313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. 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 Res 63: 149–153. [PubMed] [Google Scholar]
  65. He B, Kemppainen JA, Wilson EM. 2000. FXXLF and WXXLF sequences mediate the NH2-terminal interaction with the ligand binding domain of the androgen receptor. J Biol Chem 275: 22986–22994. [DOI] [PubMed] [Google Scholar]
  66. He B, Minges JT, Lee LW, Wilson EM. 2002. The FXXLF motif mediates androgen receptor-specific interactions with coregulators. J Biol Chem 277: 10226–10235. [DOI] [PubMed] [Google Scholar]
  67. Hearn JW, AbuAli G, Reichard CA, Reddy CA, Magi-Galluzzi C, Chang K-H, Carlson R, Rangel L, Reagan K, Davis BJ, et al. 2016. HSD3B1 and resistance to androgen deprivation therapy in prostate cancer: A multi-cohort study. Lancet Oncol 17: 1435–1444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Heemers HV. 2014. Targeting androgen receptor action for prostate cancer treatment: Does the post-receptor level provide novel opportunities? Int J Biol Sci 10: 576–587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Heemers HV, Tindall DJ. 2007. Androgen receptor (AR) coregulators: A diversity of functions converging on and regulating the AR transcriptional complex. Endocr Rev 28: 778–808. [DOI] [PubMed] [Google Scholar]
  70. Heemers HV, Tindall DJ. 2009. Unraveling the complexities of androgen receptor signaling in prostate cancer cells. Cancer Cell 15: 245–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Heemers HV, Regan KM, Schmidt LJ, Anderson SK, Ballman KV, Tindall DJ. 2009. Androgen modulation of coregulator expression in prostate cancer cells. Mol Endocrinol 23: 572–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Heemers HV, Schmidt LJ, Kidd E, Raclaw KA, Regan KM, Tindall DJ. 2010. Differential regulation of steroid nuclear receptor coregulator expression between normal and neoplastic prostate epithelial cells. Prostate 70: 959–970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Heery DM, Kalkhoven E, Hoare S, Parker MG. 1997. A signature motif in transcriptional co-activators mediates binding to nuclear receptors. Nature 387: 733–736. [DOI] [PubMed] [Google Scholar]
  74. Hofland J, van Weerden WM, Dits NFJ, Steenbergen J, van Leenders GJLH, Jenster G, Schröder FH, de Jong FH. 2010. Evidence of limited contributions for intratumoral steroidogenesis in prostate cancer. Cancer Res 70: 1256–1264. [DOI] [PubMed] [Google Scholar]
  75. Holzbeierlein J, Lal P, LaTulippe E, Smith A, Satagopan J, Zhang L, Ryan C, Smith S, Scher H, Scardino P, et al. 2004. Gene expression analysis of human prostate carcinoma during hormonal therapy identifies androgen-responsive genes and mechanisms of therapy resistance. Am J Pathol 164: 217–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Hu R, Dunn TA, Wei S, Isharwal S, Veltri RW, Humphreys E, Han M, Partin AW, Vessella RL, Isaacs WB, et al. 2009. Ligand-independent androgen receptor variants derived from splicing of cryptic exons signify hormone-refractory prostate cancer. Cancer Res 69: 16–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Huggins C, Hodges CV. 1941. Studies on prostatic cancer. I: The effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. Cancer Res 1: 293–297. [DOI] [PubMed] [Google Scholar]
  78. Ianculescu I, Wu DY, Siegmund KD, Stallcup MR. 2012. Selective roles for cAMP response element-binding protein binding protein and p300 protein as coregulators for androgen-regulated gene expression in advanced prostate cancer cells. J Biol Chem 287: 4000–4013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Imperato-McGinley J, Guerrero L, Gautier T, Peterson RE. 1974. Steroid 5α-reductase deficiency in man: An inherited form of male pseudohermaphroditism. Science 186: 1213–1215. [DOI] [PubMed] [Google Scholar]
  80. Jenster G, van der Korput HA, van Vroonhoven C, van der Kwast TH, Trapman J, Brinkmann AO. 1991. Domains of the human androgen receptor involved in steroid binding, transcriptional activation, and subcellular localization. Mol Endocrinol 5: 1396–1404. [DOI] [PubMed] [Google Scholar]
  81. Kaku N, Matsuda K, Tsujimura A, Kawata M. 2008. Characterization of nuclear import of the domain-specific androgen receptor in association with the importin α/β and Ran-Guanosine 5′-triphosphate systems. Endocrinology 149: 3960–3969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. King JC, Xu J, Wongvipat J, Hieronymus H, Carver BS, Leung DH, Taylor BS, Sander C, Cardiff RD, Couto SS, et al. 2009. Cooperativity of TMPRSS2-ERG with PI3-kinase pathway activation in prostate oncogenesis. Nat Genet 41: 524–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Knudsen KE. 2014. Hormone whodunit: Clues for solving the case of intratumor androgen production. Clin Cancer Res 20: 5343–5345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Knudsen KE, Scher HI. 2009. Starving the addiction: New opportunities for durable suppression of AR signaling in prostate cancer. Clin Cancer Res 15: 4792–4798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Labrie F, Dupont A, Simard J, Luu-The V, Bélanger A. 1993. Intracrinology: The basis for the rational design of endocrine therapy at all stages of prostate cancer. Eur Urol 24: 94–105. [DOI] [PubMed] [Google Scholar]
  86. Lamont KR, Tindall DJ. 2011. Minireview: Alternative activation pathways for the androgen receptor in prostate cancer. Mol Endocrinol 25: 897–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Li Y, Alsagabi M, Fan D, Bova GS, Tewfik AH, Dehm SM. 2011. Intragenic rearrangement and altered RNA splicing of the androgen receptor in a cell-based model of prostate cancer progression. Cancer Res 71: 2108–2117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Li Y, Hwang TH, Oseth LA, Hauge A, Vessella RL, Schmechel SC, Hirsch B, Beckman KB, Silverstein KA, Dehm SM. 2012. AR intragenic deletions linked to androgen receptor splice variant expression and activity in models of prostate cancer progression. Oncogene 31: 4759–4767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Li Y, Chan SC, Brand LJ, Hwang TH, Silverstein KAT, Dehm SM. 2013. Androgen receptor splice variants mediate enzalutamide resistance in castration-resistant prostate cancer cell lines. Cancer Res 73: 483–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Li Z, Bishop AC, Alyamani M, Garcia JA, Dreicer R, Bunch D, Liu J, Upadhyay SK, Auchus RJ, Sharifi N. 2015. Conversion of abiraterone to D4A drives anti-tumour activity in prostate cancer. Nature 523: 347–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Li Z, Alyamani M, Li J, Rogacki K, Abazeed M, Upadhyay SK, Balk SP, Taplin ME, Auchus RJ, Sharifi N. 2016. Redirecting abiraterone metabolism to fine-tune prostate cancer anti-androgen therapy. Nature 533: 547–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Li J, Alyamani M, Zhang A, Chang K-H, Berk M, Li Z, Zhu Z, Petro M, Magi-Galluzzi C, Taplin M-E, et al. 2017. Aberrant corticosteroid metabolism in tumor cells enables GR takeover in enzalutamide resistant prostate cancer. eLife 10.7554/eLife.20183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Liao RS, Ma S, Miao L, Li R, Yin Y, Raj GV. 2013. Androgen receptor-mediated non-genomic regulation of prostate cancer cell proliferation. Transl Androl Urol 2: 187–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Lin C, Yang L, Tanasa B, Hutt K, Ju B, Ohgi K, Zhang J, Rose DW, Fu XD, Glass CK, et al. 2009. Nuclear receptor-induced chromosomal proximity and DNA breaks underlie specific translocations in cancer. Cell 139: 1069–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Liu LL, Xie N, Sun S, Plymate S, Mostaghel E, Dong X. 2014. Mechanisms of the androgen receptor splicing in prostate cancer cells. Oncogene 33: 3140–3150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Lösel R, Wehling M. 2003. Nongenomic actions of steroid hormones. Nat Rev Mol Cell Biol 4: 46–56. [DOI] [PubMed] [Google Scholar]
  97. Lu J, Van der Steen T, Tindall DJ. 2015. Are androgen receptor variants a substitute for the full-length receptor? Nat Rev Urol 12: 137–144. [DOI] [PubMed] [Google Scholar]
  98. Lubahn DB, Brown TR, Simental JA, Higgs HN, Migeon CJ, Wilson EM, French FS. 1989. Sequence of the intron/exon junctions of the coding region of the human androgen receptor gene and identification of a point mutation in a family with complete androgen insensitivity. Proc Natl Acad Sci 86: 9534–9538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Mani R-S, Tomlins SA, Callahan K, Ghosh A, Nyati MK, Varambally S, Palanisamy N, Chinnaiyan AM. 2009. Induced chromosomal proximity and gene fusions in prostate cancer. Science 326: 1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Marks LS. 2004. 5α-reductase: History and clinical importance. Rev Urol 6: S11–S21. [PMC free article] [PubMed] [Google Scholar]
  101. Marshall TW, Link KA, Petre-Draviam CE, Knudsen KE. 2003. Differential requirement of SWI/SNF for androgen receptor activity. J Biol Chem 278: 30605–30613. [DOI] [PubMed] [Google Scholar]
  102. Maughan BL, Antonarakis ES. 2015. Clinical relevance of androgen receptor splice variants in castration-resistant prostate cancer. Curr Treat Options Oncol 16: 57. [DOI] [PubMed] [Google Scholar]
  103. McMullin RP, Dobi A, Mutton LN, Orosz A, Maheshwari S, Shashikant CS, Bieberich CJ. 2010. A FOXA1-binding enhancer regulates Hoxb13 expression in the prostate gland. Proc Natl Acad Sci 107: 98–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Mellinghoff IK, Vivanco I, Kwon A, Tran C, Wongvipat J, Sawyers CL. 2004. HER2/neu kinase-dependent modulation of androgen receptor function through effects on DNA binding and stability. Cancer Cell 6: 517–527. [DOI] [PubMed] [Google Scholar]
  105. Mills IG. 2014. Maintaining and reprogramming genomic androgen receptor activity in prostate cancer. Nat Rev Cancer 14: 187–198. [DOI] [PubMed] [Google Scholar]
  106. Mohler JL. 2008. Castration-recurrent prostate cancer is not androgen-independent. Adv Exp Med Biol 617: 223–234. [DOI] [PubMed] [Google Scholar]
  107. Mohler JL, Gregory CW, Ford OH, Kim D, Weaver CM, Petrusz P, Wilson EM, French FS. 2004. The androgen axis in recurrent prostate cancer. Clin Cancer Res 10: 440–448. [DOI] [PubMed] [Google Scholar]
  108. Mohler JL, Armstrong AJ, Bahnson RR, Boston B, Busby JE, D’Amico AV, Eastham JA, Enke CA, Farrington T, Higano CS, et al. 2012. Prostate cancer, Version 3.2012: Featured updates to the NCCN guidelines. J Natl Compr Canc Netw 10: 1081–107. [DOI] [PubMed] [Google Scholar]
  109. Montgomery RB, Mostaghel EA, Vessella R, Hess DL, Kalhorn TF, Higano CS, True LD, Nelson PS. 2008. Maintenance of intratumoral androgens in metastatic prostate cancer: A mechanism for castration-resistant tumor growth. Cancer Res 68: 4447–4454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Mostaghel EA. 2013. Steroid hormone synthetic pathways in prostate cancer. Transl Androl Urol 2: 212–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Mostaghel EA. 2014a. Abiraterone in the treatment of metastatic castration-resistant prostate cancer. Cancer Manag Res 6: 39–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Mostaghel EA. 2014b. Beyond T and DHT—Novel steroid derivatives capable of wild type androgen receptor activation. Int J Biol Sci 10: 602–613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Mostaghel EA, Page ST, Lin DW, Fazli L, Coleman IM, True LD, Knudsen B, Hess DL, Nelson CC, Matsumoto AM, et al. 2007. Intraprostatic androgens and androgen-regulated gene expression persist after testosterone suppression: Therapeutic implications for castration-resistant prostate cancer. Cancer Res 67: 5033–5041. [DOI] [PubMed] [Google Scholar]
  114. Mostaghel EA, Marck BT, Plymate SR, Vessella RL, Balk S, Matsumoto AM, Nelson PS, Montgomery RB. 2011. Resistance to CYP17A1 inhibition with abiraterone in castration-resistant prostate cancer: Induction of steroidogenesis and androgen receptor splice variants. Clin Cancer Res 17: 5913–5925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Mostaghel EA, Nelson PS, Lange P, Lin DW, Taplin ME, Balk S, Ellis W, Kantoff P, Marck B, Tamae D, et al. 2014. Targeted androgen pathway suppression in localized prostate cancer: A pilot study. J Clin Oncol 32: 229–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Nakamura Y, Hornsby PJ, Casson P, Morimoto R, Satoh F, Xing Y, Kennedy MR, Sasano H, Rainey WE. 2009. Type 5 17β-hydroxysteroid dehydrogenase (AKR1C3) contributes to testosterone production in the adrenal reticularis. J Clin Endocrinol Metab 94: 2192–2198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Nelson PS, Clegg N, Arnold H, Ferguson C, Bonham M, White J, Hood L, Lin B. 2002. The program of androgen-responsive genes in neoplastic prostate epithelium. Proc Natl Acad Sci 99: 11890–11895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Ni L, Llewellyn R, Kesler CT, Kelley JB, Spencer A, Snow CJ, Shank L, Paschal BM. 2013. Androgen induces a switch from cytoplasmic retention to nuclear import of the androgen receptor. Mol Cell Biol 33: 4766–4778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Nicolaides NC, Galata Z, Kino T, Chrousos GP, Charmandari E. 2010. The human glucocorticoid receptor: Molecular basis of biologic function. Steroids 75: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Nishiyama T. 2014. Serum testosterone levels after medical or surgical androgen deprivation: A comprehensive review of the literature. Urol Oncol 32: 38.e17–28. [DOI] [PubMed] [Google Scholar]
  121. Nishiyama T, Hashimoto Y, Takahashi K. 2004. The influence of androgen deprivation therapy on dihydrotestosterone levels in the prostatic tissue of patients with prostate cancer. Clin Cancer Res 10: 7121–7126. [DOI] [PubMed] [Google Scholar]
  122. Nuclear Receptors Nomenclature Committee. 1999. A unified nomenclature system for the nuclear receptor superfamily. Cell 97: 161–163. [DOI] [PubMed] [Google Scholar]
  123. Ozanne DM, Brady ME, Cook S, Gaughan L, Neal DE, Robson CN. 2000. Androgen receptor nuclear translocation is facilitated by the F-actin cross-linking protein filamin. Mol Endocrinol 14: 1618–1626. [DOI] [PubMed] [Google Scholar]
  124. Page ST, Lin DW, Mostaghel EA, Hess DL, True LD, Amory JK, Nelson PS, Matsumoto AM, Bremner WJ. 2006. Persistent intraprostatic androgen concentrations after medical castration in healthy men. J Clin Endocrinol Metab 91: 3850–3856. [DOI] [PubMed] [Google Scholar]
  125. Page ST, Lin DW, Mostaghel EA, Marck BT, Wright JL, Wu J, Amory JK, Nelson PS, Matsumoto AM. 2011. Dihydrotestosterone administration does not increase intraprostatic androgen concentrations or alter prostate androgen action in healthy men: A randomized-controlled trial. J Clin Endocrinol Metab 96: 430–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Park K, Dalton JT, Narayanan R, Barbieri CE, Hancock ML, Bostwick DG, Steiner MS, Rubin MA. 2014. TMPRSS2:ERG gene fusion predicts subsequent detection of prostate cancer in patients with high-grade prostatic intraepithelial neoplasia. J Clin Oncol 32: 206–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Pedram A, Razandi M, Sainson RCA, Kim JK, Hughes CC, Levin ER. 2007. A conserved mechanism for steroid receptor translocation to the plasma membrane. J Biol Chem 282: 22278–22288. [DOI] [PubMed] [Google Scholar]
  128. Perner S, Mosquera JM, Demichelis F, Hofer MD, Paris PL, Simko J, Collins C, Bismar TA, Chinnaiyan AM, De Marzo AM, et al. 2007. TMPRSS2-ERG fusion prostate cancer: An early molecular event associated with invasion. Am J Surg Pathol 31: 882–888. [DOI] [PubMed] [Google Scholar]
  129. Pomerantz MM, Li F, Takeda DY, Lenci R, Chonkar A, Chabot M, Cejas P, Vazquez F, Cook J, Shivdasani RA, et al. 2015a. The androgen receptor cistrome is extensively reprogrammed in human prostate tumorigenesis. Nat Genet 47: 1346–1351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Pomerantz MM, Li F, Takeda DY, Lenci R, Chonkar A, Chabot M, Cejas P, Vazquez F, Cook J, Shivdasani RA, et al. 2015b. The androgen receptor cistrome is extensively reprogrammed in human prostate tumorigenesis. Nat Genet 47: 1346–1351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Ponguta LA, Gregory CW, French FS, Wilson EM. 2008. Site-specific androgen receptor serine phosphorylation linked to epidermal growth factor-dependent growth of castration-recurrent prostate cancer. J Biol Chem 283: 20989–21001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Ravindranathan P, Lee TK, Yang L, Centenera MM, Butler L, Tilley WD, Hsieh JT, Ahn JM, Raj GV. 2013. Peptidomimetic targeting of critical androgen receptor-coregulator interactions in prostate cancer. Nat Commun 4: 1923. [DOI] [PubMed] [Google Scholar]
  133. Rittmaster RS. 1997. 5α-reductase inhibitors. J Androl 18: 582–587. [PubMed] [Google Scholar]
  134. Rittmaster R, Hahn RG, Ray P, Shannon JB, Wurzel R. 2008. Effect of dutasteride on intraprostatic androgen levels in men with benign prostatic hyperplasia or prostate cancer. Urology 72: 808–812. [DOI] [PubMed] [Google Scholar]
  135. 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: 1215–1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Rosner W, Hryb DJ, Khan MS, Nakhla AM, Romas NA. 1991. Sex hormone-binding globulin: Anatomy and physiology of a new regulatory system. J Steroid Biochem Mol Biol 40: 813–820. [DOI] [PubMed] [Google Scholar]
  137. Ruizeveld de Winter JA, Janssen PJ, Sleddens HM, Verleun-Mooijman MC, Trapman J, Brinkmann AO, Santerse AB, Schröder FH, van der Kwast TH. 1994. Androgen receptor status in localized and locally progressive hormone refractory human prostate cancer. Am J Pathol 144: 735–746. [PMC free article] [PubMed] [Google Scholar]
  138. Russell DW, Wilson JD. 1994. Steroid 5α-reductase: Two genes/two enzymes. Annu Rev Biochem 63: 25–61. [DOI] [PubMed] [Google Scholar]
  139. Ryan CJ, Tindall DJ. 2011. Androgen receptor rediscovered: The new biology and targeting the androgen receptor therapeutically. J Clin Oncol 29: 3651–3658. [DOI] [PubMed] [Google Scholar]
  140. Ryan CJ, Smith A, Lal P, Satagopan J, Reuter V, Scardino P, Gerald W, Scher HI. 2006. Persistent prostate-specific antigen expression after neoadjuvant androgen depletion: An early predictor of relapse or incomplete androgen suppression. Urology 68: 834–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Ryan CJ, Smith MR, Fizazi K, Saad F, Mulders PFA, Sternberg CN, Miller K, Logothetis CJ, Shore ND, Small EJ, et al. 2015. Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy-naïve men with metastatic castration-resistant prostate cancer (COU-AA-302): Final overall survival analysis of a randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol 16: 152–160. [DOI] [PubMed] [Google Scholar]
  142. Sadi MV, Walsh PC, Barrack ER. 1991. Immunohistochemical study of androgen receptors in metastatic prostate cancer. Comparison of receptor content and response to hormonal therapy. Cancer 67: 3057–3064. [DOI] [PubMed] [Google Scholar]
  143. Sahu B, Laakso M, Pihlajamaa P, Ovaska K, Sinielnikov I, Hautaniemi S, Jänne OA. 2013. FoxA1 specifies unique androgen and glucocorticoid receptor binding events in prostate cancer cells. Cancer Res 73: 1570–1580. [DOI] [PubMed] [Google Scholar]
  144. Scher HI, Kelly WK. 1993. Flutamide withdrawal syndrome: Its impact on clinical trials in hormone-refractory prostate cancer. J Clin Oncol 11: 1566–1572. [DOI] [PubMed] [Google Scholar]
  145. Scher HI, Sawyers CL. 2005. Biology of progressive, castration-resistant prostate cancer: Directed therapies targeting the androgen-receptor signaling axis. J Clin Oncol 23: 8253–8261. [DOI] [PubMed] [Google Scholar]
  146. Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, de Wit R, Mulders P, Chi KN, Shore ND, et al. 2012. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med 367: 1187–1197. [DOI] [PubMed] [Google Scholar]
  147. Schmidt LJ, Tindall DJ. 2011. Steroid 5α-reductase inhibitors targeting BPH and prostate cancer. J Steroid Biochem Mol Biol 125: 32–38. [DOI] [PubMed] [Google Scholar]
  148. Schröder F, Bangma C, Angulo JC, Alcaraz A, Colombel M, McNicholas T, Tammela TL, Nandy I, Castro R. 2013. Dutasteride treatment over 2 years delays prostate-specific antigen progression in patients with biochemical failure after radical therapy for prostate cancer: Results from the randomised, placebo-controlled Avodart After Radical Therapy for Prostate Cancer. Eur Urol 63: 779–787. [DOI] [PubMed] [Google Scholar]
  149. Shaffer PL, Jivan A, Dollins DE, Claessens F, Gewirth DT. 2004. Structural basis of androgen receptor binding to selective androgen response elements. Proc Natl Acad Sci 101: 4758–4763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Shah SK, Trump DL, Sartor O, Tan W, Wilding GE, Mohler JL. 2009. Phase II study of Dutasteride for recurrent prostate cancer during androgen deprivation therapy. J Urol 181: 621–626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Sharifi N. 2010. New agents and strategies for the hormonal treatment of castration-resistant prostate cancer. Expert Opin Investig Drugs 19: 837–846. [DOI] [PubMed] [Google Scholar]
  152. Sharifi N. 2013. Mechanisms of androgen receptor activation in castration-resistant prostate cancer. Endocrinology 154: 4010–4017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Sharifi N. 2014. Steroid receptors aplenty in prostate cancer. N Engl J Med 370: 970–971. [DOI] [PubMed] [Google Scholar]
  154. Sharifi N, Auchus RJ. 2012. Steroid biosynthesis and prostate cancer. Steroids 77: 719–726. [DOI] [PubMed] [Google Scholar]
  155. Sharifi N, Gulley JL, Dahut WL. 2005. Androgen deprivation therapy for prostate cancer. JAMA 294: 238–244. [DOI] [PubMed] [Google Scholar]
  156. Sharma NL, Massie CE, Ramos-Montoya A, Zecchini V, Scott HE, Lamb AD, MacArthur S, Stark R, Warren AY, Mills IG, et al. 2013. The androgen receptor induces a distinct transcriptional program in castration-resistant prostate cancer in man. Cancer Cell 23: 35–47. [DOI] [PubMed] [Google Scholar]
  157. Shiota M, Yokomizo A, Fujimoto N, Naito S. 2011. Androgen receptor cofactors in prostate cancer: Potential therapeutic targets of castration-resistant prostate cancer. Curr Cancer Drug Targets 11: 870–881. [DOI] [PubMed] [Google Scholar]
  158. Simard J, Ricketts ML, Gingras S, Soucy P, Feltus FA, Melner MH. 2005. Molecular biology of the 3β-hydroxysteroid dehydrogenase/Δ54 isomerase gene family. Endocr Rev 26: 525–582. [DOI] [PubMed] [Google Scholar]
  159. Simental JA, Sar M, Lane MV, French FS, Wilson EM. 1991. Transcriptional activation and nuclear targeting signals of the human androgen receptor. J Biol Chem 266: 510–518. [PubMed] [Google Scholar]
  160. Smith DF, Toft DO. 2008. Minireview: The intersection of steroid receptors with molecular chaperones: Observations and questions. Mol Endocrinol 22: 2229–2240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Stanbrough M, Bubley GJ, Ross K, Golub TR, Rubin MA, Penning TM, Febbo PG, Balk SP. 2006. Increased expression of genes converting adrenal androgens to testosterone in androgen-independent prostate cancer. Cancer Res 66: 2815–2825. [DOI] [PubMed] [Google Scholar]
  162. Steers WD. 2001. 5α-reductase activity in the prostate. Urology 58: 17–24; discussion 24. [DOI] [PubMed] [Google Scholar]
  163. Sun S, Sprenger CCT, Vessella RL, Haugk K, Soriano K, Mostaghel EA, Page ST, Coleman IM, Nguyen HM, Sun H, et al. 2010. Castration resistance in human prostate cancer is conferred by a frequently occurring androgen receptor splice variant. J Clin Invest 120: 2715–2730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Szmulewitz RZ, Chung E, Al-Ahmadie H, Daniel S, Kocherginsky M, Razmaria A, Zagaja GP, Brendler CB, Stadler WM, Conzen SD. 2012. Serum/glucocorticoid-regulated kinase 1 expression in primary human prostate cancers. Prostate 72: 157–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. 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. N Engl J Med 332: 1393–1398. [DOI] [PubMed] [Google Scholar]
  166. 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 Res 59: 2511–2515. [PubMed] [Google Scholar]
  167. Taplin M-E, 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. J Clin Oncol 21: 2673–2678. [DOI] [PubMed] [Google Scholar]
  168. Taplin M-E, Montgomery B, Logothetis CJ, Bubley GJ, Richie JP, Dalkin BL, Sanda MG, Davis JW, Loda M, True LD, et al. 2014. Intense androgen-deprivation therapy with abiraterone acetate plus leuprolide acetate in patients with localized high-risk prostate cancer: Results of a randomized phase II neoadjuvant study. J Clin Oncol 32: 3705–3715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Thadani-Mulero M, Nanus DM, Giannakakou P. 2012. Androgen receptor on the move: Boarding the microtubule expressway to the nucleus. Cancer Res 72: 4611–4615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. The Cancer Genome Atlas Research Network. 2015. The molecular taxonomy of primary prostate cancer. Cell 163: 1011–1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Thigpen AE, Cala KM, Russell DW. 1993. Characterization of Chinese hamster ovary cell lines expressing human steroid 5α-reductase isozymes. J Biol Chem 268: 17404–17412. [PubMed] [Google Scholar]
  172. Thirumalai A, Cooper LA, Rubinow KB, Amory JK, Lin DW, Wright JL, Marck BT, Matsumoto AM, Page ST. 2016. Stable intraprostatic dihydrotestosterone in healthy medically castrate men treated with exogenous testosterone. J Clin Endocrinol Metab 101: 2937–2944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Thomas LN, Douglas RC, Lazier CB, Too CKL, Rittmaster RS, Tindall DJ. 2008. Type 1 and type 2 5α-reductase expression in the development and progression of prostate cancer. Eur Urol 53: 244–252. [DOI] [PubMed] [Google Scholar]
  174. Tilley WD, Marcelli M, Wilson JD, McPhaul MJ. 1989. Characterization and expression of a cDNA encoding the human androgen receptor. Proc Natl Acad Sci 86: 327–331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Titus MA, Schell MJ, Lih FB, Tomer KB, Mohler JL. 2005. Testosterone and dihydrotestosterone tissue levels in recurrent prostate cancer. Clin Cancer Res 11: 4653–4657. [DOI] [PubMed] [Google Scholar]
  176. Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun X-W, Varambally S, Cao X, Tchinda J, Kuefer R, et al. 2005. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310: 644–648. [DOI] [PubMed] [Google Scholar]
  177. Tomlins SA, Bjartell A, Chinnaiyan AM, Jenster G, Nam RK, Rubin MA, Schalken JA. 2009. ETS gene fusions in prostate cancer: From discovery to daily clinical practice. Eur Urol 56: 275–286. [DOI] [PubMed] [Google Scholar]
  178. 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: 787–790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Umesono K, Evans RM. 1989. Determinants of target gene specificity for steroid/thyroid hormone receptors. Cell 57: 1139–1146. [DOI] [PubMed] [Google Scholar]
  180. Veldscholte J, Berrevoets CA, Ris-Stalpers C, Kuiper GG, Jenster G, Trapman J, Brinkmann AO, Mulder E. 1992. The androgen receptor in LNCaP cells contains a mutation in the ligand binding domain which affects steroid binding characteristics and response to antiandrogens. J Steroid Biochem Mol Biol 41: 665–669. [DOI] [PubMed] [Google Scholar]
  181. Wallén MJ, Linja M, Kaartinen K, Schleutker J, Visakorpi T. 1999. Androgen receptor gene mutations in hormone-refractory prostate cancer. J Pathol 189: 559–563. [DOI] [PubMed] [Google Scholar]
  182. Wang Q, Li W, Zhang Y, Yuan X, Xu K, Yu J, Chen Z, Beroukhim R, Wang H, Lupien M, et al. 2009. Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell 138: 245–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Wang D, Garcia-Bassets I, Benner C, Li W, Su X, Zhou Y, Qiu J, Liu W, Kaikkonen MU, Ohgi KA, et al. 2011a. Reprogramming transcription by distinct classes of enhancers functionally defined by eRNA. Nature 474: 390–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Wang Y, Lonard DM, Yu Y, Chow DC, Palzkill TG, O’Malley BW. 2011b. Small molecule inhibition of the steroid receptor coactivators, SRC-3 and SRC-1. Mol Endocrinol 25: 2041–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Wang Y, Lonard DM, Yu Y, Chow DC, Palzkill TG, Wang J, Qi R, Matzuk AJ, Song X, Madoux F, et al. 2014. Bufalin is a potent small-molecule inhibitor of the steroid receptor coactivators SRC-3 and SRC-1. Cancer Res 74: 1506–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Ware KE, Garcia-Blanco MA, Armstrong AJ, Dehm SM. 2014. Biologic and clinical significance of androgen receptor variants in castration resistant prostate cancer. Endocr Relat Cancer 21: T87–T103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Watson PA, Chen YF, Balbas MD, Wongvipat J, Socci ND, Viale A, Kim K, Sawyers CL. 2010. Constitutively active androgen receptor splice variants expressed in castration-resistant prostate cancer require full-length androgen receptor. Proc Natl Acad Sci 107: 16759–16765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Wilson JD. 2001. The role of 5α-reduction in steroid hormone physiology. Reprod Fertil Dev 13: 673–678. [DOI] [PubMed] [Google Scholar]
  189. Wilson EM, French FS. 1976. Binding properties of androgen receptors. Evidence for identical receptors in rat testis, epididymis, and prostate. J Biol Chem 251: 5620–5629. [PubMed] [Google Scholar]
  190. Xu Y, Dalrymple SL, Becker RE, Denmeade SR, Isaacs JT. 2006. Pharmacologic basis for the enhanced efficacy of dutasteride against prostatic cancers. Clin Cancer Res 12: 4072–4079. [DOI] [PubMed] [Google Scholar]
  191. Xu D, Zhan Y, Qi Y, Cao B, Bai S, Xu W, Gambhir SS, Lee P, Sartor O, Flemington EK, et al. 2015. Androgen receptor splice variants dimerize to transactivate target genes. Cancer Res 75: 3663–3671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Zhang A, Zhang J, Plymate S, Mostaghel EA. 2016. Classical and non-classical roles for pre-receptor control of DHT metabolism in prostate cancer progression. Horm Cancer 7: 104–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Zhao Y, Tindall DJ, Huang H. 2014. Modulation of androgen receptor by FOXA1 and FOXO1 factors in prostate cancer. Int J Biol Sci 10: 614–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Zhou ZX, Lane MV, Kemppainen JA, French FS, Wilson EM. 1995. Specificity of ligand-dependent androgen receptor stabilization: Receptor domain interactions influence ligand dissociation and receptor stability. Mol Endocrinol 9: 208–218. [DOI] [PubMed] [Google Scholar]
  195. Zhou HJ, Yan J, Luo W, Ayala G, Lin SH, Erdem H, Ittmann M, Tsai SY, Tsai MJ. 2005. SRC-3 is required for prostate cancer cell proliferation and survival. Cancer Res 65: 7976–7983. [DOI] [PubMed] [Google Scholar]
  196. Zhu YS, Imperato-McGinley JL. 2009. 5α-reductase isozymes and androgen actions in the prostate. Ann NY Acad Sci 1155: 43–56. [DOI] [PubMed] [Google Scholar]
  197. Ziada A, Barqawi A, Glode LM, Varella-Garcia M, Crighton F, Majeski S, Rosenblum M, Kane M, Chen L, Crawford ED. 2004. The use of trastuzumab in the treatment of hormone refractory prostate cancer; phase II trial. Prostate 60: 332–337. [DOI] [PubMed] [Google Scholar]

Articles from Cold Spring Harbor Perspectives in Medicine are provided here courtesy of Cold Spring Harbor Laboratory Press

RESOURCES