1,016
Views
14
CrossRef citations to date
0
Altmetric
Editorial

Abiraterone acetate, a first-in-class CYP17 inhibitor, establishes a new treatment paradigm in castration-resistant prostate cancer

&
Pages 1-3 | Published online: 10 Jan 2014

Androgen-deprivation therapy (ADT) has been the standard treatment for advanced and metastatic prostate adenocarcinoma for the past seven decades Citation[1]. However, clinical resistance to ADT eventually emerges and leads to a poor clinical outcome Citation[2]. While successful second-line hormone manipulations with agents such as first-generation antiandrogens (flutamide, bicalutamide and nilutamide) and the antifungal ketoconazole (that also has activity as an androgen synthesis inhibitor) are still possible, these responses are usually short lived and associated with significant toxicities Citation[3]. Chemotherapy is frequently used in this setting (with less than ideal responses) Citation[4], under the inaccurate assumption that the disease is, at this point, ‘hormone-refractory’ or ‘androgen-independent’, terms mistakenly used frequently throughout the literature. However, the re-expression of PSA Citation[5], an androgen receptor (AR)-regulated gene, in prostate cancers that have progressed after ADT, and its subsequent decline after treatment with second-line hormonal therapies has demonstrated that the AR pathway is still active at the molecular level. These data demonstrate that the tumor is not necessarily ‘androgen-independent’ Citation[6]. Instead, the term ‘castration-resistant prostate cancer’ (CRPC) is more appropriate Citation[1].

Several mechanisms have been proposed to explain the persistence of AR signaling in CRPC. Evidence has accumulated in recent years for each of these mechanisms, suggesting that these are not mutually exclusive and that substantial heterogeneity is present Citation[2]. It is becoming clear, for example, that ADT causes growth arrest, but does not induce significant apoptosis in prostate cancer cells Citation[7]. This provides them time to adapt to low androgen levels and, eventually, restart proliferating despite suppressed circulating peripheral testosterone levels Citation[8]. More recent in vitro evidence had shown that a subset of prostate cancer cells that survive androgen-depleted conditions express increased levels of stem cell markers and are able to repopulate the tumor, possibly through adaptive AR pathway changes Citation[9]. These adaptive changes include AR overexpression Citation[6,10] or gain-of-function mutations Citation[2,4] that increase receptor sensitivity to circulating androgens. Alternatively, these mutations can also allow AR to be activated by noncanonical ligands Citation[2], such as other steroids (e.g., progesterone) Citation[1], or even by agents initially used as antiandrogens in the same patient (e.g., flutamide and bicalutamide). The latter phenomenon, also known as the ‘antagonist-to-agonist’ conversion of these first generation antiandrogens, explains the ‘antiandrogen withdrawal’ effect (i.e., the decline in PSA manifested in ∼25% of patients following the discontinuation of these drugs after clinical progression) Citation[1,10]. Furthermore, the sensitivity of AR to its ligand(s) can also be modulated or enhanced by changes in the activity of coactivators and/or corepressors that play important roles in AR transcriptional activity Citation[1]. In fact, changes in the coactivator-to-corepressor ratio have been proposed as another possible explanation for the ‘antagonist-to-agonist’ conversion of antiandrogens Citation[4].

Another crucial mechanism of prostate cancer progression after ADT is based on the fact that, while gonadotropin-releasing hormone analogs suppress gonadal synthesis of testosterone, they do not affect adrenal production of androgen precursors (dehydroepiandrosterone and androstenedione) that can then become available to the prostate cancer tissue and serve as precursors to testosterone Citation[11]. The importance of these androgen precursors is very high, as the circulating concentrations of dehydroepiandrosterone are much higher than those of testosterone in healthy males. During ADT, serum testosterone and dihydrotestosterone (DHT) levels decrease by >90%, while intraprostatic testosterone and DHT levels decrease by only 60–70% and have a closer correlation with serum dehydroepiandrosterone levels. Moreover, as reported by Montgomery and colleagues, the intraprostatic concentrations of testosterone were up to fourfold higher in metastatic tissue from CRPC patients (despite castrate levels of testosterone in the serum), compared to primary prostate cancer tissue from untreated eugonadal patients Citation[12]. Taken together, these findings suggest that ADT activates an adaptive response in the prostate cancer tissue, which augments its ability to convert adrenal precursors into testosterone and DHT Citation[13], probably via upregulation of steroidogenic enzymes (e.g., AKR1C3) Citation[14], to a concentration sufficient to promote AR-dependent gene expression Citation[2,6]. Somewhat more controversial is the concept that prostate cancer tissue is able to synthesize testosterone de novo directly from cholesterol Citation[11]. Supporting this theory, some investigators have reported upregulation of the key steroidogenic enzyme CYP17 in prostate cells after ADT Citation[12]. Others, however, have suggested that CYP17 levels in prostate cancer tissue are inadequate for local de novo synthesis of testosterone directly from cholesterol, and implicated adrenal CYP17 as the major source of androgen precursors after ADT Citation[15].

In any case, adrenal androgen precursors are a crucial source of AR ligands after ADT, and attempts to suppress it have been ongoing for several years. Initially, the antifungal agent ketoconazole was used as it has inhibitory activity on several enzymes involved in androgen synthesis, such as CYP17 and CYP11, and can provide brief clinical responses. However, ketoconazole is a weak androgen synthesis inhibitor and causes substantial toxicity Citation[3]. Abiraterone acetate – a newer, more potent and better tolerated CYP17 inhibitor – accomplished a significant overall survival benefit of 3.9 months in a Phase III trial in chemotherapy-refractory CRPC patients Citation[16], leading to its recent approval by the US FDA for the treatment of prostate cancer in this clinical setting. Most importantly, these results provide proof-of-principle that the AR signaling pathway is still quite active in patients with castrate levels of peripheral testosterone and represent a therapeutic opportunity for the development of future combination therapies.

Specifically, the use of abiraterone acetate in combination with gonadotropin-releasing hormone analogs as first-line therapy for prostate cancer could potentially cause more potent upfront androgen depletion within the prostate cancer microenvironment, possibly achieving more substantial apoptosis of prostate cancer cells and denying them the opportunity to undergo adaptive changes, such as the ones mentioned previously, that would lead to the emergence of CRPC. This maximal upfront suppression of intratumoral androgen levels could significantly increase the time to clinical progression and even improve overall survival for patients with locally advanced or even metastatic disease.

Other future directions include the development of additional CYP17 inhibitors. TAK700 (orteronel), currently in Phase III testing in CRPC, was designed to preferentially inhibit the 17,20 lyase activity of CYP17 (which is necessary for androgen synthesis only) as opposed to 17-hydroxylase activity (that is required for both androgen and glucocorticoid synthesis) Citation[17]. As abiraterone blocks both the 17-hydroxylase and 17,20 lyase activities of CYP17, it requires concomitant glucocorticoid supplementation Citation[3,16]. Therefore, TAK700 was designed with the hope of obviating the need for glucocorticoid supplementation, although it remains unclear whether this specificity will be maintained with the higher doses that are required in clinical practice. Another novel agent in clinical development is galeterone (TOK-001 or VN/124-1), which is both a CYP17 inhibitor and an AR antagonist Citation[1]. The results from these clinical trials are eagerly awaited.

While the available results and future expectations for abiraterone and other anti-AR targeted therapies are encouraging, all patients that receive abiraterone under its current indication are expected to eventually progress. This highlights the multiple mechanisms for resistance observed in CRPC. The increased expression of AR mRNA variants, which lack the ligand-binding domain and are constitutively active, provides a likely explanation for the progression seen after an initial response to abiraterone Citation[18,19]. The expression of these variants increases acutely in response to androgen ablation, such as that induced by abiraterone, while it is suppressed by testosterone Citation[19]. It has been reported that the transcriptional activity of these AR variants can be blocked by MDV3100, a high-affinity AR antagonist Citation[20]. This compound was rationally designed using data from a cocrystal structure of bicalutamide bound with mutant AR to create a chemical library that was screened for AR antagonistic activity Citation[10]. In preclinical experiments Citation[10], MDV3100 does not appear (so far) to have an agonistic effect on AR, contrary to the first generation antiandrogens, reportedly because of the impairment of AR translocation and DNA binding produced by this newer drug Citation[1]. Initial clinical data shows that MDV3100 has relatively few side effects Citation[5], which, along with their noncompeting mechanisms of action, could argue in favor of its use in combination with abiraterone. Recent Phase III data demonstrate a 4.8-month overall survival benefit for chemotherapy-refractory patients treated with MDV3100 compared with placebo. However, the best use for MDV3100 is still undetermined. Similarly, its reported ability to block AR variants is yet to be tested in a rigorous clinical setting.

After many years in which new developments were stalled, excellent translational research has finally brought us to an exciting era in the management of prostate cancer by means of rationally designed new drugs. This research has not only provided medications to be added to the armamentarium for CRPC treatment, but most importantly, gave us a deeper understanding of the pathways involved in this disease and the mechanisms of resistance to hormonal therapies. With many of these mechanisms at play, sometimes simultaneously (explaining the variable and short-lived responses seen among different patients), the next logical step seems to be the use of combination regimens with these and other drugs which could potentially increase treatment efficacy. Although doing this while avoiding dose-limiting toxicity has been challenging in the past, the higher activity and the nonoverlapping side-effect profiles of these new-generation drugs increase the likelihood of success.

Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

References

  • Chen Y, Clegg NJ, Scher HI. Anti-androgens and androgen-depleting therapies in prostate cancer: new agents for an established target. Lancet Oncol.10(10), 981–991 (2009).
  • Holzbeierlein J, Lal P, LaTulippe E et al. Gene expression analysis of human prostate carcinoma during hormonal therapy identifies androgen-responsive genes and mechanisms of therapy resistance. Am. J. Pathol.164(1), 217–227 (2004).
  • Attard G, Reid AH, A’Hern R et al. Selective inhibition of CYP17 with abiraterone acetate is highly active in the treatment of castration-resistant prostate cancer. J. Clin. Oncol.27(23), 3742–3748 (2009).
  • Scher HI, Sawyers CL. Biology of progressive, castration-resistant prostate cancer: directed therapies targeting the androgen-receptor signaling axis. J. Clin. Oncol.23(32), 8253–8261 (2005).
  • Scher HI, Beer TM, Higano CS et al. Antitumour activity of MDV3100 in castration-resistant prostate cancer: a Phase 1–2 study. Lancet375(9724), 1437–1446 (2010).
  • Mostaghel EA, Page ST, Lin DW et al. Intraprostatic androgens and androgen-regulated gene expression persist after testosterone suppression: therapeutic implications for castration-resistant prostate cancer. Cancer Res.67(10), 5033–5041 (2007).
  • Ohlson N, Wikstrom P, Stattin P, Bergh A. Cell proliferation and apoptosis in prostate tumors and adjacent non-malignant prostate tissue in patients at different time-points after castration treatment. Prostate62(4), 307–315 (2005).
  • Gregory CW, Johnson RT Jr, Mohler JL, French FS, Wilson EM. Androgen receptor stabilization in recurrent prostate cancer is associated with hypersensitivity to low androgen. Cancer Res.61(7), 2892–2898 (2001).
  • Pfeiffer MJ, Smit FP, Sedelaar JP, Schalken JA. Steroidogenic enzymes and stem cell markers are upregulated during androgen deprivation in prostate cancer. Mol. Med.17(7–8), 657–664 (2011).
  • Tran C, Ouk S, Clegg NJ et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science324(5928), 787–790 (2009).
  • Locke JA, Guns ES, Lubik AA et al. Androgen levels increase by intratumoral de novo steroidogenesis during progression of castration-resistant prostate cancer. Cancer Res.68(15), 6407–6415 (2008).
  • Montgomery RB, Mostaghel EA, Vessella R et al. Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth. Cancer Res.68(11), 4447–4454 (2008).
  • Pelletier G. Expression of steroidogenic enzymes and sex-steroid receptors in human prostate. Best Pract. Res. Clin. Endocrinol. Metab.22(2), 223–228 (2008).
  • Stanbrough M, Bubley GJ, Ross K et al. Increased expression of genes converting adrenal androgens to testosterone in androgen-independent prostate cancer. Cancer Res.66(5), 2815–2825 (2006).
  • Hofland J, van Weerden WM, Dits NF et al. Evidence of limited contributions for intratumoral steroidogenesis in prostate cancer. Cancer Res.70(3), 1256–1264 (2010).
  • de Bono JS, Logothetis CJ, Molina A et al. Abiraterone and increased survival in metastatic prostate cancer. N. Engl. J. Med.364(21), 1995–2005 (2011).
  • Dreicer R, Agus DB, MacVicar GR et al. Safety, pharmacokinetics, and efficacy of TAK-700 in metastatic castration-resistant prostrate cancer: a Phase I/II, open-label study. J. Clin. Oncol. (Meeting Abstracts)28, 3084 (2010).
  • Dehm SM, Schmidt LJ, Heemers HV, Vessella RL, Tindall DJ. Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Res.68(13), 5469–5477 (2008).
  • Mostaghel EA, Marck BT, Plymate SR et al. Resistance to CYP17A1 inhibition with abiraterone in castration-resistant prostate cancer: induction of steroidogenesis and androgen receptor splice variants. Clin. Cancer Res.17(18), 5913–5925 (2011).
  • Watson PA, Chen YF, Balbas MD et al. Constitutively active androgen receptor splice variants expressed in castration-resistant prostate cancer require full-length androgen receptor. Proc. Natl Acad. Sci. USA107(39), 16759–16765 (2010).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.