Skip to main content
Top

26-04-2016 | Advanced prostate cancer | Book chapter | Article

13. Sequencing Therapies in Metastatic Castration—Resistant Prostate Cancer

Authors: Michael T. Schweizer, Bruce Montgomery

Publisher: Springer International Publishing

Abstract

Since 2004, six agents have been approved for men with castration-resistant prostate cancer (CRPC) on the basis of Phase III data indicating a survival advantage with each of these drugs. Two of these agents, abiraterone and enzalutamide, inhibit androgen receptor (AR) signaling as their primary mechanism of action. In addition, taxanes, the only class of chemotherapeutic shown to afford a survival advantage for men with CRPC, may also function to impair AR-signaling through inhibiting microtubule mediated AR nuclear trafficking. Given these overlapping mechanisms of action it is not surprising that clinical evidence of cross-resistance between these AR-directed agents has begun to emerge, and questions regarding the optimal sequence for using each of these drugs persist. It also remains unclear at what point agents with mechanisms of action not directly related to AR function (i.e. sipuleucel-t and radium-233) should be utilized. In this chapter, we will discuss the key issues surrounding the decision-making that goes into choosing one CRPC drug over another; review the biologic basis for cross-resistance between agents; and highlight some of the data in support of clinical cross-resistance between these drugs.

Introduction

The approval of docetaxel in 2004 ended a nearly 60 year drought in which nothing was shown to prolong life for patients with metastatic castration-resistant prostate cancer (mCRPC), and ushered in a new era of treatment for mCRPC [1, 2]. Since then, six therapeutic agents (including docetaxel) have been approved for the treatment of metastatic castration-resistant prostate cancer (mCRPC)—all on the basis of Phase III data indicating a survival advantage with these drugs. These agents include chemotherapeutics (e.g. docetaxel and cabazitaxel), androgen receptor (AR) directed agents (e.g. abiraterone and enzalutamide), immunotherapeutics (i.e. sipuleucel-t) and radiopharmaceuticals (i.e. radium-223) (Fig. 13.1) [19].
Since docetaxel’s approval in 2004, many subsequent approvals in the mCRPC therapeutic space have been predicated on prior docetaxel exposure (Fig. 13.2). For instance, the Phase III cabazitaxel study mandated that patients be post-docetaxel, and as such it remains only approved in docetaxel-treated patients [5]. Similarly, the approvals for abiraterone and enzalutamide were initially granted post-docetaxel, with approval for docetaxel-naïve patients only occurring after Phase III trials in that patient population were completed [3, 4, 8, 9]. While this regulatory framework was likely born out of the desire to select a patient population in which a survival benefit could be quickly demonstrated, it is not reflective of the current treatment landscape in which many options exist for men with mCRPC. Furthermore, the Phase III studies testing these newer agents did not postulate biologic rationales for the specific therapeutic sequence being tested (i.e. pre- or post-docetaxel) or incorporate biomarkers to address the issue of optimal drug sequence, and there remains no historical or biologic basis for why AR targeting agents, such as abiraterone or enzalutamide, or cabazitaxel would only work if given after docetaxel. An acknowledgment of this point is reflected in the Prostate Cancer Working Group 3 (PCWG3) guidelines regarding the conduct of clinical trials in men with mCRPC. The PCWG3 guidelines provide a revised therapeutic framework, emphasizing the sequential use of approved agents, rather than selecting drugs based on a whether or not an individual has received prior docetaxel [10].
While some of these drugs have seemingly distinct mechanisms of action (e.g. radium-223 and sipuleucel-t), there is considerable mechanistic overlap between others (e.g. abiraterone and enzalutamide). As such, it is not surprising that evidence of cross-resistance between many of these therapies is being increasingly recognized. At the heart of this cross-resistance is the fact that many of our approved therapies rely on inhibiting the same target: the lineage-survival oncogene AR [12, 13]. In addition, increasing evidence indicates that prostate cancer evolves over the course of treatment, with more resistant subclones emerging, resulting in an inherently more difficult to treat disease [1417].
Questions surrounding the optimal mCRPC treatment paradigm and how to sequence the six available mCRPC drugs remain. In this chapter, we will outline our current understanding of how to best utilize the drugs approved for men with mCRPC, and highlight some of the controversies surrounding when to use each of these agents.

Docetaxel

Taxanes (i.e. docetaxel and cabazitaxel) remain the only class of chemotherapeutics that result in improved overall survival compared to active controls for men with mCRPC [1, 2]. In recent years, several effective oral AR-directed therapies have been approved in the pre-docetaxel space, and as such practice patterns are beginning to shift towards delayed use of docetaxel in favor of these less toxic, and more easily administered agents [3, 8, 18].
Docetaxel still remains an important therapeutic option, however, and under certain circumstances may be preferred over the use of drugs like abiraterone or enzalutamide. For instance, docetaxel may be more appropriate if a patient requires rapid palliation or control of visceral metastases [19]. Docetaxel has been reported to achieve a pain response after 27 days and result in improved quality of life after 43 days [20]. To put this in context, the time to pain palliation with abiraterone has been reported to be 5.6 months [21]. While cross-trial comparisons are bias-prone, this difference is quite dramatic. It should also be noted that in both the abiraterone and enzalutamide pre-docetaxel Phase III studies, only patients who were asymptomatic or minimally symptomatic were permitted on study [3, 8]. Further complicating the issue is the recent data indicating that docetaxel results in substantial survival gains when used for hormone-sensitive metastatic prostate cancer, raising the question of whether the early use of docetaxel for men with mCRPC should be liberalized [22, 23].
For now the question regarding whether to sequence docetaxel before or after next-generation AR-directed therapies (i.e. abiraterone or enzalutamide) is not fully answered. Additional clinical trials are needed before any definitive conclusions regarding how to best utilize docetaxel in the context of effective and readily available oral agents remain.

Next Generation AR-Directed Therapies

For the majority of men with mCRPC, AR-signaling still constitutes an important driver of disease progression. Both tissue androgens and AR increase in tumor tissue as prostate cancer transitions to the castration-resistant state, and AR-regulated genes such as PSA are often expressed at high levels in men with mCRPC [24]. These observations led to renewed interest in targeting AR-signaling in mCRPC patients and subsequently prompted the development of drugs like abiraterone (an inhibitor of extragonadal androgen biosynthesis) and enzalutamide (a pure AR-antagonist) [3, 4, 8, 9, 25].
Both abiraterone and enzalutamide were initially developed in the post-docetaxel space, with their respective pivotal Phase III trials demonstrating a survival advantage compared to controls [4, 9]. Given that these studies were limited to a population that was exposed to prior docetaxel, initial drug approval was limited to patients that had already received docetaxel. Subsequent approvals in the pre-docetaxel space was only granted following publication of additional Phase III data showing a survival benefit when these drugs were used pre-docetaxel [3, 8, 26]. Consensus guidelines, such as the National Comprehensive Cancer Network (NCCN) Prostate Cancer Guidelines and the American Urological Association (AUA), recommend reserving docetaxel for patients with symptomatic, rapidly progressive or visceral disease [19, 27]. As such, practice patterns are likely shifting toward the earlier (i.e. pre-docetaxel) use of both abiraterone and enzalutamide [18].
In regard to the question of whether to use abiraterone or enzalutamide first, there are no clinical trials directly comparing these two agents to one another. Both drugs function similarly by inhibiting the ligand (e.g. testosterone, dihydrotestosterone [DHT]) AR interaction, and it is probably safe to assume that both agents have comparable efficacy when used as a first line treatment for mCRPC. The decision to choose one agent over the other may ultimately hinge on considerations independent of any anti-tumor effect. Abiraterone requires concurrent treatment with prednisone in order to blunt the mineralocorticoid side effects associated with its use [4, 8]. Therefore, in patients for whom steroids are contraindicated or undesirable (e.g. those with diabetes), enzalutamide may be preferred. It is also notable that in the pre-docetaxel enzalutamide Phase III trial patients with visceral disease were permitted; whereas, in the pre-docetaxel abiraterone Phase III trial these men were excluded [3, 8]. There is, however, no clear evidence that abiraterone is ineffective in treating visceral disease.

Radium-223

Radium-223 is a novel alpha emitting radiopharmaceutical that possesses intrinsic bone homing properties similar to that of other alkaline earth elements, such as calcium, and is approved for the treatment of symptomatic, bone-metastatic CRPC. Radium-223 is not expected to affect soft tissue metastases, and on this basis, patients with visceral metastases or nodal metastases >3 cm in the short axis were excluded from the Phase III trial that led to radium-223’s approval [7]. As such, it is only approved for mCRPC patients with metastatic disease predominantly affecting bone.
In contrast to abiraterone and enzalutamide, radium-223 was not tested in separate Phase III trials specifically designed for men that were pre- and post-docetaxel [7]. At baseline, fifty-seven percent of men enrolled to the radium-223 arm and placebo arm of the Phase III study had received prior docetaxel and the remainder were docetaxel naïve. As with abiraterone or enzalutamide, the indication for radium-223 is therefore agnostic of prior docetaxel treatment status.
In addition to location of metastatic disease, another consideration before initiating radium-223 is the fact that it did not clearly result in improved pain endpoints in its Phase III trial as assessed by validated pain assessment instruments. Given that a Phase II trial did document a palliative benefit, however, an ongoing observational study was designed to assess radium-223 effect on pain (clinicaltrials.gov: NCT02398526) [28]. Until the results of this study are reported, it should not be assumed that radium-223 would lead to a meaningful improvement in pain.

Cabazitaxel

Cabazitaxel is a newer taxane that was shown to result in prolonged survival compared to mitoxantrone when used after docetaxel [5]. As such, it is only approved for use following disease progression on a docetaxel-based regimen. While overall cabazitaxel resulted in a decreased risk of death compared to mitoxantrone, there was a higher risk of death within 30 days of receiving the last dose, likely reflecting the toxicity of this agent. This is in contrast to radium-223, which demonstrated comparable frequency of grade 3 and 4 adverse events compared to placebo. Whether cabazitaxel would prove to be as toxic in a less heavily pre-treated group of patients remains to be seen, and prospective studies assessing its effectiveness pre-docetaxel are currently underway (FIRSTANA trial [clinicaltrials.gov: NCT01308567] and TAXYNERGY trial [clinicaltrials.gov: NCT01718353]). Depending on the results of these trials, cabazitaxel use could potentially be approved for docetaxel-naïve patients, but for now its use in this patient population remains off-label.

Sipuleucel-T

Sipuleucel-T is an ex vivo autologous immunotherapy product and remains the only cancer vaccine shown to improve overall survival in Phase III testing [6, 29]. It is designed to produce an immune response toward the prostate antigen PAP. The Phase III study only included mCRPC patients with asymptomatic disease and an anticipated life expectancy of ≥6 months. The median survival of the placebo and sipuleucel-t arms was long at 21.7 months and 25.8 months, respectively, likely reflecting the relatively good prognosis of the patients enrolled to the Phase III study [6]. Similar to the radium-223 Phase III trial, enrollment to the sipuleucel-t Phase III study was not limited based on prior docetaxel exposure, and ultimately 15.5 and 12.3 % of patients randomized to sipuleucel-t and placebo arms, respectively, were post-docetaxel. Interestingly no difference in disease progression was observed between study groups, and while this might call into question the overall survival benefit observed on this study, it should be noted that two additional randomized trials reported a survival advantage with sipuleucel-t [30, 31].
Based on the aforementioned, sipuleucel-t is not approved for patients with symptomatic disease, and given that only men with a life expectancy ≥6 months were included in the pivotal Phase III trial, it is not appropriate for patients with rapidly progressive disease or those expected to live <6 months. While it is technically approved pre- or post-docetaxel, in practice most men that have progressed on docetaxel are likely in need of a therapy that will result in objective tumor control.

Cross-Resistance

Surgical or medical castration (i.e. androgen deprivation therapy; ADT) as a treatment for prostate cancer was the first example of an effective targeted cancer therapy, and to this day inhibiting AR-signaling, primarily through disrupting ligand-AR interactions, remains the mainstay of treating advanced disease [25, 32]. Until recently, progression beyond frontline ADT was felt to represent an “androgen independent” or “hormone refractory” state; however, with the recognition that the AR transcriptional program is still operative in men that progress beyond ADT, the nomenclature has shifted such that these men are now referred to as having castration-resistant prostate cancer. As discussed above, many of the newer agents approved for the treatment of mCRPC function to inhibit AR-signaling, and not surprisingly, their long-term use can lead to the emergence of a drug resistant phenotype—manifested as diminished clinical activity when these drugs are used sequentially.

Abiraterone and Enzalutamide

Two of our most effective agents for treating mCRPC patients, abiraterone and enzalutamide, both function to inhibit AR ligands (e.g. DHT and testosterone) from binding the AR [25]. Abiraterone accomplishes this through inhibiting cytochrome P450-17 (CYP17), a key family of enzymes involved in gonadal, adrenal and intratumoral androgen synthesis [3336]. The end result is testosterone levels that are significantly lower than those observed with ADT alone [37, 38]. Enzalutamide on the other hand is a pure AR-antagonist, which, unlike earlier anti-androgens (e.g. bicalutamide, nilutamide and flutamide), is able to more completely antagonize the AR. In addition, it also prevents the nuclear translocation of the AR [39].
Given their similar mechanisms of action, it is not surprising that evidence of cross-resistance between abiraterone and enzalutamide has begun to emerge. A number of mechanisms of resistance have been described that may explain how continued AR-signaling occurs in spite of treatment with either abiraterone or enzalutamide. These include: upregulation of the AR; increased extragonadal androgen synthesis; the emergence of constitutively active AR splice variants (AR-Vs); AR point mutations; AR-signaling activation via alternative pathways (e.g. AKT/mTOR/Pi3 K, HER kinases); and activation of other nuclear hormone receptors such as the glucocorticoid receptor (GR) [14, 24, 4049]. One or more of these mechanisms may provide a basis for why progression on one AR-directed agent may portend a poor response to the other drug when used second line.
Recently, evidence of clinical cross-resistance between abiraterone and enzalutamide has begun to emerge (Tables 13.1 and 13.2). While no randomized studies have evaluated if the sequence with which abiraterone and enzalutamide are given influences overall survival (i.e. abiraterone then enzalutamide vs. enzalutamide then abiraterone), several retrospective analyses have demonstrated decreased activity to the second line agent. For instance, abiraterone was reported to have a median progression free survival of 5.6 months in the Phase III trial testing it post-docetaxel [4]. When abiraterone is used to treat mCRPC patient post-docetaxel and enzalutamide, however, the median progression free survival has been reported at only 2.7 to 3.6 months [50, 51]. Likewise, when enzalutamide is given post-docetaxel and abiraterone, the median progression free survival has been reported to be 2.8 to 4.6 months, which is in contrast to the 8.3 month median progress free survival that was reported in the Phase III trial testing enzalutamide in patients that were post-docetaxel only [9, 5256].
Table 13.1
Activity of abiraterone following treatment with enzalutamide and/or docetaxel
Reference
Agent
Clinical state
Study design
Sample size
Median overal
lsurvival (mos)
≥50 % PSA decline (%)
Median progression free survival (mos)
Objective response (%)
de Bono et al. [4]
Abiraterone
Post-docetaxel
Phase III
797
14.8
29
5.6
14
Loriot et al. [50]
Abiraterone
Post-enzalutamide and docetaxel
Retrospective review
38
7.2
8
2.7
8
Noonan et al. [51]
Abiraterone
Post-enzalutamide and docetaxel
Retrospective review
30
11.7
3
3.6
0
Note Sample size is only for the cohort treated with abiraterone
Table 13.2
Activity of enzalutamide following treatment with abiraterone and/or docetaxel
Reference
Agent
Clinical state
Study design
Sample size
Overall survival (mos)
≥50 % PSA decline (%)
Progression free survival (mos)
Objective response (%)
Scher et al. [9]
Enzalutamide
Post-docetaxel
Phase III
800
18.4
54
8.3
29
Schrader et al. [56]
Enzalutamide
Post-abiraterone and post-docetaxel
Retrospective review
35
7.1
28.6
4
2.9
Bianchini et al. [54]
Enzalutamide
Post-abiraterone and post-docetaxel
Retrospective review
39
12.8
2.8
4.3
Suzman et al. [71]
Enzalutamide
Post-abiraterone
Retrospective review
30
34
4.7
Badrising et al. [53]
Enzalutamide
Post-abiraterone and post-docetaxel
Retrospective review
61
7.4
21
2.8
Cheng et al. [55]b
Enzalutamide
Post-abiraterone
Retrospective review
79
Not reached
18
4a
Enzalutamide
Post-abiraterone and post-docetaxel
Retrospective review
165
28
2.8a
Azad et al. [52]b
Enzalutamide
Post-abiraterone
Retrospective review
47
8.6
26
6.6
Enzalutamide
Post-abiraterone and post-docetaxel
Retrospective review
68
10.6
22
4.6
Note Sample size is only for the cohort treated with enzalutamide
aTime to PSA progression is reported
bThese studies report on overlapping cohorts
Upregulation of full length AR (AR-FL) and/or the presence of AR-Vs may be drivers of abiraterone and enzalutamide cross-resistance [14]. In a prospective study reported by Antonarakis et al., the presence of AR-V7 (the most prevalent AR-V) mRNA was determined by qRT-PCR on circulating tumor cells, and correlated with response (i.e. ≥50 % decline in PSA from baseline) to abiraterone (N = 31) or enzalutamide (N = 31) [14]. They reported that the presence of AR-V7 mRNA associated with a lack of PSA response to both abiraterone (0 % vs. 68 %, P = 0.004) and enzalutamide (0 % vs. 53 %, P = 0.004). In addition, time to PSA progression, clinical or radiographic progression and overall survival were all significantly shorter in men harboring an AR-V7. That study also found that high AR-FL transcript levels associated with a lack of response to abiraterone and enzalutamide. Interestingly, when compared to abiraterone and enzalutamide naïve patients, the prevalence of AR-V7 was higher in those that had been pre-treated with either abiraterone (55 % vs. 9 %) or enzalutamide (50 % vs. 15 %). In addition, pre-treatment with either agent led to high AR-FL transcript levels. It is plausible that the emergence of AR-Vs or the upregulation AR-FL following exposure to abiraterone or enzalutamide may at least partially explain the diminishing efficacy seen when these agents are used sequentially.

Taxanes and AR-Directed Agents

Somewhat surprisingly, evidence of cross-resistance between docetaxel and abiraterone has also recently begun to emerge (Table 13.3). Docetaxel likely exerts an anti-tumor effect through a variety of mechanisms independent of AR-signaling (e.g. impairing mitosis and inhibiting expression of the anti-apoptotic genes Bcl-2 and Bcl-x) [5761]. More recently pre-clinical studies have also demonstrated that docetaxel is also able to inhibit microtubule mediated AR trafficking into the nucleus, in theory preventing AR-signaling [6266].
Table 13.3
Activity of docetaxel following treatment with abiraterone
Reference
Agent
Clinical state
Study design
Sample size
Overall survival (mos)
≥50 % PSA decline (%)
Progression free survival (mos)
Objective response (%)
Tannock et al. [2]
Docetaxel
Abiraterone naïve
Phase III
335*
18.9
45
7.7**
12
Mezynski et al. [68]
Docetaxel
Post-abiraterone
Retrospective review
35
12.5
26
4.6*
11
Schweizer et al. [18]
Docetaxel
Post-abiraterone
Retrospective review
24
38***
4.4***
Abiraterone naïve
95
63***
7.6***
Azad et al. [67]
Docetaxel
Post-abiraterone
Retrospective review
37
32
Note Sample size is only for the cohort treated with docetaxel
*Only reporting on the cohort that received docetaxel 75 mg/m2 every 21 days
**Time to PSA progression is reported
***P < 0.05
Similar to the aforementioned analyses that documented cross-resistance between abiraterone and enzalutamide, clinical evidence of cross-resistance between abiraterone and docetaxel has also begun to emerge. For instance, in the randomized Phase III trial that led to the approval of docetaxel, forty-five percent of patients had a ≥50 % decline in PSA (i.e. PSA response) from baseline [2]. This is in contrast to several retrospective analyses that have reported only 26–38 % of abiraterone pre-treated men achieving a PSA response to docetaxel [18, 67, 68]. In one retrospective analysis, the clinical outcomes on docetaxel for mCRPC patients who were either abiraterone naïve or post-abiraterone were compared [18]. In that study, the median time to PSA progression (6.7 vs. 4.1, P = 0.002), median clinical or radiographic progression free survival (7.6 vs. 4.4, P = 0.003) and PSA response (63 % vs. 38 %, P = 0.02) were all significantly better in the abiraterone naïve group. Of note, abiraterone treatment status remained a significant predictor of outcome when other clinically relevant covariates were controlled for through a multivariable model. It stands to reason that similar evidence of cross-resistance may also exist between enzalutamide and docetaxel, as well as between AR-directed agents and cabazitaxel; however, to our knowledge no such clinical data has been reported.

Conclusion

As the repertoire of approved agents for the treatment of mCRPC increases, so do the number of choices we face regarding which drug to select for any given patient. At this point, all approved therapies (except cabazitaxel) are indicated pre- and post-docetaxel. Recently presented consensus guidelines on the conduct of mCRPC clinical trials encourages investigators to avoid defining trial cohorts on the basis of chemotherapy treatment status—reflecting the new mCRPC therapeutic landscape [10]. To date, it remains unclear if an optimal sequence of mCRPC drugs exists; however, specific presenting clinical features may help steer the choice of when to use each agent.
Docetaxel is probably most appropriate if a rapid palliative response is needed (e.g. patients with rapidly progressive disease or visceral metastases) [2, 20]. On the other hand, abiraterone and enzalutamide both have excellent activity in patients with mCRPC, and in contrast to docetaxel, are generally better tolerated—often making them more attractive choices for first-line mCRPC therapy, particularly for patients with significant comorbidities or impaired performance status [3, 4, 8, 9]. Evidence of cross-resistance between drugs that inhibit AR-signaling is becoming more apparent, and emerging clinical data indicates that adaptive changes in AR expression and/or emergence of constitutively active AR-Vs may drive resistance to chronic exposure to AR-directed drugs. Preliminary data indicates that docetaxel may be a better choice in the face of a resistant phenotype; however, whether there is an optimal sequence with which to use abiraterone, enzalutamide and docetaxel remains to be seen [69].
Sipuleucel-t, radium-223 and cabazitaxel have somewhat more restricted indications. Sipuleucel-t is only approved for asymptomatic patients, and as such is probably only useful in a small subset of mCRPC patients that present with slowly progressing disease [6]. It is important to note that sipuleucel-t will not prevent disease progression, so the appropriateness of delaying the initiation of drugs that can control disease (e.g. abiraterone, enzalutamide, docetaxel) must be considered. Radium-223 only targets bone metastases, and is not approved for patients with visceral disease or metastatic adenopathy >3 cm in size—limiting its usefulness in heavily pre-treated patients that are more likely to have soft tissue metastases [7]. Cabazitaxel is only approved following progression on docetaxel. In addition, it is relatively toxic and affords only a modest improvement in overall survival [5].
With so many agents to choose from, work towards developing predictive biomarkers should be prioritized. Ongoing efforts to characterize the molecular landscape of mCRPC through metastatic biopsy programs will likely play an important role in defining which group of patients stand to benefit from specific therapeutic agents. For instance, in a recent report by Robinson et al. [70] patients with mCRPC underwent a targeted biopsy followed by an integrative genomic assessment. In addition to documenting several previously described genomic aberrations (e.g. AR, ETS genes, TP53 and PTEN), this study also found a higher than expected frequency of biallelic loss of DNA damage repair pathway genes (e.g. BRCA1, BRCA2 and ATM). This finding provides a rational basis for ‘precision oncology’ trials testing agents that either induce DNA damage or impair the DNA damage repair machinery (e.g. platinum chemotherapy, PARP inhibitor) in those with evidence of biallelic loss of DNA damage repair pathway genes.
Ultimately, prospective trials are needed to delineate the role each of the aforementioned drugs will play in treating mCRPC. Specific questions that need to be addressed include: (I) Are combinations of drugs better than their sequential use? (II) Is one sequence of drug use better than another? (III) Should every mCRPC patient receive every approved drug? (IV) Can biomarkers (e.g. AR expression level, AR-V status) lead to improved patient-drug selection? There has been a lot of progress made in the treatment of mCRPC over the past decade. Many questions remain, but with continued work we will learn how to most effectively use the plethora of agents now approved for mCRPC.
Literature
1.
Petrylak DP, Tangen CM, Hussain MH, et al. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N Engl J Med. 2004;351:1513–20.CrossRefPubMed
2.
Tannock IF, de Wit R, Berry WR, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med. 2004;351:1502–12.CrossRefPubMed
3.
Beer TM, Armstrong AJ, Rathkopf DE, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. 2014.
4.
de Bono JS, Logothetis CJ, Molina A, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med. 2011;364:1995–2005.CrossRefPubMedPubMedCentral
5.
de Bono JS, Oudard S, Ozguroglu M, et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet. 2010;376:1147–54.CrossRefPubMed
6.
Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–22.CrossRefPubMed
7.
Parker C, Nilsson S, Heinrich D, et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med. 2013;369:213–23.CrossRefPubMed
8.
Ryan CJ, Smith MR, de Bono JS, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med. 2013;368:138–48.CrossRefPubMedPubMedCentral
9.
Scher HI, Fizazi K, Saad F, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012;367:1187–97.CrossRefPubMed
10.
Scher HI, Morris MJ, Stadler WM, et al. Trial Design and Objectives for Castration-Resistant Prostate Cancer: Updated Recommendations From the Prostate Cancer Clinical Trials Working Group 3. J Clin Oncol Off J Am Soc Clin Oncol. 2016.
11.
Scher HI, Halabi S, Tannock I, et al. Design and end points of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the Prostate Cancer Clinical Trials Working Group. J Clin Oncol Off J Am Soc Clin Oncol. 2008;26:1148–59.CrossRef
12.
Nelson PS. Molecular states underlying androgen receptor activation: a framework for therapeutics targeting androgen signaling in prostate cancer. J Clin Oncol Off J Am Soc Clin Oncol. 2012;30:644–6.CrossRef
13.
Garraway LA, Sellers WR. Lineage dependency and lineage-survival oncogenes in human cancer. Nat Rev Cancer. 2006;6:593–602.CrossRefPubMed
14.
Antonarakis ES, Lu C, Wang H, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med. 2014;371:1028–38.CrossRefPubMedPubMedCentral
15.
Carreira S, Romanel A, Goodall J, et al. Tumor clone dynamics in lethal prostate cancer. Sci Transl Med. 2014;6:254ra125.
16.
Haffner MC, Mosbruger T, Esopi DM, et al. Tracking the clonal origin of lethal prostate cancer. J Clin Investig. 2013;123:4918–22.CrossRefPubMedPubMedCentral
17.
Baca SC, Prandi D, Lawrence MS, et al. Punctuated evolution of prostate cancer genomes. Cell. 2013;153:666–77.CrossRefPubMedPubMedCentral
18.
Schweizer MT, Zhou XC, Wang H, et al. The influence of prior abiraterone treatment on the clinical activity of docetaxel in men with metastatic castration-resistant prostate cancer. Eur Urol. 2014;66:646–52.CrossRefPubMedPubMedCentral
19.
Mohler JL, Kantoff PW, Armstrong AJ, et al. Prostate cancer, version 2.2014. J Nat Compr Cancer Netw JNCCN. 2014;12:686–718.
20.
Berthold DR, Pond GR, Roessner M, de Wit R, Eisenberger M, Tannock AI. Treatment of hormone-refractory prostate cancer with docetaxel or mitoxantrone: relationships between prostate-specific antigen, pain, and quality of life response and survival in the TAX-327 study. Clin Cancer Res. 2008;14:2763–7.CrossRefPubMed
21.
Logothetis CJ, Basch E, Molina A, et al. Effect of abiraterone acetate and prednisone compared with placebo and prednisone on pain control and skeletal-related events in patients with metastatic castration-resistant prostate cancer: exploratory analysis of data from the COU-AA-301 randomised trial. Lancet Oncol. 2012;13:1210–7.CrossRefPubMed
22.
Sweeney C, Chen Y, Carducci MA, et al. Impact on overall survival (OS) with chemohormonal therapy versus hormonal therapy for hormone-sensitive newly metastatic prostate cancer (mPrCa): an ECOG-led phase III randomized trial. American Society of Clinical Oncology Annual Meeting, Chicago, IL; 2014.
23.
James ND, Sydes MR, Mason MD, et al. Docetaxel and/or zoledronic acid for hormone-naïve prostate cancer: first overall survival results from STAMPEDE (NCT00268476). American Society of Clinical Oncology Annual Meeting, Chicago, IL; 2015.
24.
Chen CD, Welsbie DS, Tran C, et al. Molecular determinants of resistance to antiandrogen therapy. Nat Med. 2004;10:33–9.CrossRefPubMed
25.
Schweizer MT, Antonarakis ES. Abiraterone and other novel androgen-directed strategies for the treatment of prostate cancer: a new era of hormonal therapies is born. Ther Adv Urol. 2012;4:167–78.CrossRefPubMedPubMedCentral
26.
Ryan CJ, Smith MR, Fizazi K, et al. Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy-naive 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. 2015;16:152–60.CrossRefPubMed
27.
Cookson MS, Roth BJ, Dahm P, et al. Castration-resistant prostate cancer: AUA guideline. J Urol. 2013;190:429–38.CrossRefPubMed
28.
Parker CC, Pascoe S, Chodacki A, et al. A randomized, double-blind, dose-finding, multicenter, phase 2 study of radium chloride (Ra 223) in patients with bone metastases and castration-resistant prostate cancer. Eur Urol. 2013;63:189–97.CrossRefPubMed
29.
Schweizer MT, Drake CG. Immunotherapy for prostate cancer: recent developments and future challenges. Cancer Metastasis Rev. 2014;33:641–55.CrossRefPubMedPubMedCentral
30.
Higano CS, Schellhammer PF, Small EJ, et al. Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active cellular immunotherapy with sipuleucel-T in advanced prostate cancer. Cancer. 2009;115:3670–9.CrossRefPubMed
31.
Small EJ, Schellhammer PF, Higano CS, et al. Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J Clin Oncol Off J Am Soc Clin Oncol. 2006;24:3089–94.CrossRef
32.
Huggins C, Hodges CV. Studies on prostate cancer 1: the effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. Cancer Res. 1941;1:293–7.
33.
Haidar S, Ehmer PB, Barassin S, Batzl-Hartmann C, Hartmann RW. Effects of novel 17α-hydroxylase/C17, 20-lyase (P450 17, CYP 17) inhibitors on androgen biosynthesis in vitro and in vivo. J Steroid Biochem Mol Biol. 2003;84:555–62.CrossRefPubMed
34.
Ryan CJ, Tindall DJ. Androgen receptor rediscovered: the new biology and targeting the androgen receptor therapeutically. J Clin Oncol Off J Am Soc Clin Oncol. 2011;29:3651–8.CrossRef
35.
Vasaitis TS, Bruno RD, Njar VC. CYP17 inhibitors for prostate cancer therapy. J Steroid Biochem Mol Biol. 2011;125:23–31.CrossRefPubMedPubMedCentral
36.
Ye L, Su ZJ, Ge RS. Inhibitors of testosterone biosynthetic and metabolic activation enzymes. Molecules. 2011;16:9983–10001.CrossRefPubMed
37.
Attard G, Reid AH, Yap TA, et al. Phase I clinical trial of a selective inhibitor of CYP17, abiraterone acetate, confirms that castration-resistant prostate cancer commonly remains hormone driven. J Clin Oncol Off J Am Soc Clin Oncol. 2008;26:4563–71.CrossRef
38.
O’Donnell A, Judson I, Dowsett M, et al. Hormonal impact of the 17α-hydroxylase/C(17,20)-lyase inhibitor abiraterone acetate (CB7630) in patients with prostate cancer. Br J Cancer. 2004;90:2317–25.PubMedPubMedCentral
39.
Tran C, Ouk S, Clegg NJ, et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science (New York, NY). 2009;324:787–90.CrossRef
40.
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. 2008;68:4447–54.CrossRefPubMedPubMedCentral
41.
Stanbrough M, Bubley GJ, Ross K, et al. Increased expression of genes converting adrenal androgens to testosterone in androgen-independent prostate cancer. Cancer Res. 2006;66:2815–25.CrossRefPubMed
42.
Drake JM, Graham NA, Lee JK, et al. Metastatic castration-resistant prostate cancer reveals intrapatient similarity and interpatient heterogeneity of therapeutic kinase targets. Proc Natl Acad Sci USA. 2013;110:E4762–9.CrossRefPubMedPubMedCentral
43.
Hu R, Lu C, Mostaghel EA, et al. Distinct transcriptional programs mediated by the ligand-dependent full-length androgen receptor and its splice variants in castration-resistant prostate cancer. Cancer Res. 2012;72:3457–62.CrossRefPubMedPubMedCentral
44.
Joseph JD, Lu N, Qian J, et al. A clinically relevant androgen receptor mutation confers resistance to second-generation antiandrogens enzalutamide and ARN-509. Cancer Discov. 2013;3:1020–9.CrossRefPubMed
45.
Mehra R, Kumar-Sinha C, Shankar S, et al. Characterization of bone metastases from rapid autopsies of prostate cancer patients. Clin Cancer Res Off J Am Assoc Cancer Res. 2011;17:3924–32.CrossRef
46.
Taylor BS, Schultz N, Hieronymus H, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010;18:11–22.CrossRefPubMedPubMedCentral
47.
Yeh S, Lin HK, Kang HY, Thin TH, Lin MF, Chang C. From HER2/Neu signal cascade to androgen receptor and its coactivators: a novel pathway by induction of androgen target genes through MAP kinase in prostate cancer cells. Proc Natl Acad Sci USA. 1999;96:5458–63.CrossRefPubMedPubMedCentral
48.
Arora VK, Schenkein E, Murali R, et al. Glucocorticoid receptor confers resistance to antiandrogens by bypassing androgen receptor blockade. Cell. 2013;155:1309–22.CrossRefPubMedPubMedCentral
49.
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. 2011;17:5913–25.CrossRefPubMedPubMedCentral
50.
Loriot Y, Bianchini D, Ileana E, et al. Antitumour activity of abiraterone acetate against metastatic castration-resistant prostate cancer progressing after docetaxel and enzalutamide (MDV3100). Ann Oncol Off J Eur Soc Med Oncol/ESMO. 2013;24:1807–12.CrossRef
51.
Noonan KL, North S, Bitting RL, Armstrong AJ, Ellard SL, Chi KN. Clinical activity of abiraterone acetate in patients with metastatic castration-resistant prostate cancer progressing after enzalutamide. Ann Oncol Off J Eur Soc Med Oncol/ESMO. 2013;24:1802–7.CrossRef
52.
Azad AA, Eigl BJ, Murray RN, Kollmannsberger C, Chi KN. Efficacy of enzalutamide following abiraterone acetate in chemotherapy-naive metastatic castration-resistant prostate cancer patients. Eur Urol. 2015;67:23–9.CrossRefPubMed
53.
Badrising S, van der Noort V, van Oort IM, et al. Clinical activity and tolerability of enzalutamide (MDV3100) in patients with metastatic, castration-resistant prostate cancer who progress after docetaxel and abiraterone treatment. Cancer. 2014;120:968–75.CrossRefPubMed
54.
Bianchini D, Lorente D, Rodriguez-Vida A, et al. Antitumour activity of enzalutamide (MDV3100) in patients with metastatic castration-resistant prostate cancer (CRPC) pre-treated with docetaxel and abiraterone. Eur J Cancer (Oxford, England: 1990) 2014;50:78–84.
55.
Cheng HH, Gulati R, Azad A, et al. Activity of enzalutamide in men with metastatic castration-resistant prostate cancer is affected by prior treatment with abiraterone and/or docetaxel. Prostate Cancer Prostatic Dis. 2015;18:122–7.CrossRefPubMedPubMedCentral
56.
Schrader AJ, Boegemann M, Ohlmann CH, et al. Enzalutamide in castration-resistant prostate cancer patients progressing after docetaxel and abiraterone. Eur Urol. 2014;65:30–6.CrossRefPubMed
57.
Ringel I, Horwitz SB. Studies with RP 56976 (taxotere): a semisynthetic analogue of taxol. J Natl Cancer Inst. 1991;83:288–91.CrossRefPubMed
58.
Garcia P, Braguer D, Carles G, et al. Comparative effects of taxol and Taxotere on two different human carcinoma cell lines. Cancer Chemother Pharmacol. 1994;34:335–43.CrossRefPubMed
59.
Haldar S, Basu A, Croce CM. Bcl2 is the guardian of microtubule integrity. Cancer Res. 1997;57:229–33.PubMed
60.
Pienta KJ. Preclinical mechanisms of action of docetaxel and docetaxel combinations in prostate cancer. Semin Oncol. 2001;28:3–7.CrossRefPubMed
61.
Symes JC, Kurin M, Fleshner NE, Medin JA. Fas-mediated killing of primary prostate cancer cells is increased by mitoxantrone and docetaxel. Mol Cancer Ther. 2008;7:3018–28.CrossRefPubMed
62.
Gan L, Chen S, Wang Y, et al. Inhibition of the androgen receptor as a novel mechanism of taxol chemotherapy in prostate cancer. Cancer Res. 2009;69:8386–94.CrossRefPubMedPubMedCentral
63.
Zhu ML, Horbinski CM, Garzotto M, Qian DZ, Beer TM, Kyprianou N. Tubulin-targeting chemotherapy impairs androgen receptor activity in prostate cancer. Cancer Res. 2010;70:7992–8002.CrossRefPubMedPubMedCentral
64.
Darshan MS, Loftus MS, Thadani-Mulero M, et al. Taxane-induced blockade to nuclear accumulation of the androgen receptor predicts clinical responses in metastatic prostate cancer. Cancer Res. 2011;71:6019–29.CrossRefPubMedPubMedCentral
65.
Thadani-Mulero M, Nanus DM, Giannakakou P. Androgen receptor on the move: boarding the microtubule expressway to the nucleus. Cancer Res. 2012;72:4611–5.CrossRefPubMedPubMedCentral
66.
Thadani-Mulero M, Portella L, Sun S, et al. Androgen receptor splice variants determine taxane sensitivity in prostate cancer. Cancer Res. 2014.
67.
Azad AA, Leibowitz-Amit R, Eigl BJ, et al. A retrospective, Canadian multi-center study examining the impact of prior response to abiraterone acetate on efficacy of docetaxel in metastatic castration-resistant prostate cancer. Prostate. 2014;74:1544–50.CrossRefPubMed
68.
Mezynski J, Pezaro C, Bianchini D, et al. Antitumour activity of docetaxel following treatment with the CYP17A1 inhibitor abiraterone: clinical evidence for cross-resistance? Ann Oncol Off J Eur Soc Med Oncol/ESMO. 2012;23:2943–7.CrossRef
69.
Antonarakis ES, Lu C, Chen Y, et al. AR splice variant 7 (AR-V7) and response to taxanes in men with metastatic castration-resistant prostate cancer (mCRPC). 2015 Genitourinary Cancers Symposium; Orlando, FL.
70.
Robinson D, Van Allen EM, Wu YM, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;161:1215–28.CrossRefPubMed
71.
Suzman DL, Luber B, Schweizer MT, Nadal R, Antonarakis ES. Clinical activity of enzalutamide versus docetaxel in men with castration-resistant prostate cancer progressing after abiraterone. Prostate. 2014;74:1278–85.CrossRefPubMedPubMedCentral