5-alpha reductase inhibitors use in prostatic disease and beyond
Review Article

5-alpha reductase inhibitors use in prostatic disease and beyond

Bodie Chislett1,2, David Chen1,2, Marlon L. Perera1,3, Eric Chung4, Damien Bolton1, Liang G. Qu1,2

1Department of Urology, Austin Health, Heidelberg, Melbourne, VIC, Australia; 2Young Urology Researchers Organisation (YURO), Melbourne, VIC, Australia; 3Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; 4Department of Urology, Princess Alexandra Hospital, University of Queensland, Brisbane, QLD, Australia

Contributions: (I) Conception and design: B Chislett, LG Qu; (II) Administrative support: B Chislett, LG Qu; (III) Provision of study materials or patients: B Chislett; (IV) Collection and assembly of data: B Chislett, D Chen; (V) Data analysis and interpretation: B Chislett, D Chen; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Bodie Chislett. 41 Gouldburn Street, Yarraville, VIC 3013, Australia. Email: bodiechislett@gmail.com.

Abstract: 5-alpha reductase inhibitors (5-ARIs) are commonly used and widely available, with benefits observed from their effect on androgen signalling. Their effect relies on the inhibition of the 5-alpha reductase enzyme which aids in the conversion of testosterone to dihydrotestosterone. 5-ARIs have increasing clinical relevance outside of benign prostatic hyperplasia (BPH). Such development requires clinicians to have an updated review to guide clinical practices. This review details the pharmacology and mechanisms of action for 5-ARIs and how this relates to multiple clinical indications. Of note, is the debunked association between finasteride and increased risk of high-grade prostate cancer. Furthermore, adverse effects of 5-ARI use are detailed in this review, with specific mentions to post-finasteride syndrome. In addition to overviews pertaining to BPH and prostate cancer, much attention has also been focused on severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The androgen axis may be associated with an increased virulence for SARS-CoV-2 in men, with some reporting a correlation between the severity of illness and androgenic alopecia. Since these observations, the role of antiandrogens, including 5-ARIs, has been explored further in SARS-CoV-2. Increasing understanding of pathological processes involving the androgen axis in which 5-ARIs work, has led to increasing clinical indications for 5-ARIs. Several novel off-label indications have been suggested including its potential role in the pathogenesis of SARS-CoV-2, but to date, these claims have not been substantiated. Previously held truths regarding the role of 5-ARIs and prostate carcinogenesis have been contested, inadvertently leading to the re-exploration of 5-ARIs utility in prostate cancer. With growing evidence into pathological processes involving the androgen axis, 5-ARIs are likely to become increasingly more used. This review serves as a timely update of 5 ARIs pharmacology, current indications and potential future directions.

Keywords: 5-alpha reductase inhibitors (5-ARIs); review; prostate cancer; severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2); coronavirus disease (COVID)


Submitted Oct 18, 2022. Accepted for publication Feb 21, 2023. Published online Mar 06, 2023.

doi: 10.21037/tau-22-690


Introduction

5-alpha reductase inhibitors (5-ARIs) are commonly used and widely available, with benefits observed from their effect on androgen signalling. Initially licenced for use in benign prostatic hyperplasia (BPH), finasteride and dutasteride are the two major 5-ARIs that are commercially available and in use today (1,2). Their effect relies on the inhibition of the 5-alpha reductase (5α-reductase) enzyme which aids in the conversion of testosterone to dihydrotestosterone (DHT) (3).

Since its introduction, the clinical indication for 5-ARIs has grown from BPH to include androgenetic alopecia (AGA) and hirsutism (1). As more pathological processes are identified as being mediated by DHT, the clinical indications and utility of 5-ARIs expand. The relationship between 5-ARIs and prostate cancer is of particular interest due to the androgen-driven nature of prostate carcinogenesis (3,4). Other new associations have been observed between 5-ARIs and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), including with reduced disease severity and mortality (5,6).

With new developments and increasing clinical indications, an up-to-date review of 5-ARIs is warranted, helping to inform clinical practice. This narrative review addresses the mechanism of action of 5-ARIs, potential adverse effects, current clinical indications including indications within and outside of urology.


Pharmacology

Finasteride was first licenced by the United States Food and Drug Administration (US FDA) in 1992 (1,7). It competitively inhibits the type II 5α-reductase enzymes isoenzyme, suppressing serum DHT by approximately 70% from baseline (3). Dutasteride, the first dual 5α-reductase type I and type II isoenzyme inhibitor, was approved for use a decade later and is estimated to lower serum DHT by approximately 90% (8).

5α-reductase is a nuclear-bound steroid intracellular enzyme that converts the androgen testosterone into DHT. 5-ARIs are chemically 4-azasteroids that competitively bind 5α-reductase intracellularly, primarily in the prostate stroma, leading to the peripheral inhibition of testosterone conversion into its more potent metabolite. DHT is observed as having a greater affinity than testosterone to androgen receptors (AR) in the prostate gland. A hormonal mediator, DHT modulates genes responsible for cell proliferation and can lead to hyperplasia.

5α-reductase exists as two isoenzymes, type I and type II, primarily found within the prostate stroma. Type II isoenzymes are estimated to account for two-thirds of serum DHT production and can be found outside prostate tissue, including seminal vesicles, epididymides, hair follicles, and the liver (1). Finasteride selectively inhibits type II isoenzyme and reduces serum DHT by 65% within the first 24 hours (1). Dutasteride inhibits type I and type II, and was observed to reduce serum DHT by 90% after two weeks of regular dosing (2). DHT suppression through inhibition of 5α-reductase leads to an increase of serum testosterone of 15% and 19% for finasteride and dutasteride respectfully (1,2).

5-ARIs are absorbed well orally, with a mean bioavailability of approximately 60–65% from a 1 mg dosage (1,2). With 90% of the circulating drug being protein-bound, 5-ARIs are predominantly metabolised hepatically by the cytochrome P450 enzyme. It is estimated that between 32–46% of the drug is excreted in the urine, with the remaining 51–64% excreted in faeces. The half-life of 5-ARI is approximately 6 hours in young, healthy males, increasing to 8 hours in septuagenarians (1,2).


Adverse effects

Both finasteride and dutasteride are generally well-tolerated medications, with most adverse effects reported as mild and transient (2). The most frequently reported adverse effects are related to sexual dysfunction, mainly decreased sexual libido, impotence, and ejaculatory disorders. Increasing post-market reports of sexual and depressive side-effects prompted many countries to change labelling to include these adverse side effects. Despite a growing body of literature to support the existence of adverse sexual side effects, the validity of a causal relationship between 5-ARIs and all its reported adverse effects remains unclear, with only observation studies available (9-13). The Proscar Long-Term Efficacy and Safety Study (PLESS), reported a marginal effect with 3.7% of patients treated with finasteride ceasing the medication due to adverse sexual side effects, compared to 2.1% of patients taking placebo (14). A more substantial effect was observed in a meta-analysis by Liu et al., where men taking 5-ARIs for BPH had 156% increased risk of experiencing sexual side effects (9). This study also observed dutasteride as having a greater incidence of adverse sexual side-effects than finasteride. Most literature suggests these symptoms tend to resolve within 1 month of ceasing the medication.

Several other adverse effects have been described, including depression, cardiovascular disease, and metabolic syndrome. Suggestions of an increase in the incidence of male breast cancer and dementia have not been supported by current literature (15,16). The association of 5-ARIs and depressive symptoms was first highlighted in the Prostate Cancer Prevention Trial (PCPT) (4). The PCPT however was not designed or powered to assess an association between depressive symptoms and 5-ARIs, failing to establish a causal relationship. Other studies attempting to correlate depressive symptoms and 5-ARIs have similar flaws, highlighted in a comprehensive clinical review by Saengmearnuparp et al. (17). It is reported 5-ARIs may increase insulin resistance, leading to high rates of diabetes, metabolic disease, and cardiovascular disease (18). While only observation human data exists, these claims were supported by the pre-clinical findings in rodents by Livingstone et al., where the absence of type I 5α-reductase isoenzyme was associated with hepatic steatosis and insulin resistance (19). This is in contrast to a previous publication by Hsieh et al., where they report no association between 5-ARIs and cardiovascular events during a follow-up over 1,400 men over 5 years (20). This data however failed to examine markers of metabolic disease. Longer follow-up may be required. Currently, there remains no well-established relationship between 5-ARIs and metabolic syndrome or cardiovascular disease.

A growing chorus of consumers have reported persistent adverse side effects from 5-ARIs, despite its discontinuation of the medication. These persistent symptoms have been labelled post-finasteride syndrome (PFS), embodying a constellation of sexual, physical, and neuropsychiatric symptoms that developed during or after the use of 5-ARIs. Consequently, several countries, including the US FDA, have amended warnings to include the possibility of persistent side effects (21). The pathological mechanism of PFS is unclear, though it is theorised 5-ARIs inhibit the synthesis of neurosteroids known to affect mood, cognition and libido (17). Current literature is conflicting regarding the existence of PFS, along with the dosing and duration required to be at risk. The PLESS study showed 15% of patients taking finasteride experienced sexual side effects within the first 12 months, compared to 7% in the placebo group. During the remaining 3 years of follow-up, there was no difference in reported sexual side effects between groups (14). However, only 50% of people taking finasteride reported resolution of their adverse sexual side effects following discontinuation, compared to 41% in the placebo group. Recent meta-analysis on adverse side effects of 5-ARIs did not assess persistent symptoms. Current evidence in support of PFS consists of case reports, surveys or low volume observation data. It is, for this reason, that the existence, incidence and mechanism of PFS remain debated (13,21,22). To establish the existence of PFS, quality prospective data, including placebo trials, is required.


Benign prostatic hyperplasia

BPH prevalence and associated lower urinary tract symptoms (LUTS) increases with age, affecting 80% of men aged 70 and above. Like other sex-accessory organs, the prostate is responsive to hormones and growth factors secreted for maintenance. DHT mediates intracellular apoptosis and proliferation through AR, having the highest affinity and influence on prostatic tissue. Unlike prostate cancer, there are no alterations of AR expression in BPH.

5-ARIs inhibit DHT production and are indicated for the management of enlarged prostates >30 cc on imaging, a prostate-specific antigen level of >1.5 ng/dL or palpable prostatic enlargement on a direct rectal exam (23). Similarly, they are indicated to prevent LUTS progression and reduce the risk of acute urinary retention (AUR) or prostatic surgery. 5-ARIs can be used in the medical management of BPH as either monotherapy or in combination with alpha-blockers (24). The Medical Treatment of Prostate Symptoms (MTOPS) trial highlighted finasteride as having a 34% risk reduction in participants experiencing an American Urological Association symptom score increase of greater than 4 (P=0.002), and a 66% risk reduction for combination therapy (25). In a study across 36 months by Boyle et al., finasteride reduced prostate volume by 27% compared to baseline and was found to improve urinary flow (Qmax) by 2.3 mL/s along with an increase of 3.6 points when the international prostate symptoms score (IPSS) was measured. The authors also observed that men with enlarged baseline prostates benefited the most from finasteride use with significant improvements in IPSS and Qmax metrics (26). In the PLESS, a double-blinded, placebo-controlled trial, men with symptomatic LUTS and enlarged prostates experienced improvements in symptoms (27). Finasteride reduced prostate volume by 18% compared to a 14% increase in the place group. Similarly, dutasteride has comparable results with a 57% and 55% risk reduction for AUR and BPH-related surgical intervention (28).

Patients and clinicians should expect maximal therapeutic efficacy in relieving LUTS approximately 6 months after commencing 5-ARI therapy. In the long-term, the MTOPS and Combination of dutasteride (Avodart) and Tamsulosin study (CombAT) 5-ARI monotherapy arms demonstrate 30% decrease in IPSS and 20% urinary flow at 4-year mark (14,25,29). As 5-ARI acts upon hormonal pathways, patients with larger baseline prostatic volumes experience the most clinical improvement. Patients with an initial prostatic volume of above 40 mL experience the most significant LUTS and urinary flow improvements (26). The ComBAT study findings cannot be extrapolated to smaller initial prostatic volumes of below 30 mL as it was not within the inclusion criteria.

The use of combination therapy is suggested for moderate to severe LUTS by the American Urological Association (23). CombAT was a 4-year multicentre, randomised, double-blinded parallel-group study of 4,844 men ≥50 years of age with a clinical diagnosis of BPH, IPSS ≥12, and significantly enlarged prostates. The trial demonstrated that combination therapy of dutasteride and tamsulosin was significantly superior to tamsulosin monotherapy alone but not dutasteride monotherapy at reducing the relative risk (RR) of AUR or BPH related surgery occurrence (14). CombAT also reports that combination therapy provides greater benefit in superior results in disease progression compared to both monotherapies.

There are increasing surgical modalities to manage BPH; however, transurethral resection of the prostate (TURP) remains the historical gold standard. Patients progress to surgical management after unsuccessful combination therapy. Surgical management is recommended to patients with refractory LUTS, recurrent urinary tract infections, recurrent bladder stones or gross haematuria with bladder outlet obstruction, persistent urinary retention secondary to BPH or high-pressure retention (23,30). Prospective 5-ARI administration before TURP has been indicated to reduce the level of surgical bleeding (31). There are also associated decreases in transfusion rates and operating times. However, more established evidence through trials required to confirm these findings (32).

Beyond preoperative administration of 5-ARIs in reducing surgical bleeding, 5-ARIs have a role in reducing bleeding secondary to BPH-related gross haematuria. Local prostate angiogenesis is stimulated by vascular endothelial growth factors (VEGFs), a potent signalling protein that is up-titrated by androgens including DHT (33,34). Kearney et al. demonstrated finasteride as a suitable agent for improving gross haematuria secondary to BPH regardless of anticoagulation status through the suppression of DHT (35). In their retrospective review of 53 patients given daily 5 mg finasteride for BPH-related haematuria, 94% of patients experienced improving haematuria grading and 77% no longer experienced haematuria while taking finasteride. The study also appeared to identify a time-dependent relationship between prostate size and the duration required for finasteride to improve gross haematuria.

In vitro studies demonstrated finasteride interferes with angiogenesis and causes tissue hypoxia (36,37). In a study by Lekas et al., 178 patients undergoing TURP were either allocated to finasteride or no medication prior to their operation (38). Blood loss was significantly elevated in the control group which received no medication. To assess for the impact of finasteride on BPH angiogenesis and tissue hypoxia, micro vessel density (MVD), VEGF, and hypoxia-inducible factor-1alpha (HIF-1α) were identified in vivo post-TURP. A statistically significant decrease in above factors was found in the tissue of patients on finasteride compared to those who were untreated.


Prostate cancer

The binding of androgen to AR leads to cell signalling resulting in prostate cell proliferation (39,40). By regulating prostate cell apoptosis and proliferation, androgens play a pivotal role in prostate cancer. The discovery and success of androgen deprivation therapy in treating prostate cancer caused the androgen-androgen receptor axis to be a prime target for new prostate cancer treatments. In the past, 5-ARIs have been suggested as potentially appealing prostate cancer chemopreventive agents, by inhibiting the conversion to the more potent DHT. The PCPT in the early 2000s demonstrated that 5-ARIs (finasteride), compared with placebo, had a 24.8% RR reduction in developing prostate cancer (4). However, their benefit was largely observed in low-risk disease and concerningly, a significant increase in proportion of high-grade prostate cancer was observed in men taking finasteride. Thompson et al. observed 37% of the graded prostate cancers to be Gleason score 7–10, compared to 22% of those in the placebo group, or a RR of 1.67 [95% confidence interval (CI): 1.44–1.93] (low-grade disease = Gleason ≤6; intermediate-grade disease = Gleason 7; high-grade disease = Gleason ≥8) (4). Given limited benefit in reducing significant disease and associated morbidity, 5-ARIs were never adopted or indicated as a chemopreventive agent for prostate cancer.

Since these initial findings, further research has been conducted to elucidate the relationship between 5-ARIs and prostate cancer. Findings have been mixed, though many studies have reported no discernible relationship between 5-ARIs and the incidence of high-grade prostate cancer (41-44). More recently, several notable meta-analyses have been conducted, all of which contend that 5-ARIs do not increase the incidence or risk of high-grade prostate cancer (45,46). Knijnik et al. examined the relationship between 5-ARIs and risk-stratified prostate cancer subgroups, meta-analysing 18 studies including ten randomised controlled trials (46). This meta-analysis observed no statistically significant difference between high-grade prostate cancer diagnoses and 5-ARI use (RR 0.98, 95% CI: 0.67–1.43, I2=75%). Furthermore, they observed no statistically significant difference in prostate cancer-specific mortality rates between the 5-ARI exposure arm and the control arm, a finding supported by three other meta-analyses, along with multiple other publications (42,43,45-51).

With multiple studies refuting the association of 5-ARIs and higher-grade prostate cancer, some have sought to explain the initial findings from the PCPT trial. Most theorise a sampling bias or detection bias has occurred in men using 5-ARIs, through 5-ARIs inhibiting indolent or low-grade tumours, falsely creating a higher proportion of more aggressive prostate cancers (52,53). Others suggest improved screening, with small prostate sizes from 5-ARIs leading to more accurate prostate-specific antigen testing, digital rectal examinations, and fewer sampling errors (54,55). Alternatively, the histological changes noted might mimic prostate cancer, an effect often seen with androgen deprivation therapy (53,56). Regardless, strong evidence suggests 5-ARIs have no significant impact on the incidence or mortality of higher-grade prostate cancers.

The potential for 5-ARIs to be used as chemopreventive medications should be re-examined. Multiple meta-analyses have confirmed a relationship between 5-ARIs and prostate cancer, with both finasteride and dutasteride reducing the incidence of overall and lower-grade prostate cancers (45,46,49,50). Knijnik et al. observed a RR reduction of 24% for low and intermediate-risk prostate cancer cohorts [RR 0.76, 95% CI: 0.59–0.98, I2=0.74%], a similar finding to the PCPT observations (4,46). Another study reported a RR reduction of 16% in all prostate cancers (RR 0.84, 95% CI: 0.74–0.94) and 27% in low-grade prostate cancers (RR 0.73, 95% CI: 0.60–0.88) (49). Recent publications have even suggested 5-ARIs have a potential benefit in preventing the clinical and pathological progression of prostate cancer, as is observed by the meta-analysis by Yang et al. (47).

With the potential for increased use of 5-ARIs in men with prostate cancer, clinicians must remain cognisant of their effect on prostate specific antigen (PSA) screening and surveillance. PSA is released by the epithelial cells of the prostate to aid the mobilisation of semen. In prostatic diseases such as BPH and prostate cancer, elevated PSA levels are a consequence of epithelial disruption leading to increased stromal and serum uptake (57). 5-ARIs inadvertently suppress serum PSA values within 6 months of continuous use, while poorly understood it is thought by preventing the production of DHT they limit new growth and reduce epithelial disruption (58-60). The American Urological Association suggested expecting approximately a 50% decrease in serum PSA among men using 5-ARIs (4,61). These guidelines are in line with a 2021 systematic review and meta-analysis by Sakalis et al., observing a comparable reduction in PSA when compared to dutasteride (62). However, Sakalis et al. observed finasteride reduced PSA levels by −0.62 [95% CI: −0.71 to 0.56], suggesting an underestimation when compared to conventional teaching. These findings are yet to influence current practice, where PSA values among men using 5-ARIs are routinely doubled before their application into various screening or surveillance protocols.

With substantial evidence refuting the association of 5-ARIs and high-grade prostate cancer, and proven benefits in reducing the overall incidence of prostate cancer, the lack of clinical indication for 5-ARIs as a chemopreventive agent in prostate cancer should be re-examined. More prospective data is warranted prior to further consideration of this application.


SARS-CoV-2

Despite comparable infection rates between men and women, men have been disproportionately affected by the SARS-CoV-2 with increased severity of illness and higher mortality rates (63). The pathological and immunological process leading to this discrepancy is not well elucidated. However, much attention has been focused on the androgen axis as the cause for this increased virulence in men, with some reporting a correlation between the severity of illness and androgenic alopecia (64). Since these observations, many have investigated the role antiandrogens may play, including 5-ARIs, in SARS-CoV-2.

Several publications have identified a protective benefit from antiandrogens in men with SARS-CoV-2. Potential benefits include lesser symptoms, reduced disease severity and fewer intensive care unit (ICU) admissions (5,6,65,66). These claims have been refuted in a recent meta-analysis, where there was no evidence for a protective benefit of androgen deprivation therapy from SARS-CoV-2 (67).

While the majority of research has dealt with androgen deprivation therapy, several studies have assessed the impact of antiandrogen therapy on SARS-CoV-2, with three directly addressing the potential impact of 5-ARIs (6,66,68). Most notably, Cadegiani et al. reported that men taking dutasteride had a shorter duration of illness, along with higher virologic and clinical remission rates on day seven of their illness when compared to a placebo group in their double-blinded randomised controlled trial. Participants were seen in an outpatient setting having confirmed with SARS-CoV-2 on PCR testing and subsequently randomised into either dutasteride (n=44) or placebo (n=43) arms of the trial where they were treated for 30 days or until complete resolution of their symptoms. Here they reported a higher virologic remission rate (64.3% versus 11.8%; P=0.0094) and higher clinical recovery rate (84.7% versus 57.5%; P=0.03) in the men taking dutasteride. A matched paired analysis of 944 participants, had similar findings where the absolute risk of infection on a community acquired SARS-CoV-2 infection was significantly lower in men taking 5-ARIs. They reported a RR for men taking 5-ARIs as 42.3% (399/944), compared to the paired group at 47.2% (446/944). This equated to an absolute risk reduction of 4.9% [odds ratio (OR) 0.81, 95% CI: 0.67–0.97, P=0.026]. Similarly, 77 men were followed during a prospective cohort study, where a lower proportion of ICU admissions was observed among men taking anti-androgens including 5-ARIs, [1/12 (8%) compared to 38/65 (58%), P=0.0015]. The study however reported a number of limitations, including small sample size and use of multiple anti-androgen medications with variable mechanisms of actions. These results are however encouraging, with the further investigation needed to establish the utility of 5-ARIs and their influence in the management of SARS-CoV-2.

While the exact biologic interplay between androgens and SARS-CoV-2 is unknown, it is hypothesised that SARS-CoV-2 binds with the angiotensin-converting enzyme 2 (ACE2) and enters pulmonary tissues via its spike protein after activation by transmembrane protease serine 2 (TMPRSS2) (69). Both proteins are androgen-regulated, though TMPRSS2 is particularly sensitive, with increased expression in malignant prostatic tissue (70). Higher androgen levels lead to upregulation of TMPRSS2 and theoretically increase the risk of cell-virus fusion. This association between androgens and SARS-CoV-2 is supported by the rates of hospital admissions for prepubescents and men with AGA, with lower and higher rates respectfully (71,72).

Pro-immune effects associated with oestrogen is an alternative premise (66,73). It is postulated that by inhibiting the enzymatic conversion of testosterone to DHT, the overall concentration of serum androstenedione produces a physiological shift to produce greater amounts of oestrogen. Oestrogen is known to have a pro-immune effect, in contrast to testosterones immunosuppressive effect (74).

Further study is required for 5-ARIs and their influence on pathological processes for SARS-CoV-2. Currently, the role of androgens in mediating TMPRSS2 in lung parenchyma is conflicting (75,76). Further, the pro-immune effects of elevated oestrogen have been refuted by the claim that any associated effects of increased oestrogen concentrations directly compete with the immunosuppressive effect of elevated testosterone (66).

Unlike testosterone and oestrogen, the TMPRSS2 expression theory and elevated oestrogen theory are not competitive. It is therefore possible that both proposed mechanisms of action work synergistically. Nonetheless, presently 5-ARIs are not indicated for use in preventing or managing SARS-CoV-2 due to limited prospective data.


Other non-urological indications for 5-ARIs

AGA is a form of hair loss experienced by both men and women, commonly referred to as ‘pattern baldness’, with each sex experiencing distinct distributions of hair loss. The pathological process involves hair follicles undergoing miniaturisation, referring to the production of more delicate hairs that readily fall out due to their fragile shafts (77). While the molecular process of AGA is a mystery, it is known to be mediated by DHT, evidenced by its absence in men with a 5α-reductase type II deficiency (78). As previously highlighted, two isoenzymes exist, type I located in the liver and skin, including the scalp, and type II located in hair follicles and the prostate. Commercially sold as ‘Propecia’, finasteride was first approved by the FDA in 1997 to treat AGA (1). Oral administration of finasteride has robust literature to support its use. The use of topical finasteride avoids the unwanted systemic side-effects of oral 5-ARI use and appears to be comparable to oral finasteride, though more quality long-term studies are needed (78). Dutasteride is also indicated for the treatment of AGA with some suggesting its superiority, though limited data exists comparing it to finasteride (77,79).

5-ARIs are increasingly used in hirsutism, a condition where predominantly women present with excessive hair growth (80). Excess circulating androgens stimulates terminal hair growth over androgen dependent areas. The excess hair growth is often course, like sexual or secondary hair such as those seen in male growth patterns (80,81). While often presenting as primary idiopathic Hirsutism, numerous aetiologies can lead to excess androgen production and therefore the development of hirsutism, including polycystic ovarian syndrome, acromegaly, prolactinemia or thyroid disfunction (82).

There has been increasing preclinical studies examining the benefits of 5-ARIs on reducing excessive alcohol intake in men. It is hypothesised that endogenous neuroactive steroidal properties of 5-ARIs may mediate sedative effects like alcohol in adult men (83). This is based on weak evidence presuming that 5-ARIs may reduce drinking due to its impacts on neuroactive steroids concentrations.

The role of 5-ARI for stuttering priapism is also of interest currently. This recurrent form of ischaemic priapism consists of undesirable, painful erections that last for less than 3 hours, with debilitating impacts on quality of life. Evidence for dutasteride is a promising as an emerging intervention for stuttering priapism. Daily dutasteride with consequent tapering was demonstrated to reduce episodic frequency without significant side effects (84).


Conclusions

Since their introduction in 1992 for the management of BPH, the clinical indications for 5-ARI have expanded to areas such as androgenic alopecia and hirsutism. This can be attributed to the increasing understanding of pathological processes involving the androgen axis in which 5-ARIs work. Several novel off-label indications have been suggested including its potential role in the pathogenesis of SARS-CoV-2, but to date these claims have not been substantiated. Previously held truths regarding the role of 5-ARIs and prostate carcinogenesis have been contested, inadvertently leading to the re-exploration of 5-ARIs utility in prostate cancer. Other future directions will continue to be elucidated, as the implications of 5-ARIs in androgen signalling continues to be explored.


Acknowledgments

Funding: None.


Footnote

Peer Review File: Available at https://tau.amegroups.com/article/view/10.21037/tau-22-690/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tau.amegroups.com/article/view/10.21037/tau-22-690/coif). EC serves as an unpaid editorial board member of Translational Andrology and Urology from August 2022 to July 2024. MLP reports that he has received the Australiasian Fulbright Commission Scholarship through the Kinghorn Foundation. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Pomerantz M. Propecia. International Society of Hair Restoration Surgery. 1998;8:13.1-13. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/020788s018lbl.pdf
  2. Avodart (Dutatsteride). Avodart FDA Prescribing Information. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/021319s015lbl.pdf
  3. Hsing AW, Reichardt JK, Stanczyk FZ. Hormones and prostate cancer: current perspectives and future directions. Prostate 2002;52:213-35. [Crossref] [PubMed]
  4. Thompson IM, Goodman PJ, Tangen CM, et al. The influence of finasteride on the development of prostate cancer. N Engl J Med 2003;349:215-24. [Crossref] [PubMed]
  5. Schmidt AL, Tucker MD, Bakouny Z, et al. Association Between Androgen Deprivation Therapy and Mortality Among Patients With Prostate Cancer and COVID-19. JAMA Netw Open 2021;4:e2134330. [Crossref] [PubMed]
  6. Goren A, Wambier CG, Herrera S, et al. Anti-androgens may protect against severe COVID-19 outcomes: results from a prospective cohort study of 77 hospitalized men. J Eur Acad Dermatol Venereol 2021;35:e13-5. [Crossref] [PubMed]
  7. Loughlin KR. The clinical applications of five-alpha reductase inhibitors. Can J Urol 2021;28:10584-8. [PubMed]
  8. Bramson HN, Hermann D, Batchelor KW, et al. Unique preclinical characteristics of GG745, a potent dual inhibitor of 5AR. J Pharmacol Exp Ther 1997;282:1496-502. [PubMed]
  9. Liu L, Zhao S, Li F, et al. Effect of 5α-Reductase Inhibitors on Sexual Function: A Meta-Analysis and Systematic Review of Randomized Controlled Trials. J Sex Med 2016;13:1297-310. [Crossref] [PubMed]
  10. Fertig RM, Gamret AC, Darwin E, et al. Sexual side effects of 5-α-reductase inhibitors finasteride and dutasteride: A comprehensive review. Dermatol Online J 2017;23:13030/qt24k8q743.
  11. Trost L, Saitz TR, Hellstrom WJ. Side Effects of 5-Alpha Reductase Inhibitors: A Comprehensive Review. Sex Med Rev 2013;1:24-41. [Crossref] [PubMed]
  12. Favilla V, Russo GI, Privitera S, et al. Impact of combination therapy 5-alpha reductase inhibitors (5-ARI) plus alpha-blockers (AB) on erectile dysfunction and decrease of libido in patients with LUTS/BPH: a systematic review with meta-analysis. Aging Male 2016;19:175-81. [Crossref] [PubMed]
  13. Lee S, Lee YB, Choe SJ, et al. Adverse Sexual Effects of Treatment with Finasteride or Dutasteride for Male Androgenetic Alopecia: A Systematic Review and Meta-analysis. Acta Derm Venereol 2019;99:12-7. [PubMed]
  14. Roehrborn CG, Siami P, Barkin J, et al. The effects of combination therapy with dutasteride and tamsulosin on clinical outcomes in men with symptomatic benign prostatic hyperplasia: 4-year results from the CombAT study. Eur Urol 2010;57:123-31. [Crossref] [PubMed]
  15. Welk B, McArthur E, Ordon M, et al. The risk of dementia with the use of 5 alpha reductase inhibitors. J Neurol Sci 2017;379:109-11. [Crossref] [PubMed]
  16. Wang J, Zhao S, Luo L, et al. 5-alpha Reductase Inhibitors and risk of male breast cancer: a systematic review and meta-analysis. Int Braz J Urol 2018;44:865-73. [Crossref] [PubMed]
  17. Saengmearnuparp T, Lojanapiwat B, Chattipakorn N, et al. The connection of 5-alpha reductase inhibitors to the development of depression. Biomed Pharmacother 2021;143:112100. [Crossref] [PubMed]
  18. Traish AM, Guay AT, Zitzmann M. 5α-Reductase inhibitors alter steroid metabolism and may contribute to insulin resistance, diabetes, metabolic syndrome and vascular disease: a medical hypothesis. Horm Mol Biol Clin Investig 2014;20:73-80. [Crossref] [PubMed]
  19. Livingstone DE, Barat P, Di Rollo EM, et al. 5α-Reductase type 1 deficiency or inhibition predisposes to insulin resistance, hepatic steatosis, and liver fibrosis in rodents. Diabetes 2015;64:447-58. [Crossref] [PubMed]
  20. Hsieh TF, Yang YW, Lee SS, et al. Use of 5-alpha-reductase inhibitors did not increase the risk of cardiovascular diseases in patients with benign prostate hyperplasia: a five-year follow-up study. PLoS One 2015;10:e0119694. [Crossref] [PubMed]
  21. Pereira AFJR, Coelho TOA. Post-finasteride syndrome. An Bras Dermatol 2020;95:271-7. [Crossref] [PubMed]
  22. Gray SL, Semla TP. Post-finasteride syndrome. BMJ 2019;366:l5047. [Crossref] [PubMed]
  23. Lerner LB, McVary KT, Barry MJ, et al. Management of Lower Urinary Tract Symptoms Attributed to Benign Prostatic Hyperplasia: AUA GUIDELINE PART I-Initial Work-up and Medical Management. J Urol 2021;206:806-17. [Crossref] [PubMed]
  24. Muruganandham K, Dubey D, Kapoor R. Acute urinary retention in benign prostatic hyperplasia: Risk factors and current management. Indian J Urol 2007;23:347-53. [Crossref] [PubMed]
  25. McConnell JD, Roehrborn CG, Bautista OM, et al. The long-term effect of doxazosin, finasteride, and combination therapy on the clinical progression of benign prostatic hyperplasia. N Engl J Med 2003;349:2387-98. [Crossref] [PubMed]
  26. Boyle P, Gould AL, Roehrborn CG. Prostate volume predicts outcome of treatment of benign prostatic hyperplasia with finasteride: meta-analysis of randomized clinical trials. Urology 1996;48:398-405. [Crossref] [PubMed]
  27. Roehrborn CG, Bruskewitz R, Nickel GC, et al. Urinary retention in patients with BPH treated with finasteride or placebo over 4 years. Characterization of patients and ultimate outcomes. The PLESS Study Group. Eur Urol 2000;37:528-36. [Crossref] [PubMed]
  28. McConnell JD, Bruskewitz R, Walsh P, et al. The effect of finasteride on the risk of acute urinary retention and the need for surgical treatment among men with benign prostatic hyperplasia. Finasteride Long-Term Efficacy and Safety Study Group. N Engl J Med 1998;338:557-63. [Crossref] [PubMed]
  29. Shum CF, Lau W, Teo CPC. Medical therapy for clinical benign prostatic hyperplasia: α1 Antagonists, 5α reductase inhibitors and their combination. Asian J Urol 2017;4:185-90. [Crossref] [PubMed]
  30. Bortnick E, Brown C, Simma-Chiang V, et al. Modern best practice in the management of benign prostatic hyperplasia in the elderly. Ther Adv Urol 2020;12:1756287220929486. [Crossref] [PubMed]
  31. Kloping YP, Yogiswara N, Azmi Y. The role of preoperative dutasteride in reducing bleeding during transurethral resection of the prostate: A systematic review and meta-analysis of randomized controlled trials. Asian J Urol 2022;9:18-26. [Crossref] [PubMed]
  32. Ren J, Lai S, Jiang Z, et al. A Systematic Review and Meta-Analysis of the Effects on Dutasteride Treatment for Reducing Surgical Blood Loss during Transurethral Resection of the Prostate. Urol Int 2017;98:456-65. [Crossref] [PubMed]
  33. Loureiro RM, D'Amore PA. Transcriptional regulation of vascular endothelial growth factor in cancer. Cytokine Growth Factor Rev 2005;16:77-89. [Crossref] [PubMed]
  34. Eisermann K, Fraizer G. The Androgen Receptor and VEGF: Mechanisms of Androgen-Regulated Angiogenesis in Prostate Cancer. Cancers (Basel) 2017;9:32. [Crossref] [PubMed]
  35. Kearney MC, Bingham J, Bergland R, et al. Clinical predictors in the use of finasteride for control of gross hematuria due to benign prostatic hyperplasia. J Urol 2002;167:2489-91. [Crossref] [PubMed]
  36. Pareek G, Shevchuk M, Armenakas NA, et al. The Effect of Finasteride on the Expression of Vascular Endothelial Growth Factor and Microvessel Density: A Possible Mechanism for Decreased Prostatic Bleeding in Treated Patients. J Urol 2003;169:20-3. [Crossref] [PubMed]
  37. Hochberg DA, Basillote JB, Armenakas NA, et al. Decreased Suburethral Prostatic Microvessel Density In Finasteride Treated Prostates: A Possible Mechanism For Reduced Bleeding In Benign Prostatic Hyperplasia. J Urol 2002;167:1731-3. [Crossref] [PubMed]
  38. Lekas AG, Lazaris AC, Chrisofos M, et al. Finasteride effects on hypoxia and angiogenetic markers in benign prostatic hyperplasia. Urology 2006;68:436-41. [Crossref] [PubMed]
  39. Pienta KJ, Bradley D. Mechanisms underlying the development of androgen-independent prostate cancer. Clin Cancer Res 2006;12:1665-71. [Crossref] [PubMed]
  40. Hou Z, Huang S, Li Z. Androgens in prostate cancer: A tale that never ends. Cancer Lett 2021;516:1-12. [Crossref] [PubMed]
  41. Zhu J, Gao JP, Xu AX, et al. The influence of benign prostatic hyperplasia drugs on incidence and pathology grading of prostate cancer. Zhonghua Wai Ke Za Zhi 2010;48:761-3. [PubMed]
  42. Bosland MC, Cremers RG, Kiemeney LA. Words of wisdom. Re: effect of dutasteride on the risk of prostate cancer. Eur Urol 2010;58:631-2. [Crossref] [PubMed]
  43. Thompson I Jr, Goodman P, Tangen C. Reduced Risk of Prostate Cancer With 5α-Reductase Inhibitors. J Natl Cancer Inst 2018;110:1159-60. [Crossref] [PubMed]
  44. Wilt TJ, Macdonald R, Hagerty K, et al. 5-α-Reductase inhibitors for prostate cancer chemoprevention: an updated Cochrane systematic review. BJU Int 2010;106:1444-51. [Crossref] [PubMed]
  45. Park JJ, Lee HY, Shim SR, et al. Prostate cancer specific mortality after 5α-reductase inhibitors medication in benign prostatic hyperplasia patients: systematic review and meta-analysis. Aging Male 2021;24:80-91. [Crossref] [PubMed]
  46. Knijnik PG, Brum PW, Cachoeira ET, et al. The impact of 5-alpha-reductase inhibitors on mortality in a prostate cancer chemoprevention setting: a meta-analysis. World J Urol 2021;39:365-76. [Crossref] [PubMed]
  47. Yang Y, Hu H, Zhang H, et al. 5α-Reductase Inhibitors Could Prevent the Clinical and Pathological Progression of Prostate Cancer: A Meta-analysis. Urol J 2021;18:247-51. [PubMed]
  48. Deng T, Lin X, Duan X, et al. Prostate cancer patients can benefit from 5-alpha-reductase inhibitor treatment: a meta-analysis. PeerJ 2020;8:e9282. [Crossref] [PubMed]
  49. Wu Y, Wang Y, Gu Y, et al. Prostate Cancer Risk and Prognostic Influence Among Users of 5-Alpha-Reductase Inhibitors and Alpha-Blockers: A Systematic Review and Meta-Analysis. Urology 2020;145:216-23. [Crossref] [PubMed]
  50. Hu X, Wang YH, Yang ZQ, et al. Association of 5-alpha-reductase inhibitor and prostate cancer incidence and mortality: a meta-analysis. Transl Androl Urol 2020;9:2519-32. [Crossref] [PubMed]
  51. Foldes CA, Wang R, Canfield SE. Prostate Cancer Mortality and Use of 5-Alpha Reductase Inhibitors. World J Mens Health 2020;38:139-40. [Crossref] [PubMed]
  52. Schatzl G, Madersbacher S, Haitel A, et al. Associations of serum testosterone with microvessel density, androgen receptor density and androgen receptor gene polymorphism in prostate cancer. J Urol 2003;169:1312-5. [Crossref] [PubMed]
  53. Liss MA, Thompson IM. Prostate cancer prevention with 5-alpha reductase inhibitors: concepts and controversies. Curr Opin Urol 2018;28:42-5. [Crossref] [PubMed]
  54. Thompson IM, Tangen CM, Goodman PJ, et al. Finasteride improves the sensitivity of digital rectal examination for prostate cancer detection. J Urol 2007;177:1749-52. [Crossref] [PubMed]
  55. Thompson IM, Chi C, Ankerst DP, et al. Effect of finasteride on the sensitivity of PSA for detecting prostate cancer. J Natl Cancer Inst 2006;98:1128-33. [Crossref] [PubMed]
  56. Civantos F, Soloway MS, Pinto JE. Histopathological effects of androgen deprivation in prostatic cancer. Semin Urol Oncol 1996;14:22-31. [PubMed]
  57. Nelson PS, Clegg N, Arnold H, et al. The program of androgen-responsive genes in neoplastic prostate epithelium. Proc Natl Acad Sci U S A 2002;99:11890-5. [Crossref] [PubMed]
  58. Morgia G, Urzì D, Russo GI. 5ARI and PSA: evidences. Urologia 2014;81:4-11. [Crossref] [PubMed]
  59. Pannek J, Marks LS, Pearson JD, et al. Influence of finasteride on free and total serum prostate specific antigen levels in men with benign prostatic hyperplasia. J Urol 1998;159:449-53. [Crossref] [PubMed]
  60. Etzioni RD, Howlader N, Shaw PA, et al. Long-term effects of finasteride on prostate specific antigen levels: results from the prostate cancer prevention trial. J Urol 2005;174:877-81. [Crossref] [PubMed]
  61. Carter HB, Albertsen PC, Barry MJ, et al. Early detection of prostate cancer: AUA Guideline. American Urological Association (AUA) Guideline, 2013:1-28.
  62. Sakalis V, Gkotsi A, Charpidou D, et al. The effect of pharmacotherapy on prostate volume, prostate perfusion and prostate-specific antigen (prostate morphometric parameters) in patients with lower urinary tract symptoms and benign prostatic obstruction. A systematic review and meta-analysis. Cent European J Urol 2021;74:388-421. [PubMed]
  63. Peckham H, De Gruijter NM, Raine C, et al. Male sex identified by global COVID-19 meta-analysis as a risk factor for death and ITU admission. Nat Commun 2020;11:6317. [Crossref] [PubMed]
  64. Mollica V, Rizzo A, Massari F. The pivotal role of TMPRSS2 in coronavirus disease 2019 and prostate cancer. Future Oncol 2020;16:2029-33. [Crossref] [PubMed]
  65. Ianhez M, Ramos PM, Goren A, et al. Androgen sensitivity in COVID-19 and antiandrogens: Prospective data are still needed. Dermatol Ther 2020;33:e14166. [PubMed]
  66. Lyon M, Li J, Cullen J, et al. 5α-Reductase Inhibitors Are Associated with Reduced Risk of SARS-CoV-2 Infection: A Matched-Pair, Registry-Based Analysis. J Urol 2022;207:183-9. [Crossref] [PubMed]
  67. Karimi A, Nowroozi A, Alilou S, et al. Effects of Androgen Deprivation Therapy on COVID-19 in Patients with Prostate Cancer: A Systematic Review and Meta-Analysis. Urol J 2021;18:577-84. [PubMed]
  68. Cadegiani FA, McCoy J, Gustavo Wambier C, et al. Early Antiandrogen Therapy With Dutasteride Reduces Viral Shedding, Inflammatory Responses, and Time-to-Remission in Males With COVID-19: A Randomized, Double-Blind, Placebo-Controlled Interventional Trial (EAT-DUTA AndroCoV Trial - Biochemical). Cureus 2021;13:e13047. [Crossref] [PubMed]
  69. Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020;181:271-280.e8. [Crossref] [PubMed]
  70. Tomlins SA, Rhodes DR, Perner S, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 2005;310:644-8. [Crossref] [PubMed]
  71. Wambier CG, Vaño-Galván S, McCoy J, et al. Androgenetic alopecia present in the majority of patients hospitalized with COVID-19: The "Gabrin sign". J Am Acad Dermatol 2020;83:680-2. [Crossref] [PubMed]
  72. Goren A, McCoy J, Wambier CG, et al. What does androgenetic alopecia have to do with COVID-19? An insight into a potential new therapy. Dermatol Ther 2020;33:e13365. [PubMed]
  73. Viveiros A, Rasmuson J, Vu J, et al. Sex differences in COVID-19: candidate pathways, genetics of ACE2, and sex hormones. Am J Physiol Heart Circ Physiol 2021;320:H296-304. [Crossref] [PubMed]
  74. Chang KH, Li R, Papari-Zareei M, et al. Dihydrotestosterone synthesis bypasses testosterone to drive castration-resistant prostate cancer. Proc Natl Acad Sci U S A 2011;108:13728-33. [Crossref] [PubMed]
  75. Baratchian M, McManus JM, Berk MP, et al. Androgen regulation of pulmonary AR, TMPRSS2 and ACE2 with implications for sex-discordant COVID-19 outcomes. Sci Rep 2021;11:11130. [Crossref] [PubMed]
  76. Samuel RM, Majd H, Richter MN, et al. Androgen Signaling Regulates SARS-CoV-2 Receptor Levels and Is Associated with Severe COVID-19 Symptoms in Men. Cell Stem Cell 2020;27:876-889.e12. [Crossref] [PubMed]
  77. Lolli F, Pallotti F, Rossi A, et al. Androgenetic alopecia: a review. Endocrine 2017;57:9-17. [Crossref] [PubMed]
  78. Lee SW, Juhasz M, Mobasher P, et al. A Systematic Review of Topical Finasteride in the Treatment of Androgenetic Alopecia in Men and Women. J Drugs Dermatol 2018;17:457-63. [PubMed]
  79. Dhurat R, Sharma A, Rudnicka L, et al. 5-Alpha reductase inhibitors in androgenetic alopecia: Shifting paradigms, current concepts, comparative efficacy, and safety. Dermatol Ther 2020;33:e13379. [PubMed]
  80. Heidelbaugh JJ. Endocrinology Update: Hirsutism. FP Essent 2016;451:17-24. [PubMed]
  81. Brodell LA, Mercurio MG. Hirsutism: Diagnosis and management. Gend Med 2010;7:79-87. [Crossref] [PubMed]
  82. Soares Júnior JM, Guimarães DZ, Simões RDS, et al. Systematic review of finasteride effect in women with hirsutism. Rev Assoc Med Bras (1992) 2021;67:1043-9. [PubMed]
  83. Covault J, Pond T, Feinn R, et al. Dutasteride reduces alcohol’s sedative effects in men in a human laboratory setting and reduces drinking in the natural environment. Psychopharmacology 2014;231:3609-18. [Crossref] [PubMed]
  84. Baker RC, Bergeson RL, Yi YA, et al. Dutasteride in the long-term management of stuttering priapism. Transl Androl Urol 2020;9:87-92. [Crossref] [PubMed]
Cite this article as: Chislett B, Chen D, Perera ML, Chung E, Bolton D, Qu LG. 5-alpha reductase inhibitors use in prostatic disease and beyond. Transl Androl Urol 2023;12(3):487-496. doi: 10.21037/tau-22-690

Download Citation