Skip to main content
Log in

Finasteride

A Review of its Potential in the Treatment of Benign Prostatic Hyperplasia

  • Drug Evaluation
  • Published:
Drugs Aims and scope Submit manuscript

Abstract

Synopsis

Finasteride is a novel therapeutic agent that selectively inhibits the enzyme 5α-reductase, thereby reducing prostatic dihydrotestosterone (DHT) levels and prostate size. In men with symptomatic benign prostatic hyperplasia (BPH), these effects have been associated with improvements in peak urinary flow rate and urological symptoms; withdrawal from therapy, however, results in regrowth of the adenoma and long term therapy is therefore necessary. Although the magnitude of clinical improvement seen with finasteride has been perceived to be modest [especially when compared with that associated with transurethral resection of the prostate (TURP)], it has been maintained in the medium term (up to 2 years) and thus may represent significant reversal of disease progression. Such beneficial effects, however, may not become apparent until completion of at least 6 months of therapy. Furthermore, since clinical studies have been unable to proactively identify a responsive subgroup, a trial period of 6 or possibly 12 months is necessary to assess patient responsiveness. Despite these potential shortcomings, the benefits of therapy appear to outweigh the risks. Indeed, finasteride is well tolerated; most adverse events have been related to sexual dysfunction (decreased libido, ejaculation disorders and impotence) and occurred in only a small proportion (about 2 to 3%) of patients. Moreover, although there has been concern that finasteride might mask the detection of prostate cancer through its decrementai effects on serum prostate specific antigen (PSA) levels, careful monitoring in clinical trials appears to have avoided this problem. Thorough pretreatment assessment and periodic follow-up examinations for malignancy are therefore required in clinical practice.

The role of finasteride in the treatment of patients with BPH is still emerging and will no doubt gain in clarity with further planned investigations. TURP (or other invasive procedures such as the insertion of prostatic stents in patients unsuitable for resection), continues to be the mainstay of therapy for those patients with severe symptomatic BPH. However, available data support a first line role for finasteride in the treatment of patients with uncomplicated symptomatic BPH. Within this setting, finasteride appears to offer a needed additional treatment option for those patients in whom surgery is not indicated, and may be of special benefit to the considerable proportion of patients who opt not to undergo prostatectomy.

Pharmacodynamic Properties

Competitive inhibition of 5α-reductase by finasteride has been demonstrated in a series of in vitro experiments. In animal studies, acute administration of finasteride to the rat or dog resulted in a significant reduction in prostatic dihydrotestosterone (DHT) levels and a significant increase in prostatic testosterone. With longer periods of administration (up to 12 weeks), the decrementai effects of finasteride on canine prostatic DHT levels were maintained and prostate size declined by ≈33 to 47%.

In healthy volunteers administered finasteride as a single dose, maximum suppression of plasma DHT levels (⩾50%) was shown to occur at doses greater than 0.5mg. Little variation in the magnitude of this effect was evident over a wide range of doses above 5mg. In a similar population, plasma DHT was suppressed by >60% following continued administration of finasteride 0.04 to 100 mg/day for 1 to 3 weeks. The ‘no-effect multiple-dosage’ was shown to lie between 5 and 40 μg/day. Although biochemical effects are evident with low finasteride doses, the results of phase II and III investigations suggest that dosage of 1 or 5 mg/day are required to produce clinically significant effects in patients with BPH. In healthy volunteers, no clear overall effect of finasteride on circulating testosterone was demonstrated, and where fluctuations in this parameter were recorded, values remained within the accepted normal range. The ability to detect small changes in testosterone levels, however, was limited by the small numbers of participants and the known assay variability. Other hormones [including follicle stimulating hormone (FSH), luteinizing hormone (LH), cortisol and estradiol] and serum lipids were unaffected by finasteride administration, although at supratherapeutic doses Δ4-androstenedione levels became elevated.

In men with symptomatic BPH treated with finasteride 1 or 5 mg/day for 1 year, the major reduction in prostate volume occurred in the periurethral zone, which is responsible for urethral obstruction. The serum sex hormone profile of patients with BPH administered finasteride confirmed similar findings to those recorded in studies of healthy volunteers. In addition, patients awaiting transurethral resection of the prostate (TURP) administered finasteride 1 to 100 mg/day for 1 week showed a reduction in prostatic DHT levels of more than 90% at all dosage levels with a significant increase in prostatic testosterone.

Finasteride does not appear to have a direct effect on androgenic mechanisms in vivo and had no significant affinity for the androgen receptor in vitro. As expected, at low dosages (⩾0.003 mg/kg/day), it caused dysmorphogenesis of male offspring in animal studies.

Pharmacokinetic Properties

The bioavailability of finasteride 5mg is 80% and most absorption occurs within 2 hours. Food does not affect its pharmacokinetic profile. The plasma profile of finasteride showed dose proportionality after single oral doses of 5 to 50mg, although at higher doses conflicting results were reported. Healthy volunteers administered oral finasteride 5mg showed maximum plasma concentrations of 37 to 41 μg/L approximately 2 hours later, while values for the area under the plasma concentration-time curve (AUC) were 349 to 385 μg/L · h and terminal elimination phase half-life ranged from 4.8 to 6.0h.

In elderly (⩾70 years) healthy volunteers, daily administration of finasteride 5mg for 17 days resulted in a plasma pharmacokinetic profile characteristic of slower elimination when compared with that of their younger (45 to 60 years) healthy counterparts. Plasma finasteride concentrations increased with time in both groups, although notably the increases in minimum plasma finasteride concentration (Cmin) and AUC were greater among the elderly. At steady-state, Cmin values of 8.3 and 9.6 μg/L were predicted for younger and elderly volunteers, which were marginally greater than those observed after 17 days.

Finasteride undergoes wide tissue distribution. Plasma protein binding is approximately 90% over a concentration range of 0.02 to 2 μg/L and is not affected by moderate or severe renal impairment. In humans, finasteride is extensively metabolised through oxidative mechanisms and eliminated in the faeces and urine. Patients with moderate to severe renal dysfunction showed a decrease in urinary excretion of radiolabelled finasteride which correlated with the severity of the condition, but was compensated by an increase in faecal elimination. An increase of AUC0–48h of ≈ 35% associated with renal impairment does not appear to warrant dosage adjustment in this patient group as high doses of finasteride are well tolerated.

Therapeutic Potential of Finasteride in Benign Prostatic Hyperplasia

The clinical efficacy of finasteride was first demonstrated in a series of pivotal phase II placebo-controlled, randomised, double-blind clinical investigations in which dosages of 0.2 to 40 mg/day or 5 to 80 mg/day were administered to men with symptomatic BPH for up to 24 weeks. Significant reductions in circulating DHT levels were reflected in significant prostate shrinkage after 24 weeks with dosage levels of 1 mg/day and above; concomitant reductions in prostate specific antigen (PSA) levels were also evident. Peak urinary flow rate improved by at least 3 ml/sec for most finasteride-treated groups, while urological symptoms assessed by evaluating total (obstructive and nonobstructive) and obstructive symptom scores (using an adapted Boyarski scale) showed a trend towards improvement. Cessation of therapy was associated with reversal of the beneficial clinical effects suggesting the need for continual therapy.

In well designed phase III placebo-controlled, randomised, double-blind trials, 1645 men with mild to severe symptomatic BPH were administered finasteride 1 or 5 mg/day for 1 year. Significant reductions in serum mean DHT levels (45 to 66%) again reflected in prostate shrinkage (median ≈20%) at both dosages, although 6 months of therapy was necessary before the maximum effect on the prostate became evident. Concomitant reductions in serum mean PSA levels (43 to 50%) were also observed. Peak urinary flow rate increased modestly by 1.3 to 1.7 ml/sec, although it may be argued that this degree of improvement represents a reversal of 6.5 to 7.5 years of declining function. Total symptom score decreased slightly by about 3 to 4 points (on a scale of 0 to 36) for patients receiving finasteride 5 mg/day and by about 2 to 4 points for those receiving 1 mg/day. Obstructive symptom scores followed a similar trend. Reductions in prostate size and improvements in urinary flow and symptom scores during finasteride therapy were partially offset by the larger than expected placebo response. There was no correlation between efficacy parameters and disease severity, and subgroup analysis revealed reductions in prostate size and increases in urine flow rate that were similar regardless of baseline values. In noncomparative extensions to phase HI studies the clinical effects of finasteride 5 mg/day were maintained for 2 years.

Tolerability

Available data from 1398 patients treated with finasteride 0.2 to 80 mg/day for periods of up to 1 year in phase II and III investigations show that the drug is well tolerated. Most adverse events were related to sexual dysfunction (decreased libido, ejaculation disorders and impotence) and occurred in only a small proportion of the patient population (2.2 to 3.4% vs 0.8 to 1.6% for placebo); similar findings were observed when only patients recruited to phase III studies were considered. Withdrawal rates were essentially similar for finasteride- and placebo-treated groups in phase III investigations (≈5%), although withdrawal associated with sexual adverse events was higher among finasteride recipients (0.5 to 0.9 vs 0.2%). No fatalities attributable to finasteride were noted. Finasteride did not mask the detection of prostate cancer through its effects on PSA levels. Indeed, the detection rate for prostate cancer was twice that noted for the general population (probably reflecting close patient monitoring) and most patients who developed the disease had elevated PSA levels at baseline. A greater proportion of finasteride-treated patients showed increased neutrophil counts, blood urea nitrogen and glucose levels, but these findings were not corroborated by changes in other parameters or the results of other investigations. Small increases in circulating testosterone and LH levels, and slight changes in other circulating hormone levels associated with finasteride therapy were considered not to be of clinical relevance.

Dosage and Administration

The recommended oral dosage of finasteride is 5mg daily. Dosage adjustment in patients with renal insufficiency or in the elderly appear unwarranted. Finasteride is not indicated for use in women or children. Furthermore, due to the dysmorphogenic effects of finasteride on male offspring, women who are, or who may become, pregnant should avoid exposure to the drug. In this respect, patients should avoid transvaginal exposure via the semen of their sexual partner as a precautionary measure.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  • Abrahams PH. Urodynamic results of surgery. In Hinman & Boyarsky (Eds) Benign prostatic hypertrophy, pp. 948–956. Springer-Verlag, New York, 1983

    Chapter  Google Scholar 

  • Anderson KM, Liao S. Selective retention of dihydrotestosterone by prostatic nuclei. Nature 219: 277–279, 1968

    Article  PubMed  CAS  Google Scholar 

  • Anonymous. FDC Reports Pink Sheet 54: 9–13, 1992

    Google Scholar 

  • Bardin CW, Catterall JF. Testosterone: a major determinant of extragenital sexual dimorphism. Science 211: 1285–1294, 1981

    Article  PubMed  CAS  Google Scholar 

  • Barry MJ, Mulley Jr AG, Fowler FJ, Wennberg JW. Watchful waiting vs immediate transurethral resection for symptomatic prostatism. The importance of patients’ preferences. Journal of the American Medical Association 259: 3010–3017, 1988

    Article  PubMed  CAS  Google Scholar 

  • Baum MJ, Vreeburg JTM. Copulation in castrated male rats following combined treatment with estradiol and dihydrotestosterone. Science 182: 283–285, 1973

    Article  PubMed  CAS  Google Scholar 

  • Beisland HO, Binkowitz B, Brekkan E, Ekman P, Konturri M, et al. Scandinavian clinical study of finasteride in the treatment of benign prostatic hyperplasia. European Urology 22: 271–277, 1992

    PubMed  CAS  Google Scholar 

  • Berry SJ, Coffey DS, Walsh PC, Ewing LL. The development of human benign prostatic hyperplasia with age. Journal of Urology 132: 474–479, 1984

    PubMed  CAS  Google Scholar 

  • Birkhoff JD. Natural history of benign prostatic hypertrophy. In Hinnman Jr (Ed.) Benign prostatic hypertrophy, pp. 5–9, Springer-Verlag, New York, 1983

    Chapter  Google Scholar 

  • Birkhoff JD, Wiederhorn AR, Hamilton ML, Zinsser HH. Natural history of benign prostatic hypertrophy and acute urinary retention. Urology 7: 48–52, 1976

    Article  PubMed  CAS  Google Scholar 

  • Bolognese JA, Kozioh RC, Grino PB, Patric DL, Stoner E. Validation of a symptoms questionnaire for benign prostatic hyperplasia. Prostate 21: 247–254, 1992

    Article  PubMed  CAS  Google Scholar 

  • Bosch RJLH. Pathogenesis of benign prostatic hyperplasia. European Urology (Suppl.) 20: 27–30, 1991

    Google Scholar 

  • Boyarski S, Jones G, Paulson DF, Prout Jr GR. A new look at bladder neck obstruction by the Food and Drug Administration regulators; guidelines for investigation of benign prostatic hypertrophy. Transcripts of the American Association of Genitourinary Surgery 68: 29–32, 1976

    Google Scholar 

  • Brooks JR, Berman C, Games D, Giltinan D, Gordon LR, et al. Prostatic effects induced in dogs by chronic or acute oral administration of 5α-reductase inhibitors. Prostate 9: 65–75, 1986a

    Article  PubMed  CAS  Google Scholar 

  • Brooks JR, Berman C, Primka RL, Reynolds GF, Rasmusson GH. 5α-reductase inhibitory and anti-androgenic activities of some 4-azasteroids in the rat. Steroids 47: 1–19, 1986b

    Article  PubMed  CAS  Google Scholar 

  • Bruchovsky N, Wilson JD. The conversion of testosterone to 5α-androstan-17β-ol-3-one by rat prostate in vivo and in vitro. Journal of Biological Chemistry 243: 2012–2021, 1968

    PubMed  CAS  Google Scholar 

  • Bruskewitz RC, Christensen MM. Critical evaluation of transurethral resection and incision of the prostate. Prostate 3 (Suppl.): 27–38, 1990

    Article  PubMed  CAS  Google Scholar 

  • Caine M. The present role of alpha-adrenergic blockers in the treatment of benign prostatic hypertrophy. Journal of Urology 136: 1–4, 1986

    PubMed  CAS  Google Scholar 

  • Caine M, Perlberg S, Meretyk S. A placebo-controlled, double-blind study of the effect of phenoxybenzamine in benign prostatic obstruction. British Journal of Urology 50: 551, 1978

    Article  PubMed  CAS  Google Scholar 

  • Carlin JR, Höglund P, Eriksson L-O, Christafalo P, Gregoire SL, et al. Disposition and pharmacokinetics of [14C]finasteride after oral administration in humans. Drug Metabolism and Disposition 20: 148–155, 1992

    PubMed  CAS  Google Scholar 

  • Carlin JR, Christofalo P, Vandenheuvel WJA. High-performance liquid Chromatographic determination of N-(2-methyl-2-propyl)-3-oxo-4-aza-5α-androst-1-ene-17β-carboxamide, A 4-azasteroid, in human plasma from a phase I study. Journal of Chromatography 427: 79–91, 1988

    Article  PubMed  CAS  Google Scholar 

  • Chrisp P, Goa KL. Nafarelin: a review of its pharmacodynamic and pharmacokinetic properties, and clinical potential in sex hormone-related conditions. Drugs 39: 523–551, 1990

    Article  PubMed  CAS  Google Scholar 

  • Chrisp P, Sorkin EM. Leuprorelin: a review of its pharmacology and therapeutic use in prostatic disorders. Drugs & Aging 1: 487–509, 1991

    Article  CAS  Google Scholar 

  • Clark JH, Schrader WT, O’Malley BW. Mechanism of steroid hormone action. In Wilson & Foster (Eds) Textbook of endocrinology, pp. 33–122. Philadelphia, PA, 1985

    Google Scholar 

  • Clark RL, Antonello JM, Grossman SJ, Wise LD, Anderson C, et al. External genitalia abnormalities in male rats exposed in utero to fmasteride, a 5α-reductase inhibitor. Teratology 42: 91–100, 1990

    Article  PubMed  CAS  Google Scholar 

  • Cohen SM, Taber KH, Malatesta PF, Shpungin J, Berman C. Magnetic resonance imaging of the efficacy of specific inhibition of 5α-reductase in canine spontaneous benign prostatic hyperpla-sia. Magnetic Resonance in Medicine 21: 55–70, 1991

    Article  PubMed  CAS  Google Scholar 

  • Constanzer ML, Matuszewski BK, Bayne WF. High-performance liquid Chromatographic method for the determination of finasteride in human plasma at therapeutic doses. Journal of Chromatography 566: 127–134, 1991

    Article  PubMed  CAS  Google Scholar 

  • Davis J. Are drugs worthwhile in prostatic hyperplasia? Scrip No. 1768: 24, 1992

    Google Scholar 

  • De Schepper PJ, Imperato-McGinley J, Van Hecken A, De Lepeleire I, Buntinx A, et al. Hormonal effects, tolerability and preliminary kinetics in men of MK-906, a 5α-reductase inhibitor. Steroids 56: 469–471, 1991

    Article  PubMed  Google Scholar 

  • D’Occhio MJ, Brooks DE. Effects of androgenic and oestrogenic hormones on mating behaviour in rams castrated before or after puberty. Journal of Endocrinology 86: 403–411, 1980

    Article  PubMed  Google Scholar 

  • Dørflinger T, England DM, Madsen PO, Bruskewitz RC. Urodynamic and histological correlates of benign prostatic hyperplasia. Journal of Urology 140: 1487, 1988

    PubMed  Google Scholar 

  • DuBeau CE. Finasteride for benign prostatic hyperplasia. Correspondence. New England Journal of Medicine 328: 443, 1993

    PubMed  CAS  Google Scholar 

  • Dunzendorfer U. Clinical experience with symptomatic management of BPH with terazosin. Urology (Suppl. 6) 32: 27, 1988

    CAS  Google Scholar 

  • Ekman P. BPH epidemiology and risk factors. Prostate 2 (Suppl.): 23–31, 1989

    Article  PubMed  CAS  Google Scholar 

  • Finasteride Study Group. Finasteride (MK-906) in the treatment of benign prostatic hyperplasia. Prostate, in press, 1993

  • Fowler Jr FJ, Wennberg JE, Timothy RP, Barry MJ, Mulley Jr AG, et al. Symptom status and quality of life following prostatectomy. Journal of the American Medical Association 259: 3018–3022, 1988

    Article  PubMed  Google Scholar 

  • Furuya S, Kumamoto Y, Yokoyama E, Tsukamoto T, Izumi T, et al. Alpha-adrenergic activity and urethral pressure in prostatic region in benign prostatic hypertrophy. Journal of Urology 128: 836–839, 1982

    PubMed  CAS  Google Scholar 

  • Geller J. Effect of finasteride, a 5α-reductase inhibitor on prostate tissue androgens and prostate-specific antigen. Journal of Clinical Endocrinology and Metabolism 71: 1552–1555, 1990

    Article  PubMed  CAS  Google Scholar 

  • Geller J. Pathogenesis and medical treatment of benign prostatic hyperplasia. Prostate 1 (Suppl.): 95–104, 1989

    Article  Google Scholar 

  • George FW, Johnson L, Wilson JD. The effect of a 5α-reductase inhibitor on androgen physiology in the immature male rat. Endocrinology 125: 2434–2438, 1989

    Article  PubMed  CAS  Google Scholar 

  • Gingell JC. Review of current and future approaches to the management of benign prostatic hyperplasia. Postgraduate Medical Journal 68: 702–706, 1992

    Article  PubMed  CAS  Google Scholar 

  • Gormley GJ, Stoner E, Bruskewitz RC, Imperato-McGinley J, Walsh PC, et al. The effect of finasteride in men with benign prostatic hyperplasia. New England Journal of Medicine 327: 1185–1192, 1992

    Article  PubMed  CAS  Google Scholar 

  • Gormley GJ, Stoner E, Rittmaster RS, Gregg H, Thompson DL, et al. Effects of finasteride (MK-906), a 5α-reductase inhibitor, on circulating androgens in male volunteers. Journal of Clinical Endocrinology and Metabolism 70: 1136–1141, 1990

    Article  PubMed  CAS  Google Scholar 

  • Gormley GJ, Stoner E, Slater EE. Finasteride for benign prostatic hyperplasia. Correspondence. New England Journal of Medicine 328: 443, 1993

    Google Scholar 

  • Gregoire S, Winchell G, Constanzer M, Tocco D, Jacobsen CA, et al. The effect of renal function on the disposition of finasteride, a new 5α-reductase inhibitor. Abstract 73. Journal of Clinical Pharmacology 31: 859, 1990

    Google Scholar 

  • Guess HA, Heyse JF, Gormley GJ. The effect of finasteride on prostate-specific antigen in men with benign prostatic hyperplasia. Prostate 22: 31–37, 1993

    Article  PubMed  CAS  Google Scholar 

  • Huggins C. In Vollmer & Kauppmann (Eds) Introduction in ‘Biology of the prostate and related tissues’. NCI monograph 12, Washington DC. US Government Printing Office, 1962

  • Imperato-McGinley J, Querrero L, Gautier T, Peterson RE. Steroid 5α-reductase deficiency in man: an inherited form of male pseudohermaphroditism. Science 186: 1213–1215, 1974

    Article  PubMed  CAS  Google Scholar 

  • Imperato-McGinley J, Shackleton C, Orlic S, Stoner E. C19 and C21 5β/5α metabolite ratios in subjects treated with the 5α-reductase inhibitor finasteride: comparison of male pseudo-hermaphrodites with inherited 5α-reductase deficiency. Journal of Clinical Endocrinology and Metabolism 70: 777–782, 1990

    Article  PubMed  CAS  Google Scholar 

  • Isaacs JT, Coffey DS. Etiology and disease process of benign prostatic hyperplasia. Prostate 2 (Suppl.): 33–50, 1989

    Article  PubMed  CAS  Google Scholar 

  • Janknegt RA. Surgical management for benign prostatic hyperplasia: indications, techniques and results. Prostate 2 (Suppl.): 79–93, 1989

    Article  PubMed  CAS  Google Scholar 

  • Jenkins EP, Andersson S, Imperato-McGinley J, Wilson JD, Russell DW. Genetic and pharmacological evidence for more than one human steroid 5α-reductase. Journal of Clinical Investigation 89: 293–300, 1992

    Article  PubMed  CAS  Google Scholar 

  • Kato R, Onoda K-I, Omori Y. Mechanism of thyroxine-induced increase in steroid Δ4-reductase activity in male rats. Endocrinologica Japonica 17: 215–219, 1970

    Article  CAS  Google Scholar 

  • Kirby RS. Alpha-adrenoceptor inhibitors in the treatment of benign prostatic hyperplasia. American Journal of Medicine 87 (Suppl. 2A): 26s–30s, 1989

    Article  PubMed  CAS  Google Scholar 

  • Kirby RS, Bryan J, Eardley I, Christmas TJ, Liu S, et al. Finasteride in the treatment of benign prostatic hyperplasia. A urodynamic evaluation. British Journal of Urology 70: 65–72, 1992

    Article  PubMed  CAS  Google Scholar 

  • Kirby RS, Coppinger SWC, Corcoran MV, Chappie CR, Flannigan M, et al. Prazosin in the treatment of prostatic obstruction. A placebo-controlled study. British Journal of Urology 60: 136, 1987

    Article  PubMed  CAS  Google Scholar 

  • Kontturi M. Symptoms and patient evaluation. In Altwein (Ed.) Benign prostatic hyperplasia: a diagnosis and treatment primer, p. 70, Merck and Co. Inc., Rahway, New Jersey, US 1992

    Google Scholar 

  • Lepor H. Can BPH be treated with drugs? Contemporary Urology: 15–23, June/July, 1989a

  • Lepor H. Nonoperative management of benign prostatic hyperplasia. Journal of Urology 141: 1283–1289, 1989b

    PubMed  CAS  Google Scholar 

  • Lepor H, Knapp-Maloney G, Wozniak-Petrofsky J. The safety and efficacy of terazosin for the treatment of benign prostatic hyperplasia. International Journal of Clinical Pharmacology Therapy and Toxicology 27: 392–393, 1989c

    CAS  Google Scholar 

  • Lepor H, Rigaud G. The efficacy of transurethral resection of the prostate in men with moderate symptoms of prostatism. Journal of Urology: 533–537, 1990

    Google Scholar 

  • Levine AC, Kirschenbaum A, Kaplan P, Droller MA, Gabrilove JL. Serum prostate-antigen levels in patients with benign prostatic hypertrophy treated with leuprolide. Urology 34: 10–13, 1989

    Article  PubMed  CAS  Google Scholar 

  • Liang T, Heiss CE, Cheung AH, Reynolds GF, Rasmusson GH. 4-Azasteroidal 5α-reductase inhibitors without affinity for the androgen receptor. Journal of Biological Chemistry 259: 734–739, 1984

    PubMed  CAS  Google Scholar 

  • Liang T, Cascieri MA, Cheung AH, Reynolds GF, Rasmusson GH. Species differences in prostatic steroid 5α-reductases of rat, dog, and human. Endocrinology 117: 571–579, 1985

    Article  PubMed  CAS  Google Scholar 

  • Loughlin KR. Medical and nonmedical therapies for benign prostatic hypertrophy. Geriatrics 46: 26–31, 1991

    PubMed  CAS  Google Scholar 

  • MacLusky NJ, Naftolin F. Sexual differentiation of the central nervous system. Science 211: 1294–1303, 1981

    Article  PubMed  CAS  Google Scholar 

  • Mahler CH, Peeling WB. The normal prostate. In Altwein (Ed.) Benign prostatic hyperplasia — a diagnosis and treatment primer, pp. 7–30, Merck & Co. Inc., Rahway, New Jersey, US 1992

    Google Scholar 

  • Matzkin H, Chen J, Weisman Y, Goldray D, Pappass F, et al. Prolonged treatment with finasteride (a 5α-reductase inhibitor) does not affect bone density and metabolism. Clinical Endocrinology 37: 432–436, 1992

    Article  PubMed  CAS  Google Scholar 

  • Mauvais-Jarvis P, Kuttenn F, Baudot N. Inhibition of testosterone conversion to dihydrotestosterone in men treated percutaneously by progesterone. Journal of Clinical Endocrinology and Metabolism 38: 142–147, 1974

    Article  PubMed  CAS  Google Scholar 

  • McConnell JD, Wilson JD, George FW, Geller J, Pappas F, et al. Finasteride, an inhibitor of 5α-reductase, suppresses prostatic dihydrotestosterone in men with benign prostatic hyperplasia. Journal of Clinical Endocrinology and Metabolism 74: 505–508, 1992

    Article  PubMed  CAS  Google Scholar 

  • McConnell JD. Androgen ablation and blockade in the treatment of benign prostatic hyperplasia. Urologic Clinics of North America 17: 661–670, 1990a

    PubMed  CAS  Google Scholar 

  • McConnell JD. Medical management of benign prostatic hyperplasia with androgen suppression. Prostate 3 (Suppl.): 49–59, 1990b

    Article  PubMed  CAS  Google Scholar 

  • McConnell JD, George FW, Wilson JD, Geller J, Stoner E. The effects of low-dose finasteride (MK-906) on prostatic androgen levels in men with benign prostatic hyperplasia. Abstract 313. Journal of Urology 143: 267, 1990

    Google Scholar 

  • McNeal JE. Development and comparative anatomy of the prostate. In Grayhack et al. (Eds) Benign prostatic hyperplasia, pp. 1–9, DHEW Publ. No. (NIH) 76–1113, Washington DC, 1975

    Google Scholar 

  • McNeal JE. Origin and evolution of benign prostatic enlargement. Investigative Urology 15: 340–345, 1978

    PubMed  CAS  Google Scholar 

  • McNeal JE. Anatomy of the prostate and morphogenesis of BPH. New approaches to the study of benign prostatic hyperplasia, pp. 27–53, AR Liss Inc., New York, 1984

    Google Scholar 

  • Mebust WK, Holtgrewe HL, Cockett ATK, Peters PC, Writing Committee. Transurethral prostatectomy: immediate and postoperative complications. A cooperative study of 13 participating institutions evaluating 3 885 patients. Journal of Urology 141: 243–247, 1989

    PubMed  CAS  Google Scholar 

  • Ohtawa M, Morikawa H, Shimazaki J. Pharmacokinetics and biochemical efficacy after single and multiple oral administration of N-(2-methyl-2-propyl)-3-oxo-4-aza-5α-androst-1 -ene-17β-carboxamide, a new type of specific competitive inhibitor of testosterone 5α-reductase, in volunteers. European Journal of Drug Metabolism and Pharmacokinetics 16: 15–21, 1991

    Article  PubMed  CAS  Google Scholar 

  • Peeling WB. Diagnostic assessment of benign prostatic hyperplasia. Prostate 2 (Suppl.): 51–68, 1989

    Article  PubMed  CAS  Google Scholar 

  • Peterson RE, Imperato-McGinley J, Gautier T, Sturla E. Male pseudohermaphroditism due to steroid 5α-reductase deficiency. American Journal of Medicine 62: 170–191, 1977

    Article  PubMed  CAS  Google Scholar 

  • Rasmusson GH, Reynolds GF, Steinberg NG, Walton E, Patel GF, et al. Azasteroids: structure-activity relationships for inhibition of 5α-reductase and of androgen receptor binding. Journal of Medicinal Chemistry 29: 2298–2315, 1986

    Article  PubMed  CAS  Google Scholar 

  • Resko JA, Ellinwood WE, Pasztor LM, Buhl AE. Sex steroids in the umbilical circulation of fetal rhesus monkeys from the time of gonadal differentiation. Journal of Clinical Endocrinology and Metabolism 50: 900–905, 1980

    Article  PubMed  CAS  Google Scholar 

  • Rittmaster RS, Magor KE, Manning AP, Norman RW, Lazier CB. Differential effect of 5α-reductase inhibition and castration on androgen-regulated gene expression in rat prostate. Molecular Endocrinology 5: 1023–1029, 1991

    Article  PubMed  CAS  Google Scholar 

  • Rittmaster RS, Lemay A, Zwicker H, Capizzi TM, Winch S, et al. Effect of finasteride, a 5α-reductase inhibitor, on serum gonad-otropins in normal men. Journal of Clinical Endocrinology and Metabolism 75: 484–488, 1992

    Article  PubMed  CAS  Google Scholar 

  • Rittmaster RS, Stoner E, Thompson DL, Nance D, Lasseter KC. Effect of MK-906, a specific 5α-reductase inhibitor, on serum androgens and androgen conjugates in normal men. Journal of Andrology 10: 259–262, 1989

    PubMed  CAS  Google Scholar 

  • Robel P, Baulieu EE. Le testicule endocrine — mechanisme d’action moléculaire des hormones steroides masculines. In Mauvais-Jarvis P (Ed.) Medecine de la reproductive masculine. Flammarion Medical Sciences: p. 94, 1984

    Google Scholar 

  • Roehrborn CG, McConnell JD, Eddy DM. Outcome analysis after treatment for benign prostatic hyperplasia (BPH) by various modalities — a confidence profile analysis. Abstract. Journal of Urology 145 (Suppl.): 364A, 1991

    Google Scholar 

  • Roos NP, Wennberg JE, Malenka DJ, Fisher ES, McPherson K, et al. Mortality and reoperation after open and transurethral resection of the prostate for benign prostatic hyperplasia. New England Journal of Medicine 320; 1120–124, 1989

    Article  PubMed  CAS  Google Scholar 

  • Roy AB. The steroid 5α-reductase activity of rat liver and prostate. Biochimie 53: 1032–1040, 1971

    Article  Google Scholar 

  • Schröder FH, Altwein JE. Development of benign prostatic hyperplasia. In Altwein JE (Ed.) Benign prostatic hyperplasia: a diagnosis and treatment primer. pp. 31–50. Merck and Co. Inc. Rahway, New Jersey, US 1992

    Google Scholar 

  • Schröder FH, Blom JHM. Natural history of benign prostatic hyperplasia (BPH). Prostate 2 (Suppl.): 17–22, 1989

    Article  PubMed  Google Scholar 

  • Spencer JR, Torrado T, Sanchez RS, Vaughan Jr ED, Imperato-McGinley J. Effects of flutamide and finasteride on rat testicular descent. Endocrinology 129: 741–748, 1991

    Article  PubMed  CAS  Google Scholar 

  • Stoner E, Finasteride Study Group. The clinical effects of a 5α-reductase inhibitor, finasteride, on benign prostatic hyperplasia. Journal of Urology 147: 1298–1302, 1992

    PubMed  CAS  Google Scholar 

  • Tempany CMC, Partin AW, Zerhouni EA, Zinreich SJ, Walsh PC. The influence of finasteride on the volume of the peripheral and periurethral zones of the prostate in men with benign prostatic hyperplasia. Prostate 22: 39–42, 1993

    Article  PubMed  CAS  Google Scholar 

  • Tenover JS, Zeitner ME, Plymate SR. Effects of 24-week administration of a 5-alpha reductase inhibitor (MK-906) on serum levels of testosterone (T), free T, and gonadotrophins in men. Abstract 583. 71st Annual Meeting of the Endocrine Society, Seattle, Washington, 1989.

    Google Scholar 

  • Thigpen AE, Davis DL, Milatovich A, Mendonca BB, Imperato-McGinley J, et al. Molecular genetics of steroid 5α-reductase 2 deficiency. Journal of Clinical Investigation 90: 799–809, 1992

    Article  PubMed  CAS  Google Scholar 

  • The MK-906 (Finasteride) Study Group. One-year experience in the treatment of benign prostatic hyperplasia with finasteride. Journal of Andrology 12: 372–375, 1991

    Google Scholar 

  • The Proscar Study Group. Treatment of benign prostatic hyperplasia (BPH) with proscar, a 5α-reductase inhibitor. Abstract. Workshop Conference on Androgen Therapy, Marco Island, Florida, January, 1990

  • Vermeulen A, Giagulli VA, De Schepper P, Buntinx A, Stoner E. Hormonal effects of an orally active 4-azasteroid inhibitor of 5α-reductase in humans. Prostate 14: 45–53, 1989

    Article  PubMed  CAS  Google Scholar 

  • Watanabe H. Natural history of benign prostatic hypertrophy. Ultrasound in Medicine and Biology 12: 567–571, 1986

    Article  PubMed  CAS  Google Scholar 

  • Wilde MI, Fitton A, McTavish D. Alfuzosin: a review of its pharmacodynamic and pharmacokinetic properties and therapeutic potential in benign prostatic hyperplasia. Drugs 45: 410–429, 1993a

    Article  PubMed  CAS  Google Scholar 

  • Wilde MI, Fitton A, Sorkin EM. Terazosin: a review of its phar-macodynamic and pharmacokinetic properties and therapeutic potential in benign prostatic hyperplasia. Drugs & Aging 3: 258–277, 1993b

    Article  CAS  Google Scholar 

  • Wilson JD, Griffin JE, George FW. Sexual differentiation: early hormone synthesis and action. Biology of Reproduction 22: 9–17, 1980

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Additional information

Various sections of the manuscript reviewed by: U. Dunzendorfer, Maingau Krankenhaus, Frankfurt, Federal Republic of Germany; P. Ekman, Department of Urology, Karolinska Hospital, Stockholm, Sweden; J. Geller, Department of Medical Education, Mercy Hospital and Medical Center, San Diego, California, USA; F.W. George, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; J.C. Gingell, Department of Urology, Southmead Hospital, Westbury-on-Trym, Bristol, England; J.W. Goldzieher, Department of Obstetrics and Gynecology, Baylor College of Medicine, San Antonio, Texas, USA; D. Horii, Kashima Laboratory, Mitsubishi-Kasei Institute of Toxicological and Environmental Sciences, Ibaraki, Japan; H. Matzkin, Department of Urology, Tel Aviv-Elias Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; J. Nacey, Department of Surgery, Wellington School of Medicine, Wellington, New Zealand; W.B. Peeling, St Woolos Hospital, Newport, Gwent, Wales; R.S. Rittmaster, Department of Medicine and Physiology, Dalhousie University, Halifax, Nova Scotia, Canada; P.J. De Schepper, Department of Pharmacology and Clinical Pharmacology, School of Medicine, University of Leuven, Leuven, Belgium.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Peters, D.H., Sorkin, E.M. Finasteride. Drugs 46, 177–208 (1993). https://doi.org/10.2165/00003495-199346010-00010

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003495-199346010-00010

Keywords

Navigation