22
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Supraphysiological Testosterone Therapy as Treatment for Castration-Resistant Prostate Cancer

      discussion

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Blocking androgen signaling has been the focus of treatment for advanced and metastatic prostate cancer (PC) for the past 70 years (1). First-line androgen deprivation therapy (ADT), either through surgical or medical castration (luteinizing hormone-releasing hormone agonists and antiandrogens), holds promise for PC patients; however, the disease inevitably progresses to castration resistance (2). Second-generation ADT, abiraterone acetate (AA), and enzalutamide (ENZ), have been effective for a subset of patients with castration-resistant PC (CRPC) with relatively short median survival benefits (~3–5 months) (3–5). Concerted effort in the field, including evidence from our group, clearly demonstrates a sustained AR activity in the CRPC tumors including (1) amplification of AR, (2) AR mutations, (3) expression of AR splice variants that are constitutively active, (4) altered milieu of AR coactivators and corepressors, and (5) intracrine synthesis of androgens to support CRPC progression (6, 7). The addiction of PC to the AR signaling paradoxically creates a therapeutic vulnerability that has recently attracted increasing attention. While ADT causes regression of PC, high level of androgen can also inhibit PC progression. The concept of cancer suppression using excessive hormone therapy was introduced by earlier work from Huggins in 1940: “malignant cells can regress from too little or too much hormone” (8). In relation to PC, AR regulates proliferation as well as differentiation of prostate epithelial and cancer cells but it has not been established what conditions support one over the other. Interestingly activation of AR with excessive hormone (i.e., supraphysiological levels of testosterone; SPT) was shown to inhibit growth of CRPC in vitro by negative effects on proliferation and increased expression of some of the AR-regulated genes that are expressed in differentiated luminal epithelium, e.g., prostate-specific antigen. Multiple preclinical studies demonstrated that SPT inhibits growth of PC cells that express AR (9–21), with evidence suggesting that higher levels of AR might lead to more pronounced SPT effects in certain phenotypes of CRPC [reviewed in Ref. (22)]. However, AR by itself is not necessarily sufficient for the SPT-induced growth inhibition; cellular context (23) and AR-regulated transcriptome in its entirety will need to be assessed to delineate the molecular effect of SPT (24). Mechanistically, SPT-induced cell growth inhibition involves (1) cell-cycle arrest, (2) disruption of AR-mediated DNA licensing, (3) DNA damage, (4) transcriptional repression of AR and its variants, (5) transcriptional reprogramming, (6) cellular quiescence or senescence, and (7) induction of apoptosis [reviewed in Ref. (22)]. However, these effects were demonstrated exclusively in cell line models, and whether they play a significant biological role in SPT-induced tumor inhibition in patients remains to be determined. Clinical use of testosterone (T) supplementation in PC has been limited and provided controversial results. Two older National Prostatic Cancer Project trials that used T-supplementation to normal levels with a goal to enhance the effectiveness of chemotherapy reported disappointing results (25, 26). Additional two phase I trials, which did not achieve consistent supraphysiological T levels, showed minimally reduced disease progression (27, 28). In contrast, several other studies showed that T-supplementation to normal-supraphysiological range (303−2637 ng/dl), specifically in symptomatic hypogonadal PC patients, provided prolonged disease control (as measured by sustainably low-PSA level) (29–31). In our opinion, the lack of favorable response in some of the clinical trials is, at least in part, due to the absence of a supraphysiological level of T as well as the unselected patient population. With advanced understanding of the biology and AR involvement in CRPC progression, leveraging the active AR signaling to explore therapeutic opportunity has recently received renewed attention in clinical settings. Dr. Denmeade’s group at John Hopkins University pioneered a therapy termed “bipolar androgen therapy” (BAT) as a treatment for PC patients. With BAT treatment, PC patients receive intermittent T injections at doses shown to produce a spike in serum T to supraphysiological levels, followed by a decline to below normal at the end of a 28-day treatment cycle (32). This cycling strategy was developed based on the most common molecular hallmark of CRPC–overexpression of AR (33) and the potential growth inhibitory effect of SPT in AR-overexpressing PC. Rapidly cycling of T from SPT (~1,500 ng/dl) to below normal T levels (~150 ng/dl) was expected to blunt the adaptive changes in AR expression, thereby delaying the emergence of resistance. In these proof-of-principle BAT trials, one in CRPC showed radiographic response rates of ~50% in men (32), and one in hormone-sensitive PC showed favorable PSA responses (34). Promising results of these trials led to a new clinical trial, in which asymptomatic CRPC patients that progressed on AA or ENZ receive BAT, and after progression on BAT the patients are re-challenged with AA or ENZ. This trial aims to evaluate the efficacy of BAT in patients who progressed on secondary ADT and assess whether BAT re-sensitizes CRPC to secondary ADT. Recent data from this trial showed a PSA50 response in 9/30 ENZ-resistant patients on BAT, and, importantly, 15/21 patients who progressed on BAT showed a PSA50 response upon ENZ re-challenge (35). These results are encouraging. However, additional analyses and larger number of patients are needed to correlate tumor/radiographic vs. PSA responses in individual patients. One of the reasons is that PSA changes do not necessarily associate with tumor regression in advanced CRPC. PSA, an AR-regulated gene, is highly sensitive to AR activation/inhibition and can rise upon SPT and decline upon ADT. PSA response might not faithfully reflect radiographic responses in advanced CRPC which growth often does not rely solely on AR signaling [e.g., FGF signaling (36)]. Bipolar androgen therapy shows great clinical promise in a subset of patients. However, universal to all cancer treatment modalities, not all patients respond to this treatment and resistance to BAT develops. Therefore, there is an opportunity to improve this therapy. It is notable that a critical step in drug development, determining the optimal dosing schedule, was bypassed in the clinical development of BAT. Despite the clinical efficacy of BAT, there were by far no clinical data to support the hypotheses that cycling SPT (i.e., BAT) mitigates the development of resistance or that BAT represents the optimal mode for administering SPT. Notably, several preclinical studies have consistently demonstrated that SPT delivered on a continuous basis inhibits the growth of PC cells (13, 20, 21). While several small clinical trials of continuous T administration in men with CRPC have been carried out, they did not achieve SPT levels. Cycling or not cycling—that is the question. While we currently do not have sufficient evidence whether BAT results in better clinical outcome than continuous SPT, it is possible that long-term continuous SPT and BAT could alter AR signaling differently. One would anticipate that continuous SPT might trigger more pronounced differentiation, potentially causing a change from a “low-T” oncogenic AR transcriptome to that of a more differentiating SPT transcriptome (24). Meanwhile, BAT might provide better efficacy if cell-cycle relicensing effects and DNA damage are the critical mechanism of action (37, 38). While BAT was associated with improved quality of life (34, 35), this effect diminished over the course of a cycle of BAT, presumably due to T levels falling below normal range. It is possible that quality of life metrics will be better with continuous SPT but there also might be increase in negative side effects. While T therapy has been reported to be generally safe, with a small subset of patients experiencing severe cardiovascular-related complications (27–32, 39–42), continuous SPT has not been tested and monitoring will be essential. Careful evaluation of effects of BAT vs. continuous SPT on tumor progression, as well as any potential health benefits or side effects will be required to make final decision. Interestingly, cycling of ADT, intermittent ADT, has been evaluated in PC extensively since its introduction in mid-1980s. However, intermittent ADT was not found to be inferior to continuous ADT with respect to the overall survival but it was shown to improve patients’ quality of life, and therefore it is thought to be a viable option for patients who experience significant adverse effects of continuous ADT [for review see Ref. (43, 44)]. In addition, intermittent AA therapy has been recently shown to delay the development of resistance from 16.5 (continuous treatment) to 27 months (45). Whether the intermittent therapy diversifies the residual tumor clones or re-sensitize the residual clones to a therapy that formerly failed remains scientifically and clinically important. In summary, we will need to seek answers to multiple important questions before unleashing the full potential of SPT therapy in CRPC: (1) which mode of SPT, BAT or continuous SPT, represents the optimal administration regimen for tumor growth inhibition; (2) what population of patients will benefit from SPT therapy; (3) is there a way to prolong the treatment response; and (4) what are the mechanisms of resistance, as these will be diverse in different tumor phenotypes. To address these questions, systematic preclinical trials will need to be performed, and pre-treatment and on-treatment clinical specimens will be essential to identify mechanisms of SPT action and biomarkers that predict SPT response. Author Contributions H-ML and EC jointly conceptualized and wrote the manuscript. Conflict of Interest Statement The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

          Related collections

          Most cited references41

          • Record: found
          • Abstract: found
          • Article: not found

          Androgen Receptor Pathway-Independent Prostate Cancer Is Sustained through FGF Signaling.

          Androgen receptor (AR) signaling is a distinctive feature of prostate carcinoma (PC) and represents the major therapeutic target for treating metastatic prostate cancer (mPC). Though highly effective, AR antagonism can produce tumors that bypass a functional requirement for AR, often through neuroendocrine (NE) transdifferentiation. Through the molecular assessment of mPCs over two decades, we find a phenotypic shift has occurred in mPC with the emergence of an AR-null NE-null phenotype. These "double-negative" PCs are notable for elevated FGF and MAPK pathway activity, which can bypass AR dependence. Pharmacological inhibitors of MAPK or FGFR repressed the growth of double-negative PCs in vitro and in vivo. Our results indicate that FGF/MAPK blockade may be particularly efficacious against mPCs with an AR-null phenotype.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Integrating evolutionary dynamics into treatment of metastatic castrate-resistant prostate cancer

            Abiraterone treats metastatic castrate-resistant prostate cancer by inhibiting CYP17A, an enzyme for testosterone auto-production. With standard dosing, evolution of resistance with treatment failure (radiographic progression) occurs at a median of ~16.5 months. We hypothesize time to progression (TTP) could be increased by integrating evolutionary dynamics into therapy. We developed an evolutionary game theory model using Lotka–Volterra equations with three competing cancer “species”: androgen dependent, androgen producing, and androgen independent. Simulations with standard abiraterone dosing demonstrate strong selection for androgen-independent cells and rapid treatment failure. Adaptive therapy, using patient-specific tumor dynamics to inform on/off treatment cycles, suppresses proliferation of androgen-independent cells and lowers cumulative drug dose. In a pilot clinical trial, 10 of 11 patients maintained stable oscillations of tumor burdens; median TTP is at least 27 months with reduced cumulative drug use of 47% of standard dosing. The outcomes show significant improvement over published studies and a contemporaneous population.
              Bookmark
              • Record: found
              • Abstract: not found
              • Article: not found

              Mechanisms underlying the development of androgen-independent prostate cancer.

                Bookmark

                Author and article information

                Contributors
                Journal
                Front Oncol
                Front Oncol
                Front. Oncol.
                Frontiers in Oncology
                Frontiers Media S.A.
                2234-943X
                22 May 2018
                2018
                : 8
                : 167
                Affiliations
                Department of Urology, University of Washington School of Medicine , Seattle, WA, United States
                Author notes

                Edited by: Fabio Grizzi, Humanitas Research Hospital, Italy

                Reviewed by: Momoe Itsumi, Tokyo Medical and Dental University, Japan; Benyi Li, University of Kansas Medical Center, United States

                *Correspondence: Eva Corey, ecorey@ 123456uw.edu

                Specialty section: This article was submitted to Genitourinary Oncology, a section of the journal Frontiers in Oncology

                Article
                10.3389/fonc.2018.00167
                5972313
                29872642
                f9c4707b-2f00-4699-a2d4-62753d271a4c
                Copyright © 2018 Lam and Corey.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 10 January 2018
                : 01 May 2018
                Page count
                Figures: 0, Tables: 0, Equations: 0, References: 45, Pages: 4, Words: 3166
                Funding
                Funded by: National Institutes of Health 10.13039/100000002
                Award ID: R21 CA194798, P50 CA097186, P01 CA163227, P30 CA015704
                Categories
                Oncology
                Opinion

                Oncology & Radiotherapy
                supraphysiological testosterone,bipolar androgen therapy,castration-resistant prostate cancer,enzalutamide,abiraterone

                Comments

                Comment on this article