Skip to main content

Advances in hormonal therapies for hormone naïve and castration-resistant prostate cancers with or without previous chemotherapy

Abstract

Hormonal manipulation plays a significant role in the treatment of advanced hormone naïve prostate cancer and castration-resistant prostate cancer (CRPC) with or without previous chemotherapy. Combination of gonadotropin releasing hormone (GnRH) agonists and androgen receptor (AR) antagonists (combined androgen blockade; CAB) is the first line therapy for advanced hormone naïve prostate cancer, but current strategies are developing novel GnRH antagonists to overcome disadvantages associated with GnRH agonist monotherapy and CAB in the clinical setting. Abiraterone acetate and enzalutamide are hormonal agents currently available for patients with CRPC and are both shown to improve overall survival versus placebo. Recently, in clinical trials, testosterone has been administered in cycles with existing surgical and chemical androgen deprivation therapies (ADT) (intermittent therapy) to CRPC patients of different stages (low risk, metastatic) to abate symptoms of testosterone deficiency and reduce cost of treatment from current hormonal therapies for patients with CRPC. This review will provide an overview on the therapeutic roles of hormonal manipulation in advanced hormone naïve and castration-resistant prostate cancers, as well as the development of novel hormonal therapies currently in preclinical and clinical trials.

Background

Prostate cancer is the most reported male cancer as well as the second leading cause of cancer-related deaths in Western men, excluding non-melanoma skin diseases [1]. Ever since 1941, when it was discovered that lowering testosterone levels via orchiectomy or estrogen injections improved symptoms in patients with metastatic, exogenous hormone naïve disease, androgen deprivation therapy (ADT) became the mainstay treatment for locally advanced prostate cancer (clinical tumor stages T3–T4, PSA > 20 ng/mL) [2]. However, despite reducing testosterone production to castrate levels (≤50 ng/dL), many patients will relapse and develop castration-resistant disease within 2–3 years post treatment, that is often more aggressive, currently incurable and has a poor prognosis with only 16–18 months of survival [3, 4]. This review will provide an overview on the therapeutic roles of hormonal manipulation in advanced hormone naïve and castration-resistant prostate cancers, as well as the development of novel hormonal therapies currently in preclinical and clinical trials. This will include the potential use of testosterone, although critically involved in cancer growth and progression, as a therapy for patients with castration-resistant prostate cancer (CRPC).

Physiology of androgen biosynthesis and action

Testosterone is a hormone produced primarily by Leydig cells of testes and in smaller quantities by the adrenal glands [5, 6]. The secretion of testosterone in testes and adrenal glands is regulated by luteinising hormone (LH) and adrenocorticotropic hormone (ACTH), respectively; both of which are derived from the pituitary gland [58]. Inactive androgen receptors (AR) are located in the cytoplasm and are bound by heat-shock protein (HSP) chaperones including HSP90, HSP70 and HSP40 [9]. Following activation by androgens, AR dissociates from the HSP complex, dimerizes and binds onto HSP27 which allows localization of AR to the nucleus and subsequent transcription of downstream target genes such as prostate-specific antigen (PSA), a glycoprotein that is also referred as kallikrein-3 (KLK3) [911]. Regulation of AR-specific genes is important for the development and maturation of prostate and other male sexual characteristics. Studies have shown that testosterone deficiency is associated with erectile dysfunction, low libido, reduced muscle and physical strength [12, 13]. Testosterone is further required for the growth and survival of prostate tumors, and ADT is commonly used to suppress disease progression among patients [2, 14, 15].

Interestingly, low testosterone levels have been linked to increased risk of developing high-grade prostate cancer [16, 17]. A study by Schatzl et al. [17] found that prostate cancer patients with partial androgen deficiency (n = 52; total serum testosterone <3.0 ng/mL) at diagnosis had significantly higher Gleason scores than patients with normal serum testosterone levels (n = 104; >3.0 ng/mL) (Gleason score 7.4 ± 1.2 vs. 6.2 ± 1.4, respectively; p = 0.001). Other studies have reported an association between low testosterone, prostate cancer risk and tumor reoccurrence [18, 19], suggesting that ADT may have adverse effects on patients with prostate cancer. Nevertheless ADT has been shown to significantly improve clinical outcomes in combination with other therapies (e.g. radiotherapy) which are discussed in further detail in this review.

Locally advanced hormone naïve prostate cancer

GnRH agonists—overview

Prior to the use of immunotherapy and chemotherapy, ADT is the first-line therapy for locally advanced or high-risk hormone naïve prostate cancer (clinical tumor stages T3–T4, PSA > 20 ng/mL) [20]. Gonadotropin releasing hormone (GnRH) agonists are the standard hormonal agents for ADT due to their reversibility, allowing intermittent use when required. GnRH agonists can also prevent complications associated with orchiectomy, such as physical discomfort [20, 21]. However recently, a 10-year study by Van Hemelrijck et al. has suggested that patients treated with GnRH agonists were at higher risk for deep-venous thrombosis (DVT) and pulmonary embolism than those who underwent orchiectomy (Absolute risk value 4.08 vs. 1.40, respectively) [22].

GnRH agonists are very long acting drugs which bind to the gonadotrophin receptors on the pituitary gland. Their initial binding causes a marked increase in luteinising hormones (LH) and follicle stimulating hormones (FSH) secretion, resulting in a transient increase in testosterone levels [23, 24]. This “flare” can induce adverse symptoms such as bone pain, spinal cord compression, cardiovascular events, and ureteral blockage among patients [24, 25]. Thus, to abate this effect, GnRH agonists are usually accompanied with AR antagonists and this is often referred as combined androgen blockade (CAB) therapy (Table 1). Within approximately 3–4 weeks, the pituitary GnRH receptors undergo desensitization and suppression, thus lowering serum testosterone to castrate levels (≤50 ng/dL) [26]. There are currently four United States (US) Food and Drug Administration (FDA) -approved GnRH agonists for advanced hormone naïve prostate cancer patients: goserelin acetate (Zoladex®), leuprolide (Lupron Depot®), histrelin acetate (Supprelin LA®) and triptorelin pamaote (Trelstar Depot®, Trelstar LA®) (Table 1) [2731].

Table 1 FDA-approved hormonal therapies for advanced hormone naïve prostate cancer and metastasis castration-resistant prostate cancer

GnRH agonists—clinical advancement for prostate cancer therapy

At present, anticancer strategies have taken an interest in supplementing GnRH agonists with concurrent radiotherapies to optimize efficacy of treatment for locally advanced or high-risk prostate cancer [32]. The 10-year overall survival rate for patients treated with radiotherapy and adjuvant ADT with goserelin, is evidently greater than radiation monotherapy (49–58.1 vs. 39–39.8 %, respectively; p < 0.002) [33, 34]. Likewise, a combination of ADT and radiotherapy is more effective when compared to ADT alone [35]. As shown by the SPCG-7 trial, the cumulative incidence for prostate cancer-specific mortality at 10 years was 23.9 % for the group treated with ADT alone (3.75 or 11.25 mg GnRH agonist leuprorelin plus 250 mg AR antagonist flutamide for 3 months) and 11.9 % in the group treated with ADT in combination with radiotherapy [35].

Further studies have sought to deliver goserelin to the tumor via nanoparticle carriers. In the clinical setting, nanoparticles are macromolecular, conductive materials (metal or semi-metal; size range 5–250 nm) that carry and deliver anticancer drugs to the vicinity of the tumor and solely penetrate target tissue by converting absorbed light photons at wavelengths near the infrared spectrum (800–2500 nm) into heat, and this is often referred as photothermal therapy [3638]. The selective delivery of nanoparticles to solid tumors among patients is highly dependent on their large molecular size and how cancer cells develop in contrast to normal cells [36]. Most solid tumors increase the production of new blood vessels (angiogenesis) to enhance vascular permeability and provide more nutrients to sustain their growth [39]. However, unlike normal blood vessels, the endothelial junctions of tumor blood vessels are more loose due to poor development and lack efficient lymphatic drainage, which allows the macromolecular nanoparticles to easily pass through as well as accumulate at the tumor site for effective treatment [39]. This phenomenon known as the “enhanced permeability and retention” (EPR) effect differentiates most solid tumors from normal tissues and is essential for the therapeutic properties of nanoparticles [36, 39].

Goserelin-conjugated gold rod-shaped nanoparticles (commonly known as nanorods) were used in a recent in vivo study by Wolfe et al. and these demonstrated a significant increase in radiosensitization of PC3 xenograft models, as compared to standard pegylated gold nanorods (1.36 ± 0.06 vs. 1.19 ± 0.04, respectively). Treatment with the goserelin-conjugated gold nanorods plus radiotherapy delayed tumor growth by 17 ± 1 days versus standard pegylated gold nanorods plus radiotherapy or radiotherapy alone in PC3 xenograft models (p < 0.001) [40]. Likewise, another study reported that goserelin loaded nanoparticles can induce growth inhibition and apoptosis in LNCaP and DU145 cell lines [41]. These preclinical findings are promising and warrant further investigation.

Efficacy of GnRH agonists in prostate cancer treatment is likely due to their specificity toward GnRH receptors found in the pituitary gland; however increasing evidence has indicated that the presence of GnRH receptors in tumors of the prostate, as demonstrated by the in vitro and in vivo studies mentioned previously, and of other organs, such as breast, uterus and ovary might be their additional molecular targets [4042]. There are currently two types of GnRH receptors (GnRH receptors I and II) found in prostate cancer cells in vitro [43, 44].

Better outcomes with radiotherapy are achieved with long-term adjuvant ADT (2–3 years) than short-term adjuvant ADT (<2 years). A phase III randomised, controlled trial (ClinicalTrials.gov, number NCT02175212) suggests that advanced or high-risk prostate cancer patients receiving long-term adjuvant ADT (3.6 mg subcutaneous goserelin; after 1 month, 10.8 mg was given every 3 months for 28 months) plus high dose radiotherapy (dose range 76–82 Gy) within 5 years had significantly improved biochemical disease-free survival (90 vs. 81 %, respectively; p = 0.01), metastasis-free survival (94 vs. 83 %, respectively; p = 0.009), and overall survival (95 vs. 86 %, respectively; p = 0.01) versus those in the short-term ADT plus high dose radiotherapy group (same analog and dose regimen, for 4 months) [45].

As with radiotherapy, chemotherapeutic drugs such as docetaxel and estramustine are far more beneficial with adjuvant goserelin ADT. A randomised phase III trial (ClinicalTrials.gov, number NCT00055731) with 413 advanced or high-risk prostate cancer patients were administered to either ADT (10.8 mg goserelin every 3 months for 36 months) with docetaxel (70 mg/m2; four cycles on Day 2) and estramustine (10 mg/kg per day; for 5 days every 3 weeks) or ADT alone [46]. In median follow-up of 8.8 years, patients who received ADT, docetaxel and estramustine had fewer relapses and mortality than those who received only ADT; 88 of 207 (43 %) patients versus 111 of 206 (54 %) patients, respectively [46]. 8-year relapse-free survival was 62 % in the ADT, docetaxel and estramustine group compared to 50 % in the ADT only group whilst reporting no treatment-related deaths (p = 0.017) [46].

Clinical observations have shown no significant differences in efficacy between the two GnRH agonists, leuprolide (3.75 and 7.5 mg) and goserelin (3.6 mg) [47]. It is noteworthy, however, that a substantial proportion of patients (26.3, 25 and 35 % of patients who received 3.75 mg leuprolide, 7.5 mg leuprolide, and 3.6 mg goserelin, respectively) did not achieve castration levels of testosterone (≤50 ng/dL) regardless of which GnRH agonist was used in the study [47]. Future research to improve the efficacy of chemical castration may therefore be needed.

GnRH antagonists—overview

There are two second-line GnRH antagonists for patients with locally advanced or high-risk prostate cancer, degarelix (Firmagon®) and abarelix (Plenaxis®), although the latter has been restricted to patients with no alternative therapy because of associated severe allergic reactions (Table 1) [48, 49], where 3 of the 81 (3.7 %) patients had immediate-onset histamine surges and systemic allergic reactions upon treatment [24, 50].

GnRH antagonists directly inhibit pituitary GnRH receptors and do not cause an initial LH, FSH and testosterone surge or “flare” as opposed to GnRH agonists, thereby suppressing testosterone to castrate levels (≤50 ng/dL) with minimal delay. A randomized, controlled phase III trial (CS21) has shown that patients receiving either low or high dose of the GnRH antagonist degarelix (Firmagon®) subcutaneously for locally advanced or high-risk prostate cancer (240 mg followed by either 80 or 160 mg monthly, respectively) were more likely to reach castration levels of testosterone by day 3 post treatment than those receiving a GnRH agonist (leuprolide; intramuscular; 7.5 mg monthly), with 95.5–96.1 % patients vs. 0 % patients, respectively [51]. Combined with this, PSA was decreased significantly faster in the degarelix group versus the leuprolide group [51].

GnRH antagonists—clinical advancement for prostate cancer therapy

To date, there are clear advantages of GnRH antagonists (particularly degarelix) over GnRH agonists [5156]. At 1 year follow-up, degarelix (subcutaneous; 240 mg followed by 80 mg every month) has reduced recurrence of elevated PSA in locally advanced or high-risk patients compared to those receiving leuprolide (7.5 mg every month) (7.7 vs. 12.9 %, respectively; baseline PSA > 20 ng/mL; p = 0.04) [52]. Likewise, a study by Albertson et al. revealed a significant reduction in cardiovascular risk after 1 year treatment with degarelix when compared to GnRH agonists (p = 0.002) [53]. Klotz et al. [54] reported that patients treated with degarelix had significant improvements in PSA progression-free survival (baseline PSA > 20 ng/mL; p = 0.052), overall survival (p = 0.023) and fewer musculoskeletal events and urinary tract events compared to the leuprolide group and the goserelin group.

GnRH agonists and GnRH antagonists have differential effects on FSH secretion. A study by Crawford et al. [57] reported a significant suppression in FSH levels from baseline among patients who switched from GnRH agonist leuprolide to GnRH antagonist degarelix for 1 year. Median FSH was 1.20 IU/L in the degarelix group (subcutaneous; 240 mg followed by 80 mg every month) and 4.40 IU/L the leuprolide group (intramuscular; 7.5 mg) (p < 0.0001) [57]. Elevated FSH levels were common among patients who received treatment with either a GnRH agonist or orchiectomy, or those who have switched from degarelix to leuprolide [58, 59]. In vitro studies detected overexpression of FSH receptors in prostate cancer compared to benign prostatic epithelium [60]. Moreover, a FSH value more than the lowest tertile (>4.8 mIU/mL) may be associated with shorter time to CRPC progression [61]. Taken together, the efficacy of GnRH antagonists may be due to lack of FSH stimulation as opposed to GnRH agonists.

Because degarelix is the only efficient GnRH antagonist for prostate cancer, alternative GnRH antagonists are currently in development [6266]. A 10-amino acid peptide (decapeptide) GnRH antagonist acyline is reported to significantly lower serum LH, FSH and testosterone levels below baseline values for 15 and 21 days among healthy men (cohort 1 and cohort 2) following treatment with acyline as either a single dose (subcutaneous; 300 µg/kg on day 0; cohort 1 baseline values: LH = 3.3 ± 0.7 IU/L, FSH = 2.7 ± 0.8 IU/L, testosterone = 21.6 ± 4.2 nmol/L) or multiple doses (subcutaneous; 75 µg/kg on days 0, 2, 4, 6 and 8; cohort 2 baseline values: LH = 4.0 ± 0.4 IU/L, FSH = 2.0 ± 0.3 IU/L, testosterone = 26.1 ± 4.2 nmol/L), respectively (p < 0.05) [65]. In a recent study by Festuccia et al. ozarelix, a decapeptide GnRH antagonist, significantly restores cell sensitivity to human recombinant tumor necrosis factor-related apoptosis inducing ligand (TRAIL)-mediated apoptosis in androgen-independent prostate cancer cell lines DU145 and PC3 (~60–70 % apoptotic cells; 20 ng/mL ozarelix plus 500 ng/mL TRAIL) versus cells treated with TRAIL alone (~10 % apoptotic cells; 500 ng/mL TRAIL) [66]. TRAIL is a member of the tumor necrosis factor (TNF) superfamily and resistance is usually acquired by cancer tissue to prevent apoptosis and subsequent cell death [67]. A novel non-peptide GnRH antagonist TAK-375 (Relugolix®) has recently completed phase I trials with healthy men and is progressing onto phase II [62]. Unlike peptide GnRH antagonist degarelix, non-peptide GnRH antagonists can be taken orally which can prevent disadvantages associated with subcutaneous and intramuscular routes of administration; for instance, adverse reactions to injection site and high dosage intake for efficacy [68].

GnRH antagonists are proven to be effective therapies clinically. However, the currently inevitable disease progression to castration-resistant prostate cancer (CRPC) renders this therapy, like all other therapies for advanced prostate cancer, inefficient. In a novel study, patients with locally advanced or high-risk prostate cancer underwent radical prostatectomy (RP) with or without prior neoadjuvant degarelix (subcutaneous 240 mg; administered 7 days before study) [69]. RP samples were collected from each patient for analysis. Degarelix-treated samples had lower mRNA expression of cell proliferation gene Ki-67 (MKI67) and cell cycle regulatory gene cyclin D1 (CCND1), as opposed to untreated samples (p = 0.003) [69]. Expression levels of AR-regulated genes PSA (KLK3) and fatty acid synthase (FASN) were significantly downregulated following degarelix treatment (p = 0.005 and p = 0.0002, respectively). However, malignant epithelial cells in degarelix-treated samples had higher estrogen receptor 1 (ESR1) expression versus untreated samples (24 vs. 8 %, respectively; p = 0.0002) [69]. This therefore indicates that GnRH antagonists may not inhibit intrinsic pathways mediated from growth promoters such as ESR1 which could assist in the development of castration resistance and further disease progression among patients.

Castration-resistant prostate cancer (CRPC)

Overview

Many studies have reported that CRPC remains androgen-dependent and is driven by intra-tumoral androgens [7073]. This phenomenon is due to AR reactivation which most commonly occurs through either overexpression or mutation of wild-type AR, with about 30 and 20–40 % present in CRPC patients, respectively [7476]. Overexpression of wild-type AR may be induced by the long noncoding HOX transcript antisense RNA (HOTAIR) which has been shown to prevent binding of AR protein to the E3 ubiquitin ligase MDM2 which directs AR’s proteasomal degradation [77]. AR overexpression often leads to enhanced hypersensitivity toward low circulating androgens, which enables prostate cancer cells to further progress despite the use of ADT [78, 79]. Chen et al. [78] found that AR shRNA-infected prostate cancer tumors in castrated male mice grew more slowly compared to the empty vector control. This indicates a distinct selection for cells that avoided AR knockdown for tumor growth. Consistent with this finding, in vivo and in vitro studies have shown that AR upregulates M-phase cell cycle regulatory genes such as CDC20, CDK1 and UBE2C and stimulate proliferation [80]. These observations suggest that increased AR regulation is important for developing resistance to hormonal therapy.

Mutations in the ligand-binding domain (LBD) of AR are known to switch non-steroidal AR antagonists into agonists; a phenomenon known as anti-androgen withdrawal syndrome (AAWS). Recent studies indicate that AAWS occurs with AR antagonists enzalutamide (MDV3100) and ARN-509 due to an AR-F876L point mutation [81, 82]. Interestingly, exposure to CAB therapy (GnRH agonist plus AR antagonist) has minimal effect on the secretion of testosterone precursor androstenediol and is shown to stimulate antagonist-to-agonist switch T877 mutation in AR [83]. A clinical study by Taplin et al. reported an increase in the AR-T877A point mutation among 5 of 16 (31.3 %) patients administrated with CAB therapy (ADT via LnRH agonist or orchiectomy plus AR antagonist flutamide) versus 0 of 17 (0 %) patients administered to ADT only [84]. Although CAB therapy can avoid testosterone surges when compared to ADT monotherapy, its use remains controversial due to increased risk of AAWS and resistance to castration.

Other mechanisms of CRPC involve AR splice variants (AR-Vs) which lack the C-terminus including the LBD and are constitutively active in the absence of androgens [85]. Detection of AR splice variants 6 (AR-V6; contiguously-spliced AR exons 1/2/3/2b) and 7 (AR-V7 contiguously-spliced AR exons 1/2/3/CE3) in CRPC tumors is associated with resistance to enzalutamide, an AR antagonist that specifically targets the LBD of AR [86, 87]. Recently, third generation AR antagonists were developed that are directed against domains other than the LBD of AR to improve efficacy of CRPC treatment. EPI-001 is a novel AR antagonist that targets the N-terminal domain of AR [88]. Consisting of four stereoisomers (EPI-002, EPI-003, EPI-004 and EPI-005), EPI-001 is revealed to inhibit transcription of AR-regulated genes in response to synthetic androgen R1881 [88]. EPI-506, a derivative of EPI-001, also targets the N-terminal domain of AR and is currently in phase I trials (ClinicalTrials.gov, number NCT02606123) [89]. The DNA-binding domain (DBD) is an alternative site for inhibition of AR-Vs. Recent evidences have shown that DBD-targeting compound VPC-14449 (4-(4-(4,5-bromo-1H-imidazol-1-yl)thiazol-2-yl)morpholine) (100 mg/kg; 4 days) suppressed tumor size and serum PSA levels in LNCaP xenograft models [90]. Existing antimicrobial drugs can induce suppression of full length AR (AR with C-terminal of LBD) and AR splice variant expression including nigericin (antibiotic) and niclosamide (anthelmintics) [91, 92].

Abiraterone acetate

Abiraterone acetate (Zytiga®) is an inhibitor of CYP17 (17α-hydroxy/17,20-lyase), an enzyme responsible for the intratumoral androgen synthesis of adrenal androgens and testosterone precursor dehydroepiandrosterone (DHEA) [93]. Clinically, abiraterone acetate (oral; four 250 mg tablets once daily with oral prednisone 5 mg twice daily) is shown to significantly improve median overall survival compared to placebo in metastatic CRPC (mCRPC; histologically confirmed prostate cancer, N1/M1, more than two [or three in other studies] consecutive rises in PSA levels over 25 % above nadir value following surgical or chemical ADT for at least 1 week) patients with or without previous chemotherapy [9497]. A randomized phase III trial demonstrates that abiraterone acetate significantly reduced pain intensity in chemotherapy naïve mCRPC patients versus the placebo group (26.7 vs. 18.4 months, respectively; p = 0.0490) [98]. Abiraterone acetate received FDA approval for mCRPC patients with or without previous chemotherapy in 2011 and 2012, respectively [27, 94, 96, 99] (Table 1).

Since its approval, the treatment of mCRPC patients with abiraterone acetate has become popular. Truven Health Analytics MarketScan® and electronic medical record (EMR) databases in the US have shown that 67 % mCRPC patients had abiraterone acetate as first-line therapy in 2013 compared to only 15 % using chemotherapeutic drug docetaxel [100]. In 2010, 1 year before the approval of abiraterone acetate, about 91 % mCRPC patients received docetaxel as first-line therapy [100]. The positive clinical outcomes associated with abiraterone acetate highlights the important role of androgen biosynthesis in CRPC.

Enzalutamide

Enzalutamide (Xtandi®) is an AR antagonist that has been approved by FDA for post-chemotherapeutic and chemotherapy naïve mCRPC patients in 2012 and 2014, respectively (Table 1) [101, 102]. A double-blind, phase II study, AFFIRM, has shown that enzalutamide (oral; four 40 mg capsules for median 8.3 months) improved median overall survival compared to the placebo group with previous chemotherapy (18.4 vs. 13.6 months, respectively; p < 0.001) [101]. A similar trend occurred for mCRPC patients without previous chemotherapy in another study, PREVAIL (32.4 vs. 30.2 months, respectively) [102].

There is debate on whether enzalutamide is more effective among patients with advanced hormone naïve prostate cancer than standard ADT (GnRH agonist plus AR antagonist), with a novel randomized phase II trial currently being performed (ClinicalTrials.gov, number NCT02278185) [103].

Potential use of testosterone for CRPC therapy

For decades it was thought that testosterone played a pivotal role in prostate cancer onset; however, recent studies have found no statistically significant association between testosterone administration (testosterone replacement therapy) and prostate cancer risk or mortality [104107]. A recent study has shown that testosterone might have therapeutic benefits for patients with asymptomatic mCRPC (histologically confirmed prostate cancer, rising PSA levels following surgical or chemical ADT for at least 4 weeks, castrate serum testosterone ≤50 ng/dL, no cancer-related pain, ≤5 asymptomatic bone metastases, ≤10 total sites of metastases including soft tissue) [108]. In 6 of the 14 (42.9 %) patients there were PSA reductions after three 28-day cycles of testosterone (intramuscular; 400 mg) plus 14 days of the chemotherapy drug etoposide (oral; 100 mg) [108]. Of these 14 patients, 4 (28.6 %) patients had >50 % PSA reductions from mean baseline (21.7 ng/mL; range 1.4–819.1 ng/mL). Low-grade (grade 1–2) alopecia (56.3 %), fatigue (56.3 %) and nausea (62.5 %) were most common among patients post treatment, followed by lower incidences of high-grade (grade 3–4) neutropenia (12.5 %) and pulmonary embolism (12.5 %) [108]. Likewise, a prior study by Morris et al. reported low-grade fatigue as the most common toxicity among all three cohorts of patients (75 %; n = 3 [cohort 1], n = 3 [cohort 2] and n = 6 [cohort 3]) with progressive mCRPC (histologically confirmed prostate cancer, 25 % increase in PSA over three tests) following high-dose exogenous testosterone (transdermal; 5 mg; patch [cohorts 1 and 2] or gel [cohort 3]; 1 week, 4 weeks, or until progression for cohorts 1, 2 and 3, respectively) [109]. No high-grade toxicities related to treatment were observed [109]. PSA suppression was seen in 1 of 3 (33.3 %), 2 of 3 (66.7 %) and 4 of 6 (66.7 %) patients from cohorts 1, 2 and 3, respectively [109]. However, despite this unique finding, the timing in which testosterone supplementation occurs is critical. Patients must previously be treated with ADT via surgical castration or GnRH agonist for at least 1 year prior to study to avoid potential adverse effects associated with significantly higher levels of testosterone or flares among patients with existing bone metastases and nodal metastases causing increased rates of fractures at metastatic site and ureteral obstruction, respectively [108, 109]. Higher levels of testosterone may also lead to increased risk of cancer progression. A number of publications have documented the potential for testosterone therapy to induce progression among patients with hormone naïve prostate cancer [2, 15]. On the contrary, a randomized phase I study reported no significant relationship between dose or serum testosterone levels and median time to progression (p = 0.072 and p = 0.14, respectively) among patients with low risk CRPC (histologically confirmed prostate cancer, PSA ≤ 3.0 ng/mL; rising PSA after surgical or chemical ADT) [110]. The recent studies highlight that testosterone is well tolerated among patients with asymptomatic and progressive mCRPC, and low risk CRPC but future investigation is needed [108110].

In patients with increasing PSA levels due to relapse following local therapy (radical prostatectomy, radiation therapy) treatment with testosterone decreased PSA to undetectable levels (<0.05 ng/ml). Feltquate et al. have shown that 2 of 9 (22.2 %) patients with non-castrate testosterone levels (mean 382 ng/dL; range 181–654 ng/dL) and PSA relapse after radiation therapy achieved undetectable PSA levels after 28-day cycles of testosterone repletion (transdermal gel; 5 g daily) on days 1 to 7 plus GnRH agonist (either leuprolide 7.5 mg intramuscular or goserelin 3.6 mg subcutaneous) on day 1 [111]. With the same dose and regimen, 8 of 17 (47 %) patients with PSA relapse following radical prostatectomy had achieved undetectable PSA levels [111]. However, prior to testosterone repletion, patients were required to undergo 12-week induction cycles of GnRH agonist (leuprolide 7.5 mg intramuscular or goserelin 3.6 mg subcutaneous) on day 1 plus AR antagonist bicalutamide (50 mg daily) on days 1 to 28 to deplete PSA levels to <1 ng/mL [111]. Patients who failed to achieve a PSA of <1 ng/mL after the initial induction did not received testosterone repletion in the study [111]. Likewise, a phase II trial reported that 5 of 38 (13 %) non-castrate patients (testosterone levels >150 ng/dL) with histologically confirmed prostate cancer and PSA relapse following prostatectomy or radiation therapy achieved “treatment-specific” undetectable PSA endpoint (PSA < 0.05 ng/mL or PSA < 0.5 ng/mL for patients with prior prostatectomy or radiation therapy, respectively) at 18 months after receiving nine 3-week cycles of testosterone repletion (transdermal gel; 5 g daily for 3 days) in combination with 3-month depot leuprolide (intramuscular; 22.5 mg) on Day 1 of cycles 1, 5 and 9, and docetaxel (70 mg/m2) on Day 1 per cycle [112]. Of these 5 patients, 3 had prostatectomy and 2 had radiation therapy prior to study [112].

As demonstrated by recent clinical trials, testosterone administration is generally supplemented with cycles of ADT to deplete serum androgen and PSA levels for a certain period of time. The process of cycling between androgen repletion and depletion, known as intermittent therapy, was first introduced in 1986 by Klotz et al. [113] to minimize toxicity and adverse side effects associated with continuous ADT. However, in addition to stopping the use of ADT, recent studies have taken interest in modifying androgen repletion with testosterone therapy to not only alleviate side effects associated with ADT but also to potentially reduce cost of hormonal treatment for CRPC patients (Table 2) [114123]. Interestingly, intermittent therapy is thought to prolong androgen dependence in prostate cancer when compared to continuous ADT; however, at present there is no statistically significant evidence to suggest that intermittent therapy without testosterone administration during the repletion stage improves overall survival of patients with PSA relapse following local therapy versus continuous ADT [124, 125]. Long term randomised clinical trials should be performed to determine if testosterone administration during intermittent therapy can significantly affect overall survival among patients with prostate cancer.

Table 2 Prices of several hormonal agents for prostate cancer in Australia (as of January 2016)

Conclusions

Hormonal therapy is the mainstay of treatment in the clinical management of pre- and post-castration-resistant prostate cancers. Until recently, GnRH agonists comprised the first-line therapy for advanced hormone naïve prostate cancer and are generally combined with AR antagonists to avoid initial testosterone flares, and either radiotherapy or chemotherapy to improve clinical outcomes. Progress has been made since the discovery of degarelix with the development of novel GnRH antagonists such as the non-peptide GnRH antagonist TAK-375 which is entering phase II trials. Due to advanced knowledge on AR reactivation and castration resistance, two hormonal agents have been approved by FDA for patients with CRPC: abiraterone acetate and enzalutamide. Abiraterone acetate has particularly gained vast interest because it directly inhibits biosynthesis of testosterone precursors, unlike current hormonal therapies. Although still early in clinical trials, the use of testosterone therapy could be expanded to patients with CRPC. It would be interesting to see if testosterone therapy could be translated into larger randomized clinical trials for further analysis, and subsequently allow the development of more effective therapeutic methods to manage the disease.

Abbreviations

ACTH:

adrenocorticotropic hormones

ADT:

androgen deprivation therapies

AR:

androgen receptor

CRPC:

castration-resistant prostate cancer

CAB:

combined androgen blockade

DHEA:

dehydroepiandrosterone

DBD:

DNA-binding domain

FSH:

follicle stimulating hormones

GnRH:

gonadotropin releasing hormone

HSP:

heat-shock protein

KLK3:

kallikrein-3

LH:

luteinising hormones

PSA:

prostate-specific antigen

References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65:5–29.

    Article  PubMed  Google Scholar 

  2. Huggins C, Hodges CV. Studies on prostatic cancer. Cancer Res. 1941;1:297.

    Google Scholar 

  3. Feldman BJ, Feldman D. The development of androgen-independent prostate cancer. Nat Rev Cancer. 2001;1:34–45.

    Article  CAS  PubMed  Google Scholar 

  4. Di Lorenzo G, Buonerba C, Autorino R, De Placido S, Sternberg CN. Castration-resistant prostate cancer. Drugs. 2010;70:983–1000.

    Article  PubMed  Google Scholar 

  5. Mendelson C, Dufau M, Catt K. Gonadotropin binding and stimulation of cyclic adenosine 3′:5′-monophosphate and testosterone production in isolated Leydig cells. J Biol Chem. 1975;250:8818–23.

    CAS  PubMed  Google Scholar 

  6. Rainey WE, Nakamura Y. Regulation of the adrenal androgen biosynthesis. J Steroid Biochem Mol Biol. 2008;108:281–6.

    Article  CAS  PubMed  Google Scholar 

  7. Saez JM. Leydig cells: endocrine, paracrine, and autocrine regulation. Endocr Rev. 1994;15:574–626.

    Article  CAS  PubMed  Google Scholar 

  8. Franchimont P. Regulation of gonadal androgen secretion. Horm Res Paediatr. 1983;18:7–17.

    Article  CAS  Google Scholar 

  9. Azad AA, Zoubeidi A, Gleave ME, Chi KN. Targeting heat shock proteins in metastatic castration-resistant prostate cancer. Nat Rev Urol. 2015;12:26–36.

    Article  CAS  PubMed  Google Scholar 

  10. Trapman J, Cleutjens KB. Androgen-regulated gene expression in prostate cancer. In: Seminars in cancer biology. Amsterdam: Elsevier; 1997. p. 29–36.

  11. Balk SP, Ko Y-J, Bubley GJ. Biology of prostate-specific antigen. J Clin Oncol. 2003;21:383–91.

    Article  CAS  PubMed  Google Scholar 

  12. Smith J, Bennett S, Evans L, Kynaston H, Parmar M, Mason M, et al. The effects of induced hypogonadism on arterial stiffness, body composition, and metabolic parameters in males with prostate cancer. J Clin Endocrinol Metab. 2001;86:4261–7.

    Article  CAS  PubMed  Google Scholar 

  13. Traish AM, Guay A, Feeley R, Saad F. The dark side of testosterone deficiency: I. metabolic syndrome and erectile dysfunction. J Androl. 2009;30:10–22.

    Article  CAS  PubMed  Google Scholar 

  14. Heidenreich A, Bastian PJ, Bellmunt J, Bolla M, Joniau S, van der Kwast T, et al. EAU guidelines on prostate cancer. Part 1: screening, diagnosis, and local treatment with curative intent—update 2013. Eur Urol. 2014;65:124–37.

    Article  PubMed  Google Scholar 

  15. Prout GR, Brewer WR. Response of men with advanced prostatic carcinoma to exogenous administration of testosterone. Cancer. 1967;20:1871–8.

    Article  PubMed  Google Scholar 

  16. Hoffman MA, DeWolf WC, Morgentaler A. Is low serum free testosterone a marker for high grade prostate cancer? J Urol. 2000;163:824–7.

    Article  CAS  PubMed  Google Scholar 

  17. Schatzl G, Madersbacher S, Thurridl T, Waldmüller J, Kramer G, Haitel A, et al. High-grade prostate cancer is associated with low serum testosterone levels. Prostate. 2001;47:52–8.

    Article  CAS  PubMed  Google Scholar 

  18. Teloken C. Low serum testosterone levels are associated with positive surgical margins in radical retropubic prostatectomy: hypogonadism represents bad prognosis in prostate cancer. J Urol. 2005;174:2178–80.

    Article  CAS  PubMed  Google Scholar 

  19. Morgentaler A. Prevalence of prostate cancer among hypogonadal men with prostate-specific antigen levels of 4.0 ng/mL or less. Urology. 2006;68:1263–7.

    Article  PubMed  Google Scholar 

  20. Heidenreich A, Bastian PJ, Bellmunt J, Bolla M, Joniau S, van der Kwast T, et al. EAU guidelines on prostate cancer. Part II: treatment of advanced, relapsing, and castration-resistant prostate cancer. Eur Urol. 2014;65:467–79.

    Article  CAS  PubMed  Google Scholar 

  21. Seidenfeld J, Samson DJ. Single-therapy androgen suppression in men with advanced prostate cancer: a systematic review and. Ann Intern Med. 2000;132:566–77.

    Article  CAS  PubMed  Google Scholar 

  22. Van Hemelrijck M. Risk of thromboembolic diseases in men with prostate cancer: results from the population-based PCBaSe Sweden. Lancet Oncol. 2010;11:450–8.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Schulze H, Senge T. Influence of different types of antiandrogens on luteinizing hormone-releasing hormone analogue-induced testosterone surge in patients with metastatic carcinoma of the prostate. J Urol. 1990;144:934–41.

    CAS  PubMed  Google Scholar 

  24. Van Poppel H, Nilsson S. Testosterone surge: rationale for gonadotropin-releasing hormone blockers? Urology. 2008;71:1001–6.

    Article  PubMed  Google Scholar 

  25. Cook T, Sheridan WP. Development of GnRH antagonists for prostate cancer: new approaches to treatment. Oncologist. 2000;5:162–8.

    Article  CAS  PubMed  Google Scholar 

  26. Limonta P, Manea M. Gonadotropin-releasing hormone receptors as molecular therapeutic targets in prostate cancer: current options and emerging strategies. Cancer Treat Rev. 2013;39:647–63.

    Article  CAS  PubMed  Google Scholar 

  27. US Food and Drug Administration. Approved drug products with therapeutic equivalence evaluations: orange book [Electronic version]. 2016. http://www.accessdata.fda.gov/scripts/cder/ob/default.cfm. Accessed 18 Jan 2016.

  28. US Food and Drug Administration. Goserelin: highlights of prescribing information 2015. http://www.accessdata.fda.gov/drugsatfda_docs/label/2015/019726s059,020578s037lbl.pdf. Accessed 18 Jan 2016.

  29. US Food and Drug Administration. Lupron depot: highlights of prescribing information 2014. http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/020517s036_019732s041lbl.pdf. Accessed 18 Jan 2016.

  30. US Food and Drug Administration. Supprelin LA (histrelin acetate): highlights of prescribing information 2012. http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/022058s009lbl.pdf. Accessed 18 Jan 2016.

  31. Millar J. Triptorelin approved for prostate cancer treatment. Am J Health Syst Pharm. 2000;57:1386.

    CAS  PubMed  Google Scholar 

  32. Mohiuddin JJ, Baker BR, Chen RC. Radiotherapy for high-risk prostate cancer. Nat Rev Urol. 2015;12:145–54.

    Article  PubMed  Google Scholar 

  33. Pilepich MV, Winter K, Lawton CA, Krisch RE, Wolkov HB, Movsas B, et al. Androgen suppression adjuvant to definitive radiotherapy in prostate carcinoma—long-term results of phase III RTOG 85–31. Int J Radiat Oncol Biol Phys. 2005;61:1285–90.

    Article  CAS  PubMed  Google Scholar 

  34. Bolla M, Van Tienhoven G, Warde P, Dubois JB, Mirimanoff R-O, Storme G, et al. External irradiation with or without long-term androgen suppression for prostate cancer with high metastatic risk: 10-year results of an EORTC randomised study. Lancet Oncol. 2010;11:1066–73.

    Article  CAS  PubMed  Google Scholar 

  35. Widmark A. Endocrine treatment, with or without radiotherapy, in locally advanced prostate cancer (SPCG-7/SFUO-3): an open randomised phase III trial. Lancet. 2009;373:301–8.

    Article  CAS  PubMed  Google Scholar 

  36. Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J Control Release. 2000;65:271–84.

    Article  CAS  PubMed  Google Scholar 

  37. Gullotti E, Yeo Y. Extracellularly activated nanocarriers: a new paradigm of tumor targeted drug delivery. Mol Pharm. 2009;6:1041–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Alexis F, Pridgen E, Molnar LK, Farokhzad OC. Factors affecting the clearance and biodistribution of polymeric nanoparticles. Mol Pharm. 2008;5:505–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Greish K. Enhanced permeability and retention of macromolecular drugs in solid tumors: a royal gate for targeted anticancer nanomedicines. J Drug Target. 2007;15:457–64.

    Article  CAS  PubMed  Google Scholar 

  40. Wolfe T, Chatterjee D, Lee J, Grant JD, Bhattarai S, Tailor R, et al. Targeted gold nanoparticles enhance sensitization of prostate tumors to megavoltage radiation therapy in vivo. Nanomed Nanotechnol Biol Med. 2015;11:1277–83.

    Article  CAS  Google Scholar 

  41. Tomar P, Jain N, Agarwal G, Dixit V. Goserelin loaded nanoparticles inhibit growth and induce apoptosis in human prostate cancer cell lines. Drug Deliv Transl Res. 2012;2:265–71.

    Article  CAS  PubMed  Google Scholar 

  42. Cheng CK, Leung PCK. Molecular biology of gonadotropin-releasing hormone (GnRH)-I, GnRH-II, and their receptors in humans. Endocr Rev. 2005;26:283–306.

    Article  CAS  PubMed  Google Scholar 

  43. Fekete M, Redding TW, Comaru-Schally AM, Pontes JE, Connelly RW, Srkalovic G, et al. Receptors for luteinizing hormone-releasing hormone, somatostatin, prolactin, and epidermal growth factor in rat and human prostate cancers and in benign prostate hyperplasia. Prostate. 1989;14:191–208.

    Article  CAS  PubMed  Google Scholar 

  44. Darby S, Stockley J, Khan MM, Robson CN, Leung HY, Gnanapragasam VJ. Expression of GnRH type II is regulated by the androgen receptor in prostate cancer. Endocr Relat Cancer. 2007;14:613–24.

    Article  CAS  PubMed  Google Scholar 

  45. Zapatero A, Guerrero A, Maldonado X, Alvarez A, San Segundo CG, Rodríguez MAC, et al. High-dose radiotherapy with short-term or long-term androgen deprivation in localised prostate cancer (DART01/05 GICOR): a randomised, controlled, phase 3 trial. Lancet Oncol. 2015;16:320–7.

    Article  CAS  PubMed  Google Scholar 

  46. Fizazi K, Faivre L, Lesaunier F, Delva R, Gravis G, Rolland F, et al. Androgen deprivation therapy plus docetaxel and estramustine versus androgen deprivation therapy alone for high-risk localised prostate cancer (GETUG 12): a phase 3 randomised controlled trial. Lancet Oncol. 2015;16:787–94.

    Article  CAS  PubMed  Google Scholar 

  47. Silva É, Ferreira U, Matheus W, Faria E, Silva G, Saito M, et al. Goserelin versus leuprolide in the chemical castration of patients with prostate cancer. Int Urol Nephrol. 2012;44:1039–44.

    Article  CAS  PubMed  Google Scholar 

  48. US Food and Drug Administration. Plenaxis (abarelix) injectable suspension. 2015. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2003/21-320_Plenaxis.cfm. Accessed 18 Jan 2016.

  49. Steinberg M. Degarelix: a gonadotropin-releasing hormone antagonist for the management of prostate cancer. Clin Ther. 2009;31:2312–31.

    Article  CAS  PubMed  Google Scholar 

  50. Huhtaniemi I, White R, McArdle CA, Persson B-E. Will GnRH antagonists improve prostate cancer treatment? Trends Endocrinol Metab. 2009;20:43–50.

    Article  CAS  PubMed  Google Scholar 

  51. Klotz L, Boccon-Gibod L, Shore ND, Andreou C, Persson B-E, Cantor P, et al. The efficacy and safety of degarelix: a 12-month, comparative, randomized, open-label, parallel-group phase III study in patients with prostate cancer. BJU Int. 2008;102:1531–8.

    Article  CAS  PubMed  Google Scholar 

  52. Tombal B, Miller K, Boccon-Gibod L, Schröder F, Shore N, Crawford ED, et al. Additional analysis of the secondary end point of biochemical recurrence rate in a phase 3 trial (CS21) comparing degarelix 80 mg versus leuprolide in prostate cancer patients segmented by baseline characteristics. Eur Urol. 2010;57:836–42.

    Article  PubMed  Google Scholar 

  53. Albertsen PC, Klotz L, Tombal B, Grady J, Olesen TK, Nilsson J. Cardiovascular morbidity associated with gonadotropin releasing hormone agonists and an antagonist. Eur Urol. 2014;65:565–73.

    Article  CAS  PubMed  Google Scholar 

  54. Klotz L, Miller K, Crawford ED, Shore N, Tombal B, Karup C, et al. Disease control outcomes from analysis of pooled individual patient data from five comparative randomised clinical trials of degarelix versus luteinising hormone-releasing hormone agonists. Eur Urol. 2014;66:1101–8.

    Article  CAS  PubMed  Google Scholar 

  55. Schröder FH, Tombal B, Miller K, Boccon-Gibod L, Shore ND, Crawford ED, et al. Changes in alkaline phosphatase levels in patients with prostate cancer receiving degarelix or leuprolide: results from a 12-month, comparative, phase III study. BJU Int. 2010;106:182–7.

    Article  PubMed  CAS  Google Scholar 

  56. Cui Y, Zong H, Yan H, Li N, Zhang Y. Degarelix versus goserelin plus bicalutamide therapy for lower urinary tract symptom relief, prostate volume reduction and quality of life improvement in men with prostate cancer: a systematic review and meta-analysis. Urol Int. 2014;93:152–9.

    Article  CAS  PubMed  Google Scholar 

  57. Crawford ED, Tombal B, Miller K, Boccon-Gibod L, Schröder F, Shore N, et al. A phase III extension trial with a 1-arm crossover from leuprolide to degarelix: comparison of gonadotropin-releasing hormone agonist and antagonist effect on prostate cancer. J Urol. 2011;186:889–97.

    Article  CAS  PubMed  Google Scholar 

  58. Huhtaniemi IT, Dahl KD, Rannikko S, Hsueh AJW. Serum bioactive and immunoreactive follicle-stimulating hormone in prostatic cancer patients during gonadotropin-releasing hormone agonist treatment and after orchidectomy. J Clin Endocrinol Metab. 1988;66:308–13.

    Article  CAS  PubMed  Google Scholar 

  59. Miyazawa Y, Kato H, Arai S, Furuya Y, Sekine Y, Nomura M, et al. Clinical endocrinological evaluation of the gonadal axis (testosterone, LH and FSH) in prostate cancer patients switched from a GnRH antagonist to a LHRH agonist. Basic Clin Androl. 2015;25:1–8.

    Article  Google Scholar 

  60. Soulitzis N, Karyotis I, Delakas D, Spandidos DA. Expression analysis of peptide growth factors VEGF, FGF2, TGFB1, EGF and IGF1 in prostate cancer and benign prostatic hyperplasia. Int J Oncol. 2006;29:305–14.

    CAS  PubMed  Google Scholar 

  61. Hoare D, Skinner TA, Black A, Robert Siemens D. Serum follicle-stimulating hormone levels predict time to development of castration-resistant prostate cancer. Can Urol Assoc J. 2015;9:122–7.

    PubMed  PubMed Central  Google Scholar 

  62. MacLean DB, Shi H, Faessel HM, Saad F. Medical castration using the investigational oral GnRH antagonist TAK-385 (relugolix): phase 1 study in healthy males. J Clin Endocrinol Metab. 2015;100:4579–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Zhang G, Wang T, Gao L, Quan D. Oral delivery of oil-based formulation for a novel synthetic cationic peptide of GnRH (gonadotropin-releasing hormone) antagonist for prostate cancer treatment. Int J Pharm. 2013;450:138–44.

    Article  CAS  PubMed  Google Scholar 

  64. Herbst KL, Anawalt BD, Amory JK, Bremner WJ. Acyline: the first study in humans of a potent, new gonadotropin-releasing hormone antagonist. J Clin Endocrinol Metab. 2002;87:3215–20.

    Article  CAS  PubMed  Google Scholar 

  65. Herbst KL, Coviello AD, Page S, Amory JK, Anawalt BD, Bremner WJ. A single dose of the potent gonadotropin-releasing hormone antagonist acyline suppresses gonadotropins and testosterone for 2 weeks in healthy young men. J Clin Endocrinol Metab. 2004;89:5959–65.

    Article  CAS  PubMed  Google Scholar 

  66. Festuccia C, Dondi D, Piccolella M, Locatelli A, Gravina GL, Tombolini V, et al. Ozarelix, a fourth generation GnRH antagonist, induces apoptosis in hormone refractory androgen receptor negative prostate cancer cells modulating expression and activity of death receptors. Prostate. 2010;70:1340–9.

    CAS  PubMed  Google Scholar 

  67. Zhang L, Fang B. Mechanisms of resistance to TRAIL-induced apoptosis in cancer. Cancer Gene Ther. 2005;12:228.

    Article  CAS  PubMed  Google Scholar 

  68. Millar RP, Zhu Y-F, Struthers RS. Progress towards the development of non-peptide orally-active gonadotropin-releasing hormone (GnRH) antagonists: therapeutic implications. Br Med Bull. 2000;56:761–72.

    Article  CAS  PubMed  Google Scholar 

  69. Shaw GL, Whitaker H, Corcoran M, Dunning MJ, Luxton H, Kay J, et al. The early effects of rapid androgen deprivation on human prostate cancer. Eur Urol. 2015. doi:10.1016/j.eururo.2015.10.042.

  70. Stanbrough M, Bubley GJ, Ross K, Golub TR, Rubin MA, Penning TM, et al. Increased expression of genes converting adrenal androgens to testosterone in androgen-independent prostate cancer. Cancer Res. 2006;66:2815–25.

    Article  CAS  PubMed  Google Scholar 

  71. Montgomery RB, Mostaghel EA, Vessella R, Hess DL, Kalhorn TF, Higano CS, et al. Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth. Cancer Res. 2008;68:4447–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Dillard PR, Lin M-F, Khan SA. Androgen-independent prostate cancer cells acquire the complete steroidogenic potential of synthesizing testosterone from cholesterol. Mol Cell Endocrinol. 2008;295:115–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Pfeiffer MJ, Smit FP, Sedelaar JP, Schalken JA. Steroidogenic enzymes and stem cell markers are upregulated during androgen deprivation in prostate cancer. Mol Med. 2011;17:657.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Visakorpi T, Hyytinen E, Koivisto P, Tanner M, Keinänen R, Palmberg C, et al. In vivo amplification of the androgen receptor gene and progression of human prostate cancer. Nat Genet. 1995;9:401–6.

    Article  CAS  PubMed  Google Scholar 

  75. Koivisto P, Kononen J, Palmberg C, Tammela T, Hyytinen E, Isola J, et al. Androgen receptor gene amplification: a possible molecular mechanism for androgen deprivation therapy failure in prostate cancer. Cancer Res. 1997;57:314–9.

    CAS  PubMed  Google Scholar 

  76. Taplin M-E, Balk SP. Androgen receptor: a key molecule in the progression of prostate cancer to hormone independence. J Cell Biochem. 2004;91:483–90.

    Article  CAS  PubMed  Google Scholar 

  77. Zhang A, Zhao Jonathan C, Kim J, K-w Fong, Yang Yeqing A, Chakravarti D, et al. LncRNA HOTAIR enhances the androgen-receptor-mediated transcriptional program and drives castration-resistant prostate cancer. Cell Rep. 2015;13:209–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Chen CD, Welsbie DS, Tran C, Baek SH, Chen R, Vessella R, et al. Molecular determinants of resistance to antiandrogen therapy. Nat Med. 2004;10:33–9.

    Article  PubMed  CAS  Google Scholar 

  79. Waltering KK, Helenius MA, Sahu B, Manni V, Linja MJ, Jänne OA, et al. Increased expression of androgen receptor sensitizes prostate cancer cells to low levels of androgens. Cancer Res. 2009;69:8141–9.

    Article  CAS  PubMed  Google Scholar 

  80. Wang Q, Li W, Zhang Y, Yuan X, Xu K, Yu J, et al. Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell. 2009;138:245–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Korpal M, Korn JM, Gao X, Rakiec DP, Ruddy DA, Doshi S, et al. An F876L mutation in androgen receptor confers genetic and phenotypic resistance to MDV3100 (enzalutamide). Cancer Discov. 2013;3:1030–43.

    Article  CAS  PubMed  Google Scholar 

  82. Joseph JD, Lu N, Qian J, Sensintaffar J, Shao G, Brigham D, et al. A Clinically relevant androgen receptor mutation confers resistance to second-generation antiandrogens enzalutamide and ARN-509. Cancer Discov. 2013;3:1020–9.

    Article  CAS  PubMed  Google Scholar 

  83. Mizokami A, Koh E, Fujita H, Maeda Y, Egawa M, Koshida K, et al. The adrenal androgen androstenediol is present in prostate cancer tissue after androgen deprivation therapy and activates mutated androgen receptor. Cancer Res. 2004;64:765–71.

    Article  CAS  PubMed  Google Scholar 

  84. Taplin M-E, Bubley GJ, Ko Y-J, Small EJ, Upton M, Rajeshkumar B, et al. Selection for androgen receptor mutations in prostate cancers treated with androgen antagonist. Cancer Res. 1999;59:2511–5.

    CAS  PubMed  Google Scholar 

  85. Dehm SM, Tindall DJ. Alternatively spliced androgen receptor variants. Endocr Relat Cancer. 2011;18:R183–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med. 2014;371:1028–38.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Li Y, Chan SC, Brand LJ, Hwang TH, Silverstein KAT, Dehm SM. Androgen receptor splice variants mediate enzalutamide resistance in castration-resistant prostate cancer cell lines. Cancer Res. 2013;73:483–9.

    Article  CAS  PubMed  Google Scholar 

  88. Myung J-K, Banuelos CA, Fernandez JG, Mawji NR, Wang J, Tien AH, et al. An androgen receptor N-terminal domain antagonist for treating prostate cancer. J Clin Invest. 2013;123:2948–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. ClinicalTrials.gov. Safety and anti-tumor study of oral EPI-506 for patients with metastatic castration-resistant prostate cancer. 2015. https://clinicaltrials.gov/ct2/show/NCT02606123. Accessed 20 Dec 2015.

  90. Dalal K, Roshan-Moniri M, Sharma A, Li H, Ban F, Hassona MD, et al. Selectively targeting the DNA-binding domain of the androgen receptor as a prospective therapy for prostate cancer. J Biol Chem. 2014;289:26417–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Mashima T, Okabe S, Seimiya H. Pharmacological targeting of constitutively active truncated androgen receptor by nigericin and suppression of hormone-refractory prostate cancer cell growth. Mol Pharmacol. 2010;78:846–54.

    Article  CAS  PubMed  Google Scholar 

  92. Liu C, Lou W, Zhu Y, Nadiminty N, Schwartz CT, Evans CP, et al. Niclosamide inhibits androgen receptor variants expression and overcomes enzalutamide resistance in castration-resistant prostate cancer. Clin Cancer Res. 2014;20:3198–210.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Vasaitis TS, Bruno RD, Njar VCO. CYP17 inhibitors for prostate cancer therapy. J Steroid Biochem Mol Biol. 2011;125:23–31.

    Article  CAS  PubMed  Google Scholar 

  94. de Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med. 2011;364:1995–2005.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Fizazi K, Scher HI, Molina A, Logothetis CJ, Chi KN, Jones RJ, et al. Abiraterone acetate for treatment of metastatic castration-resistant prostate cancer: final overall survival analysis of the COU-AA-301 randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 2012;13:983–92.

    Article  CAS  PubMed  Google Scholar 

  96. Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med. 2013;368:138–48.

    Article  CAS  PubMed  Google Scholar 

  97. Fleming MT, Morris MJ, Heller G, Scher HI. Post-therapy changes in PSA as an outcome measure in prostate cancer clinical trials. Nat Clin Prac Oncol. 2006;3:658–67.

    Article  CAS  Google Scholar 

  98. Basch E, Autio K, Ryan CJ, Mulders P, Shore N, Kheoh T, et al. Abiraterone acetate plus prednisone versus prednisone alone in chemotherapy-naive men with metastatic castration-resistant prostate cancer: patient-reported outcome results of a randomised phase 3 trial. Lancet Oncol. 2013;14:1193–9.

    Article  CAS  PubMed  Google Scholar 

  99. US Food and Drug Administration. Expanded approval for abiraterone acetate. 2016. http://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm331628.htm. Accessed 18 Jan 2016.

  100. Flaig TW, Potluri RC, Ng Y, Todd MB, Mehra M. Treatment evolution for metastatic castration-resistant prostate cancer with recent introduction of novel agents: retrospective analysis of real-world data. Cancer Med. 2015. n/a-n/a.

  101. Scher HI, Fizazi K, Saad F, Taplin M-E, Sternberg CN, Miller K, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012;367:1187–97.

    Article  CAS  PubMed  Google Scholar 

  102. Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. 2014;371:424–33.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. ClinicalTrials.gov. Enzalutamide versus standard androgen deprivation therapy for the treatment hormone sensitive prostate cancer. 2015. https://clinicaltrials.gov/ct2/show/NCT02278185. Accessed 20 Dec 2015.

  104. Rhoden EL, Morgentaler A. Risks of testosterone-replacement therapy and recommendations for monitoring. N Engl J Med. 2004;350:482–92.

    Article  CAS  PubMed  Google Scholar 

  105. Group EHaPCC, Roddam A, Allen N, Appleby P, Key T. Endogenous sex hormones and prostate cancer: a collaborative analysis of 18 prospective studies. J Natl Cancer Inst. 2008;100:170–83.

    Article  CAS  Google Scholar 

  106. Morgentaler A, Lipshultz LI, Bennett R, Sweeney M, Avila D Jr, Khera M. Testosterone therapy in men with untreated prostate cancer. J Urol. 2011;185:1256–61.

    Article  CAS  PubMed  Google Scholar 

  107. Rhoden EL, Morgentaler A. Testosterone replacement therapy in hypogonadal men at high risk for prostate cancer: results of 1 year of treatment in men with prostatic intraepithelial neoplasia. J Urol. 2003;170:2348–51.

    Article  CAS  PubMed  Google Scholar 

  108. Schweizer MT, Antonarakis ES, Wang H, Ajiboye AS, Spitz A, Cao H, et al. Effect of bipolar androgen therapy for asymptomatic men with castration-resistant prostate cancer: Results from a pilot clinical study. Sci Transl Med. 2015;7:269.

    Google Scholar 

  109. Morris MJ, Huang D, Kelly WK, Slovin SF, Stephenson RD, Eicher C, et al. Phase 1 trial of high-dose exogenous testosterone in patients with castration-resistant metastatic prostate cancer. Eur Urol. 2009;56:237–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Szmulewitz R, Mohile S, Posadas E, Kunnavakkam R, Karrison T, Manchen E, et al. A randomized phase 1 study of testosterone replacement for patients with low-risk castration-resistant prostate cancer. Eur Urol. 2009;56:97–104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Feltquate D, Nordquist L, Eicher C, Morris M, Smaletz O, Slovin S, et al. Rapid androgen cycling as treatment for patients with prostate cancer. Clin Cancer Res. 2006;12:7414–21.

    Article  CAS  PubMed  Google Scholar 

  112. Rathkopf D, Carducci MA, Morris MJ, Slovin SF, Eisenberger MA, Pili R, et al. Phase II trial of docetaxel with rapid androgen cycling for progressive noncastrate prostate cancer. J Clin Oncol. 2008;26:2959–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Klotz LH, Herr HW, Morse MJ, Whitmore WF. Intermittent endocrine therapy for advanced prostate cancer. Cancer. 1986;58:2546–50.

    Article  CAS  PubMed  Google Scholar 

  114. The Pharmaceutical Benefits Scheme. Goserelin. 2016. http://www.pbs.gov.au/medicine/item/1454m-8093y. Accessed 18 Jan 2016.

  115. The Pharmaceutical Benefits Scheme. Triptorelin. 2016. http://www.pbs.gov.au/medicine/item/5297t-9378n-9379p. Accessed 18 Jan 2016.

  116. The Pharmaceutical Benefits Scheme. Goserelin and bicalutamide. 2016. http://www.pbs.gov.au/medicine/item/9064c-9065d-9066e. Accessed 18 Jan 2016.

  117. The Pharmaceutical Benefits Scheme. Leuprorelin. 2016. http://www.pbs.gov.au/medicine/item/10255r-10256t-8707g-8708h-8709j-8859g-8875d-8876e-8877f. Accessed 18 Jan 2016.

  118. The Pharmaceutical Benefits Scheme. Degarelix. 2016. http://www.pbs.gov.au/medicine/item/2784m-2785n. Accessed 18 Jan 2016.

  119. The Pharmaceutical Benefits Scheme. Bicalutamide. 2016. http://www.pbs.gov.au/medicine/item/8094b. Accessed 18 Jan 2016.

  120. The Pharmaceutical Benefits Scheme. Flutamide. 2016. http://www.pbs.gov.au/medicine/item/1417n. Accessed 18 Jan 2016.

  121. The Pharmaceutical Benefits Scheme. Nilutamide. 2016. http://www.pbs.gov.au/medicine/item/8131y. Accessed 18 Jan 2016.

  122. The Pharmaceutical Benefits Scheme. Abiraterone. 2016. http://www.pbs.gov.au/medicine/item/2698b. Accessed 18 Jan 2016.

  123. The Pharmaceutical Benefits Scheme. Enzalutamide. 2016. http://www.pbs.gov.au/medicine/item/10174l. Accessed 18 Jan 2016.

  124. Crook JM, O’Callaghan CJ, Duncan G, Dearnaley DP, Higano CS, Horwitz EM, et al. Intermittent androgen suppression for rising PSA level after radiotherapy. N Engl J Med. 2012;367:895–903.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Hussain M, Tangen CM, Berry DL, Higano CS, Crawford ED, Liu G, et al. Intermittent versus continuous androgen deprivation in prostate cancer. N Engl J Med. 2013;368:1314–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Soloway MS, Schellhammer PF, Smith JA, Chodak GW, Kennealey GT. Bicalutamide in the treatment of advanced prostatic carcinoma: a phase II multicenter trial. Urology. 1996;47:33–7 (discussion 48–53).

    Article  CAS  PubMed  Google Scholar 

  127. Brogden RN, Clissold SP. Flutamide. A preliminary review of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy in advanced prostatic cancer. Drugs. 1989;38:185–203.

    Article  CAS  PubMed  Google Scholar 

  128. US Food and Drug Administration. XTANDI® (enzalutamide): highlights of prescribing information. 2014. http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/203415s003lbl.pdf. Accessed 18 Jan 2016.

Download references

Authors’ contributions

TP conceived of the review, reviewed the literature and drafted the manuscript. MCS, HL, DJR, and MCD revised the intellectual content and contributed to the writing of the manuscript. KR conceived of the review, drafted and finalised the manuscript. All authors read and approved the final manuscript.

Acknowledgements

This work was supported by the Royal Brisbane and Women’s Hospital Research Foundation and Pathology Queensland (T.P, H.L, M.C.D, K.R.); the Movember Foundation; the Prostate Cancer Foundation of Australia through a Movember Revolutionary Team Award (C.M.S.); and the Australian Government Department of Health (C.M.S.).

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

Not applicable.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kerry Richard.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pham, T., Sadowski, M.C., Li, H. et al. Advances in hormonal therapies for hormone naïve and castration-resistant prostate cancers with or without previous chemotherapy. Exp Hematol Oncol 5, 15 (2015). https://doi.org/10.1186/s40164-016-0046-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40164-016-0046-1

Keywords