• No results found

Antiandrogens in the Treatment of Prostate Cancer

N/A
N/A
Protected

Academic year: 2021

Share "Antiandrogens in the Treatment of Prostate Cancer"

Copied!
9
0
0
Show more ( Page)

Full text

(1)

Review – Prostate Cancer

Antiandrogens in the Treatment of Prostate Cancer

Manfred P. Wirth

*

, Oliver W. Hakenberg, Michael Froehner

Department of Urology, University Hospital ‘‘Carl Gustav Carus’’, Technical University of Dresden, Fetscherstrasse 74, D-01307 Dresden, Germany

1. Introduction

Since the discovery of the beneficial effect of androgen deprivation by orchiectomy on prostate cancer disease, the stage-corrected survival rates of prostate cancer patients treated by androgen

abla-tion have not been further improved [1,2]. Regard-less of the obvious symptomatic benefit in advanced disease, definite evidence for any prolongation of survival by endocrine treatment not combined with curative local treatment (radical prostatectomy or external beam radiotherapy) is still lacking. Early a v a i l a b l e a t w w w . s c i e n c e d i r e c t . c o m

j o u r n a l h o m e p a g e : w w w . e u r o p e a n u r o l o g y . c o m

Article info

Article history:

Accepted August 22, 2006 Published online ahead of print on September 11, 2006 Keywords: Prostate cancer Hormonal treatment Antiandrogens Bicalutamide Flutamide Nilutamide Cyproterone acetate Abstract

Objectives: To give an overview on the contemporary role of

antiandro-gens in prostate cancer treatment.

Methods: A review of the literature was performed concerning

pharma-cologic properties, possible indications and side effects of antiandrogens in the treatment of early and advanced prostate cancer.

Results: One steroidal and three non-steroidal antiandrogens are in

common use for the treatment of prostate cancer. Monotherapy with non-steroidal antiandrogens may prevent osteoporosis, loss of muscu-lature and may preserve sexual activity in a proportion of patients and therefore has advantages in quality of life compared to castration. In patients with localized disease managed by watchful waiting or in an adjuvant setting, there are no studies showing an advantage in early versus delayed treatment with antiandrogens regarding clinical progres-sion. In locally advanced non-metastatic prostate cancer, antiandrogen monotherapy seems to be an alternative to castration treatment if treatment is required. In patients with metastatic disease and a high tumor burden, antiandrogen monotherapy is inferior to castration. In advanced disease, combined androgen blockade can provide a small survival advantage, which, however, has to be balanced against increased side effects and costs.

Conclusions: Antiandrogens are a treatment option in some patients

with prostate cancer. However, it has to be taken into account that the hormonal effect is inferior to castration.

#2006 European Association of Urology. Published by Elsevier B.V. All rights reserved.

* Corresponding author. Tel. +49 351 4582447; Fax: +49 351 4584333. E-mail address:Manfred.Wirth@uniklinikum-dresden.de(M.P. Wirth).

(2)

studies demonstrated a survival advantage, but these studies do not meet contemporary require-ments for comparative clinical trials any more and can therefore not be regarded as conclusive[1].

Surgical or chemical castration is accompanied by adverse effects and results in a decrease in quality of life if undertaken early in the course of the disease (Table 1). Most patients with metastatic prostate cancer will develop hormone-refractory disease within 12 to 18 months. Longer response periods may be expected when hormonal treatment is started at earlier stages[3,4]. Long-term androgen depriva-tion, however, increases the risk of adverse effects such as osteoporosis and pathologic fractures[5].

Prostate cancer today is diagnosed with increas-ing frequency in physically and sexually active younger men and although prostate cancer is increasingly detected in localized and potentially

curable tumor stages[6], a considerable proportion of patients will experience treatment failure and will become candidates for hormonal treatment [1]. Therefore, considerable effort has been undertaken to assess the effect of early, adjuvant or neoadjuvant hormonal manipulations and to decrease the side effects of this treatment modality [3]. This article summarizes the current role of antiandrogens in the treatment of prostate cancer.

2. Properties and side effects of currently available antiandrogens

Nonsteroidal antiandrogens (bicalutamide, fluta-mide, nilutamide) competitively inhibit the binding of androgens to the androgen receptor. As a consequence, the serum testosterone levels are Table 1 – Pros and cons of different modalities of hormonal treatment for prostate cancer

Treatment modality Pro Con

Orchiectomy low long-term costs high short-term costs definitive androgen ablation surgical complications possible

no problems with compliance psychological side effects, cognitive decline long term side effects (anaemia and osteoporosis) loss of libido and potency

loss of physical activity loss of muscle mass, weight gain hot flushes

irreversible measure

LHRH analogues preservation of body integrity side effects comparable with surgical castration temporary treatment possible ‘‘flare-up’’ phenomenon

treatment reversible high long-term costs regular contact with physician

LHRH antagonists no ‘‘flare-up’’ phenomenon anaphylaxis Nonsteroidal antiandrogens temporary adjuvant or

intermittent therapy possible

high long-term costs (bicalutamide)

preservation of sexual function possible gynecomastia and breast pain frequent oral applicability, partially once daily hepatotoxicity and diarrhoea

(especially flutamide) favourable tolerability profile non-compliance

osteoporosis probably less frequent less effective in high tumor burden (metastatic disease)

step-up treatment possible limited approval treatment reversible

Steroidal antiandrogens temporary adjuvant or intermittent therapy possible

side effects comparable with surgical castration

oral applicability inferior to castration in terms of time to progression

step-up treatment possible hepatotoxicity with long term use treatment reversible limited approval

Maximal (combined) androgen blockade

blockade of both testicular and adrenal androgens

combined side effects of castration and oral antiandrogens

possible small survival advantage high long-term costs treatment reversible

Estrogens treatment reversible cardiovascular toxicity low costs

(3)

not suppressed and may even increase. In contrast, the only available steroidal antiandrogen cyproter-one acetate in addition to antiandrogenic also has progestational and antigonadotrophic properties. Its application via a feedback suppression of pituitary LHRH release thus leads to a reduction of serum testosterone levels. Cyproterone aceteate medica-tion in contrast to the use of non-steroidal anti-androgens, thus results in the suppression of libido and erectile function and causes side effects similar to castration[7].

Bicalutamide is the most extensively studied nonsteroidal antiandrogen [7]. Compared to LHRH agonist treatment bicalutamide monotherapy results in reduced fat accumulation, increased bone density and fewer bothersome adverse effects both in animal studies [8] as well as clinically [9,10]. However, gynecomastia and breast pain are frequent side effects of bicalutamide monotherapy and occur in 70–80% of patients [11–13]. This can partially be prevented by local radiotherapy or tamoxifen treat-ment. In randomised studies, tamoxifen prevented gynecomastia effectively (odds ratio 0.1,p= 0.0009), whereas prophylactic breast irradiation reduced the incidence of gynecomastia by about 50% (odds ratio 0.51,p= 0.008),[14,15]. Tamoxifen did not add mean-ingful toxicity to bicalutamide monotherapy[15]. It is, however, still unclear what effect the estrogen-antagonist tamoxifen has on prostate cancer. There-fore, the routine use of tamoxifen to prevent gyne-comastia cannot be recommended at present while the use of prophylactic or therapeutic breast irradia-tion is established. After the development of bicalu-tamide-induced gynecomastia and/or breast pain, about one third of patients may be expected to benefit from breast irradiation (two fractions of 6 Gy)[16].

Another problem of non-steroidal antiandrogens is the interaction with other drugs due to inter-ference with plasma protein binding. The use of antiandrogens in patients taking drugs with high plasma protein binding such as warfarin, phenytoin or theophylline will increase the free drug serum concentration of these substances which may result in increased effects or side effects of these drugs[17]. Table 2summarizes the properties and side effects of the four antiandrogens in clinical use. There are relatively few data available for nilutamide. Although direct comparisons are not available, compared to the other antiandrogens bicalutamide seems to have some advantages in terms of the side effect profile. Hepatotoxicity, the most serious side effect of nilutamide, flutamide and cyproterone acetate is relatively uncommon with bicalutamide. This relative advantage of bicalutamide is partially outweighed by the high rate of gynecomastia. Taken together, the side effect profile of non-steroidal antiandrogens compares favourably with castra-tion, nevertheless, uncritical long-term use is inappropriate and harbours the risk of adverse events and even decreased survival [18]. Intermit-tent LHRH analogue treatment is an alternative to antiandrogens to reduce side effects caused by castration. Osteoporosis can also be prevented by bisphosphonate treatment.

3. Antiandrogen monotherapy 3.1. Locally advanced or metastatic disease

In two combined randomised trials involving 480 patients with locally advanced non-metastatic Table 2 – Properties and side effects of the currently available antiandrogens

Bicalutamide Flutamide Nilutamide Cyproterone acetate

Half-life: 7 days 5–6 hours 2 days 30–40 hours Dosage: 150 mg daily* 3250 mg daily 3100 mg dailyy 3100 mg daily

Side effects: hepatotoxicity (rarely)

diarrhoea visual disturbances (delayed adaptation to darkness, 33%)z

lower testosterone levels

hot flushes

hepatotoxicity (more frequent than other antiandrogens, may be fatal) alcohol intolerance (20%)z loss of libido breast tenderness impotence (20%) nausea impotence (80%) gynaecomastia (70%) gynaecomastia hepatotoxicity (rarely, may be fatal)

rarely gynaecomastia

nausea

interstitial pneumonitis8

cardiovascular toxicity (4–40%§)

hepatotoxicity (may be fatal) diarrhoea

impotence (20%)

impotence (50%),

* Monotherapy, 50 mg daily when combined with LHRH agonists. y Not licenced for monotherapy.

z Not seen with other non-steroidal antiandrogens.

§ Hypotension, tachycardia, heart failure, syncope, myocardial infarct, haemorrhage, cerebrovascular accident, cardiovascular disorder, retinal

vascular disorder, embolus, pulmonary embolism, superficial and deep thrombophlebitis, thrombosis, retinal vein thrombosis, phlebitis, vascular headache, shock.

(4)

disease (clinical stages T3-T4N0M0), bicalutamide monotherapy (150 mg daily) was compared with castration. After a median follow-up of 6.3 years, there was no detectable difference concerning time to progression or overall survival[19]. This study, however, was not able to demonstrate equivalence between both treatments since the upper 1-sided 95% confidence interval was greater than the value of 1.25 needed to reject the hypothesis that bicalutamide was at least 25% inferior to castration [19]. Thus, there was no unequivocal proof in this trial that bicalutamide monotherapy was equally effective as castration treatment. Advantages for

bicalutamide monotherapy, however, were

observed concerning sexual interest and physical capacity although detailed data on sexual potency were not reported [19]. Other studies suggest that approximately one third of patients receiving 150 mg bicalutamide daily maintain sexual func-tion during long term treatment[20].

In a prospective randomised study including 220 patients with stage C or D prostate cancer, bicalu-tamide monotherapy (150 mg daily) was compared to combined androgen blockade. Patients in the bicalutamide arm underwent castration at disease progression. Again, there were no detectable differ-ences concerning disease-specific or overall survival [21] while subgroup analysis suggested an unex-plained increased overall mortality in patients with high-grade disease treated with bicalutamide only[21].

Tyrrell and co-workers compared bicalutamide monotherapy (150 mg daily) with castration as treatment for locally advanced non-metastatic or metastatic prostate cancer (n= 1453). Bicalutamide monotherapy was as effective as castration in non-metastatic patients, but there was a small survival advantage for castration in the M1 subgroup. This difference was perhaps partially outweighed by a better tolerability profile and a higher quality of life in patients treated with bicalutamide monotherapy [22].

Thus, after adequate patient information, bica-lutamide monotherapy is an option for younger and sexually active patients with locally advanced disease and in selected patients with metastatic prostate cancer. In men with a high disease burden (PSA values >400 ng/ml), castration definitely remains superior to antiandrogen monotherapy [23,24].

Schro¨der and co-workers reported the final results of a randomised EORTC trial comparing flutamide versus cyproterone acetate monothera-pies in men with metastatic prostate cancer and favourable prognostic factors [25]. They found no

significant difference between the two treatment arms concerning time to progression, disease-specific or overall survival. The side effect analysis favoured cyproterone acetate regarding gynecomas-tia, diarrhoea and nausea[25]. In a phase III study of metastatic prostate cancer treatment with com-bined androgen blockade (goserelin acetate plus cyproterone acetate) versus goserelin acetate alone or cyproterone acetate alone with 525 patients, monotherapy with the antiandrogen cyproterone acetate was inferior to LHRH monotherapy in delaying time to progression (difference in the median delay of time to progression was 3 months,

p<0.02) [26]. Current recommendations of the American Society of Clinical Oncology therefore do not recommend the use of cyproterone acetate for monotherapy[27,28].

The supplementation of antiandrogen monother-apy with a 5a-reductase inhibitors is possible. It has been suggested that such a combination treatment may increase the antiandrogenic activity with low additional toxicity. About one third of patients treated in this way may be expected to maintain spontaneous erections [29]. No data on the long-term efficacy of such treatment regimens has been reported and this combination increases treatment costs considerably.

3.2. Adjuvant and primary antiandrogenic treatment in early prostate cancer

Again, bicalutamide is the most extensively studied drug for this application. In the ongoing Early Prostate Cancer Program (EPC) bicalutamide is being evaluated as an adjuvant treatment for early prostate cancer (stages T1b-4N0-1M0), whereby antiandrogenic monotherapy together with ‘watch-ful waiting’ as a primary treatment strategy is also defined as ‘adjuvant’. The program consists of three double-blind, parallel-group, placebo-controlled trials (North America, (trial 23, n= 3292), Europe/ rest of world (trial 24,n= 3603), Scandinavia (trial 25,

n= 1218)[11,30]. While in the North American trial, all patients underwent curative treatment prior to study entry, watchful waiting was allowed as ‘primary treatment’ besides radical prostatectomy or radiotherapy in trials 24 and 25 and in the Scandinavian trial comprised 82% of patients [11,30]. The study treatment was given for two years or until disease progression in trial 23 and until disease progression in trials 24 and 25. A maximum treatment duration of five years was recommended for patients receiving treatment adjuvant to curative measures [18,31]. Because of these differences in treatment schedules and in

(5)

patient selection, the results of a combined analysis of the three studies have to be interpreted with caution. Time to objective clinical progression and survival were the primary study end points, time to treatment failure, PSA progression and toxicity as secondary end points. Side effects (gynecomastia/ breast pain) were the most common reason for discontinuation of treatment in the bicalutamide group and disease progression in the placebo group [11]. At the time of the most recently published analysis, at a median follow-up of 7.4 years, there was no benefit in progression-free survival for the bicalutamide group in the entire study population [18]. There was even a trend towards decreased overall survival for localized prostate cancer under watchful waiting when treated with bicalutamide (hazard ratio 1.16, 95% confidence interval 0.99– 1.37,p= 0.07). In all patients with locally advanced disease, however, bicalutamide significantly improved progression-free survival irrespective of the primary treatment applied. Overall survival in patients with locally advanced disease was improved in the bicalutamide group only in patients treated by radiotherapy (hazard ratio 0.65, 95% confidence interval 0.44–0.95, p= 0.03) and these patients had a lower risk of prostate cancer-related mortality. In the bicalutamide group of patients with locally advanced disease under watchful waiting overall survival was not significantly improved (hazard ratio 0.81, 95% confidence inter-val 0.66–1.01,p= 0.06) nor was any survival differ-ence seen in the radical prostatectomy subgroup [18].

Overall, the currently available data from the ongoing EPC studies suggest that early or adjuvant hormonal therapy is not beneficial in patients with localized disease while it seems of proven benefit in prolonging progression-free survival in patients with locally advanced disease irrespective of the primary treatment and in prolonging overall survi-val in patients undergoing primary radiotherapy [18,31].

4. Combined androgen blockade

The superiority of combined (‘‘complete’’) androgen blockade (i.e. the combination of an antiandrogen with orchiectomy or medical castration) as the primary treatment for advanced prostate cancer has not been proven conclusively up to date despite a large number of randomised trials[32,33]. A meta-analysis of 20 trials found no differences between combined androgen blockade and castration when 2-year overall survival was evaluated. Based on the

10 trials with available 5-year survival data, this same meta-analysis, however, did show a signifi-cant survival advantage for combined androgen blockade[32]. No evidence is available supporting a greater benefit of combined treatment in patients with favorable prognostic factors.

Combined androgen blockade using a non-ste-roidal antiandrogen tends to be superior to those using cyproterone acetate in delaying progression [26,32,33]. The limited advantage of combined treatment over monotherapy must, however, be balanced against higher side effects and costs. The additional costs for one quality-adjusted life-year gained by using combined androgen blockade over orchiectomy alone has been estimated to be US$1 million [Prostate Cancer Trialists’ Collaborative Group 2000]. Combined androgen blockade is an option for patients with advanced or metastatic prostate cancer. It is, however, not the standard form of androgen ablative therapy for primary application[33].

5. Adjuvant treatment after radiotherapy or radical prostatectomy

A great body of data from prospective randomised trials supports the use of adjuvant hormonal treatment after external beam radiotherapy. High-risk patients with locally advanced disease or positive lymph nodes seem to benefit from immediate androgen deprivation after external beam radiotherapy whereas in earlier stages the differences tend to diminish [27]. The beneficial effect of adjuvant hormonal therapy in the external beam radiotherapy setting has been attributed to an elimination of occult metastases and to a potential additive effect by induction of apoptosis [34]. It is, however, still unknown to which degree the observed survival advantages may be attributed to hormonal therapy alone. Data from the Bicalutamide Early Prostate Cancer Program suggest that bicalutamide monotherapy may possibly be similarly effective as castration in the adjuvant setting after radiotherapy [18], ran-domised comparisons are, however, not available up to date.

Conclusive evidence from randomised studies on the efficacy of adjuvant endocrine treatment and particularly of adjuvant antiandrogen treatment after radical prostatectomy is still lacking. In patients with positive lymph nodes, a small random-ized study demonstrated a survival advantage for immediate hormonal therapy (castration) versus treatment at metastatic or symptomatic progression

(6)

[35]. In a multicentric randomised trial of 352 patients with locally advanced, lymph node-nega-tive prostate cancer after radical prostatectomy, adjuvant flutamide was compared to no adjuvant treatment. Tumor progression was significantly delayed in the flutamide arm (p= 0.0041) but there was no difference in overall survival after a median follow-up of 6.1 years (p= 0.92)[36]. Similarly, the EPC studies on bicalutamide have so far only shown a delay in PSA-defined progression without any effect on overall survival in patients after radical prostatectomy (see above).

6. Neoadjuvant treatment prior to radiotherapy or radical prostatectomy

Prior to radical prostatectomy, neoadjuvant hormo-nal treatment is not recommended for routine clinical use[23]. In patients with locally advanced prostate cancer selected for external beam radiotherapy, however, in one radomised trial, neoadjuvant com-bined androgen blockade (flutamide and goserelin) significantly improved local control, progression rate and overall survival in the Gleason score 2–6 subgroup [37]. Comparable evidence is not available for anti-androgenic monotherapy. Considering the prostate volume reduction, bicalutamide monotherapy seems to be less effective than LHRH analogues[38].

7. Step-up hormonal treatment

Because of the uncertainties surrounding the tim-ing, side effects and costs of hormonal treatment, alternative treatments especially in patients with a low tumor burden have been considered. The so-called step-up treatment (stepwise escalation of androgen withdrawal, for instance starting with an antiandrogen and later adding a 5-a-reductase inhibitor) is frequently used in clinical practice. However, hardly any studies investigating this treatment option have been published. Oh and co-workers treated patients with PSA relapse after curative treatment or newly diagnosed metastatic disease initially with flutamide and finasteride. The authors suggested that such a strategy is feasible and effective in terms of PSA control. All patients who failed under the initial oral regimen experi-enced more than 50% decline in PSA when even-tually androgen ablation treatment was undertaken [29]. Randomized studies comparing step-up hor-monal treatment with early or deferred con-ventional androgen deprivation might be of considerable clinical interest.

8. Prevention of ‘‘flare-up’’ phenomenon in patients receiving LHRH agonists

When given for the first time, long-acting LHRH agonists cause a transient testosterone increase by stimulating pituitary LHRH receptors. This so called ‘‘flare-up’’ phenomenon may cause fatal complica-tions in patients with far advanced disease. Simul-taneous antiandrogen application may decrease the incidence of initial disease progression. It has been recommended to start such treatment at the same day as the first LHRH analogue depot injection and maintain it for a two week period. In high risk patients (impending spinal cord compression), however, surgical castration should be preferred [23].

9. Antiandrogen withdrawal syndrome and secondary hormonal manipulations

Failure after initial hormonal treatment does not necessarily mean treatment refractory disease progression. Secondary hormonal manipulations are a safe and effective but short-term option in patients with failure after primary hormonal therapy [4]. When an antiandrogen is part of the treatment regimen, discontinuation may result in the so-called antiandrogen withdrawal syndrome, described for the first time by Kelly and Scher[39]. The incidence of this effect has been reported to be 15 to 20% and it lasts for five months on average [4]. Replacement of an initially given antiandrogen by an alternative substance may also result in a PSA response [40]. Mutations in the androgen receptor gene are supposed to account for this phenomenon by enabling the antiandrogens to act as receptor agonists [41]. Similar observations were reported after replacement of bicalutamide or flutamide as part of a failing combined andro-gen blockade by nilutamide [42]. After initial antiandrogen monotherapy, orchiectomy at the time of failure of initial treatment may lead to a PSA response and to symptomatic improvement [43]. Second-line treatment with bicalutamide has been shown to improve symptoms and decrease pain in patients without prior antiandrogen therapy [4]. A 50% PSA decrease has been described after second line treatment with non-steroidal antiandrogens in 14–50% of cases [4]. Responders to second-line hormonal treatment may be expected to survive significantly longer than non-responders [40]. Overall, however, it is still doubtful whether secondary hormonal manipulations do actually improve survival [4].

(7)

However, the suppression of testicular androgens should be continued when the disease is hormone refractory, since a withdrawal of treatment may decrease survival [4].

10. Conclusions

Antiandrogens are an established treatment option in patients with prostate cancer either as mono-therapy or in combination with LHRH analogues. In patients with a limited tumor burden, non-ste-roidal antiandrogen monotherapy seems as effec-tive as castration. Antiandrogens which preserve testosterone levels can also preserve libido and sexual function in previously sexually active patients. Thus some side effects of castration may be avoided by antiandrogen monotherapy, but frequent gynecomastia (bicalutamide) and the risk of hepatotoxicity (cyproterone, nilutamide, flutamide) and a hitherto unexplained trend towards increased mortality when given to patients with localized prostate cancer prohibit their uncri-tical use. These risks have to be individually balanced against improved quality of life compared to castration treatment. Antiandrogens may be used as part of combined androgen blockade regimens. PSA progression under combined andro-gen blockade may respond to discontinuation or change of the antiandrogen.

Conflict of interest

Professor M. P. Wirth is principal investigator of the ‘‘Bicalutamide Early Prostate Cancer Program’’ which is supported by AstraZeneca.

References

[1] Walsh PC, deWeese TL, Eisenberger MA. A structured debate: immediate versus deferred androgen suppression in prostate cancer – evidence for deferred treatment. J Urol 2001;166:508–15.

[2] Huggins C, Hodges CV. Studies on prostatic cancer: I. The effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. Cancer Res 1941;1:293–7.

[3] Ryan CJ, Small EJ. Early versus delayed androgen depriva-tion for prostate cancer: new fuel for an old debate. J Clin Oncol 2005;23:8225–31.

[4] Lam JS, Leppert JT, Vemulapalli SN, Shvarts O, Belldegrun AS. Secondary hormonal therapy for advanced prostate cancer. J Urol 2006;175:27–34.

[5] Shahinian VB, Kuo YF, Freeman JL, Goodwin JS. Risk of fracture after androgen deprivation for prostate cancer. N Engl J Med 2005;352:154–64.

[6] Routh JC, Leibovich BC. Adenocarcinoma of the prostate: epidemiological trends, screening, diagnosis, and surgical management of localized disease. Mayo Clin Proc 2005;80:899–907.

[7] Anderson J. The role of antiandrogen monotherapy in the treatment of prostate cancer. BJU Int 2003;91:455–61. [8] Lefort M, Curiel MD, Carrascal MT, Mendez-Davila C, De la

Piedra C. Comparative effects of bicalutamide versus orchidectomy on bone mineral density, bone remodelling and bone biomechanics in healthy rats. Urol Int 2005; 74:301–7.

[9] Smith MR, Goode M, Zietman AL, McGovern FJ, Lee H, Finkelstein JS. Bicalutamide monotherapy versus leupro-lide monotherapy for prostate cancer: effects on bone mineral density and body composition. J Clin Oncol 2004;22:2546–53.

[10] Sieber PR, Keiller DL, Kahnoski RJ, Gallo J, McFadden S. Bicalutamide 150 mg maintains bone mineral density during monotherapy for localized or locally advanced prostate cancer. J Urol 2004;171:2272–6.

[11] Wirth M, Tyrrell C, Delaere K, Sanchez-Chapado M, Ramon J, Wallace DM, et al. Bicalutamide (‘Casodex’) 150 mg in addition to standard care in patients with nonmetastatic prostate cancer: updated results from a randomised double-blind phase III study (median follow-up 5.1 y) in the early prostate cancer programme. Prostate Cancer Prostatic Dis 2005;8:194–200.

[12] Abrahamsson PA, Anderson J, Boccon-Gibod L, Schulman C, Studer UE, Wirth M. Risks and benefits of hormonal manipulation as monotherapy or adjuvant treatment in localised prostate cancer. Eur Urol 2005;48:900–5. [13] Tyrrell CJ, Payne H, See WA, McLeod DG, Wirth MP,

Iversen P, et al. Bicalutamide (‘Casodex’) 150 mg as adju-vant to radiotherapy in patients with localised or locally advanced prostate cancer: results from the randomised Early Prostate Cancer Programme. Radiother Oncol 2005;76:4–10.

[14] Perdona S, Autorino R, De Placido S, D’Armiento M, Gallo A, Damiano R, et al. Efficacy of tamoxifen and radio-therapy for prevention and treatment of gynaecomastia and breast pain caused by bicalutamide in prostate can-cer: a randomised controlled trial. Lancet Oncol 2005;6: 295–300.

[15] Boccardo F, Rubagotti A, Battaglia M, Di Tonno P, Selvaggi FP, Conti G, et al. Evaluation of tamoxifen and anastrozole in the prevention of gynecomastia and breast pain induced by bicalutamide monotherapy of prostate cancer. J Clin Oncol 2005;23:808–15.

[16] Van Poppel H, Tyrrell CJ, Haustermans K, Van Cangh P, Keuppens F, Colombeau P, et al. Efficacy and tolerability of radiotherapy as treatment for bicalutamide-induced gynaecomastia and breast pain in prostate cancer. Eur Urol 2005;47:587–92.

[17] Encyclopedia of cancer. Encyclopedia of Cancer: Antian-drogens: eNotes.com LLC (http://www.enotes.com/ cancer-encyclopedia), 2006.

(8)

[18] McLeod DG, Iversen P, See WA, Morris T, Armstrong J, Wirth MP, et al. Bicalutamide 150 mg plus standard care vs standard care alone for early prostate cancer. BJU Int 2006;97:247–54.

[19] Iversen P, Tyrrell CJ, Kaisary AV, Anderson JB, van Poppel H, Tammela TLJ, et al. Bicalutamide monotherapy com-pared with castration in patients with nonmetastatic locally advanced prostate cancer: 6.3 years of followup. J Urol 2000;164:1579–82.

[20] Iversen P. Bicalutamide monotherapy for early state pros-tate cancer. J Urol 2003;170:S48–54.

[21] Boccardo F, Barichello M, Battaglia M, Carmignani G, Comeri G, Ferraris V, et al. Bicalutamide monotherapy versus flutamide plus goserelin in prostate cancer: updated results of a multicentric trial. Eur Urol 2002;42:481–90. [22] Tyrrell CJ, Kaisary AV, Iversen P, Anderson JB, Baert L,

Tammela T, et al. A randomised comparison of ‘Casodex’ (bicalutamide) 150 mg monotherapy versus castration in the treatment of metastatic and locally advanced prostate cancer. Eur Urol 1998;33:447–56.

[23] Aus G, Abbou CC, Heidenreich A, Schmid HP, van Poppel H, Wolff J, Zattoni F. EAU guidelines on prostate cancer. Eur Urol 2005;48:546–51.

[24] Kaisary AV, Iversen P, Tyrrell CJ, Carroll K, Morris T. Is there a role for antiandrogen monotherapy in patients with metastatic prostate cancer? Prostate Cancer Pro-static Dis 1999;4:196–203.

[25] Schro¨der FH, Whelan P, de Reijke TM, Kurth KH, Pavone-Macaluso M, Mattelaer J, et al. Metastatic prostate cancer treated by flutamide versus cyproterone acetate. Final analysis of the ‘‘European Organization for Research and Treatment of Cancer’’ (EORTC) Protocol 30892. Eur Urol 2004;45:457–64.

[26] Thorpe SC, Azmatullah S, Fellows GJ, Gingell JC, O’Boyle PJ. A prospective randomized study to compare goserelin acetate (Zoladex) versus cyproterone acetate (Cyprostat) versus a combination of the two in the treatment of metastatic prostatic carcinoma. Eur Urol 1996;29:47–54. [27] Loblaw DA, Mendelson DS, Talcott JA, Virgo KS,

Somer-field MR, Ben-Josef E, et al. American Society of Clinical Oncology recommendations for the initial hormonal management of androgen-sensitive metastatic, recur-rent, or progressive prostate cancer. J Clin Oncol 2004; 22:2927–41.

[28] Sharifi N, Gulley JL, Dahut WL. Androgen deprivation therapy for prostate cancer. JAMA 2005;294:238–44. [29] Oh WK, Manola J, Bittmann L, Brufsky A, Kaplan ID, Smith

MR, et al. Finasteride and flutamide therapy in patients with advanced prostate cancer: response to subsequent castration and long-term follow-up. Urology 2003;62: 99–104.

[30] Wirth M, Tyrrell C, Wallace M, Delaere KP, Sanchez-Cha-pado M, Ramon J, et al. Bicalutamide (Casodex) 150 mg as immediate therapy in patients with localized or locally advanced prostate cancer significantly reduces the risk of disease progression. Urology 2001;58:146–51.

[31] Wirth MP, See WA, McLeod DG, Iversen P, Morris T, Carroll K, et al. Bicalutamide 150 mg in addition to standard care

in patients with localized or locally advanced prostate cancer: results from the second analysis of the early prostate cancer program at median followup of 5.4 years. J Urol 2004;172:1865–70.

[32] Samson DJ, Seidenfeld J, Schmitt B, Hasselblad V, Albert-sen PC, Bennett CL, et al. Systematic review and meta-analysis of monotherapy compared with combined androgen blockade for patients with advanced prostate carcinoma. Cancer Res 2002;95:361–76.

[33] Prostate Cancer Trialists’ Collaborative Group. Maximum androgen blockade in advanced prostate cancer: an over-view of the randomised trials. Lancet Oncol 2000;355: 1491–8.

[34] Bolla M, Collette L, Blank L, Warde P, Dubois JB, Mirimanoff RO, et al. Long-term results with immediate androgen suppression and external irradiation in patients with locally advanced prostate cancer (an EORTC study): a phase III randomised trial. Lancet 2002;360:103–6.

[35] Messing EM, Manola J, Sarosdy M, Wilding G, Crawford ED, Trump D. Immediate hormonal therapy compared with observation after radical prostatectomy and pelvic lym-phadenectomy in men with node-positive prostate can-cer. New Engl J Med 1999;341:1781–8.

[36] Wirth MP, Weissbach L, Marx FJ, Heckl W, Jellinghaus W, Riedmiller H, et al. Prospective randomized trial com-paring flutamide as adjuvant treatment versus observa-tion after radical prostatectomy for locally advanced, lymph node-negative prostate cancer. Eur Urol 2004; 45:267–70.

[37] Pilepich MV, Winter K, John MJ, Mesic JB, Sause W, Rubin P, et al. Phase III radiation therapy oncology group (RTOG) trial 86-10 of androgen deprivation adjuvant to definitive radiotherapy in locally advanced carcinoma of the pros-tate. Int J Radiat Oncol Biol Phys 2001;50:1243–52. [38] Henderson A, Langley SE, Laing RW. Is bicalutamide

equivalent to goserelin for prostate volume reduction before radiation therapy? A prospective, observational study. Clin Oncol (R Coll Radiol) 2003;15:316–7.

[39] Kelly WK, Scher HI. Prostate specific antigen decline after antiandrogen withdrawal: the flutamide withdrawal syn-drome. J Urol 1993;149:607–9.

[40] Kojima S, Suzuki H, Akakura K, Shimbo M, Ichikawa T, Ito H. Alternative antiandrogens to treat prostate cancer relapse after initial hormone therapy. J Urol 2004;171: 679–83.

[41] Yoshida T, Kinoshita H, Segawa T, Nakamura E, Inoue T, Shimizu Y, et al. Antiandrogen bicalutamide promotes tumor growth in a novel androgen-dependent prostate cancer xenograft model derived from a bicalutamide-treated patient. Cancer Res 2005;65:9611–6.

[42] Nakabayashi M, Regan MM, Lifsey D, Kantoff PW, Taplin ME, Sartor O, et al. Efficacy of nilutamide as secondary hormonal therapy in androgen-independent prostate cancer. BJU Int 2005;96:783–6.

[43] Tan A, Tuckey J, Rice M. Orchidectomy following failure of antiandrogen monotherapy in patients with metastatic prostate cancer. Eur Urol 2001;40:130–3.

(9)

Editorial Comment

Steven P. Balk, Glenn J. Bubley, Beth Israel Deaconess Medical Center,

Harvard Medical School, Boston, MA, United States sbalk@bidmc.harvard.edu

Mary-Ellen Taplin,Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States

This article is a timely review of androgen recep-tor antagonist (antiandrogen) clinical trials and the current role of these drugs in prostate cancer treat-ment. The nonsteroidal antiandrogens were widely used in the late 1970s and 1980s in combination with orchiectomy or luteinising hormone-releasing hormone (LHRH) agonist therapies (combined androgen blockade) to prevent androgen receptor activation by residual adrenal androgens, but ran-domised trials showed only a small survival advan-tage with increased toxicity. These results led to a marked decline in the initial use of antiandrogen combination therapies and the conclusion that complete androgen receptor blockade is not more effective than monotherapy. However, it now appears clear that the androgen receptor is reacti-vated and largely refractory to antiandrogens in prostate cancers that relapse subsequent to andro-gen-deprivation therapy, indicating that complete androgen receptor blockade is not achieved with currently available antiandrogens and that more potent antagonists (possibly functioning through alternative mechanisms such as androgen receptor degradation) may be more effective[1].

Antiandrogens (primarily bicalutamide) have also been examined as initial monotherapy and appear comparable to castration in some stages of disease, although high doses may be required. In contrast, bicalutamide monotherapy may be inferior to pla-cebo in patients in watchful waiting therapy and inferior to castration in patients with greater disease

burden (and may therefore be less effective in patients with short prostate-specific antigen dou-bling times). Nonetheless, as emphasised in the review, bicalutamide may be preferable in some patients due to a better side effect profile than cas-tration (likely reflecting, in part, normal or increased levels of testosterone, which can be converted to estrogen by aromatase) [2]. However, it should be emphasised that these hormonal differences could also affect efficacy, including the apparent synergy between radiation and androgen deprivation [3]. Based on these observations, it is prudent to limit hormonal therapy (antiandrogen or castration) to patients who have been shown to benefit [4], to consider intermittent androgen-deprivation ther- apyasanalternative,andtouseantiandrogenmono-therapy cautiously in cases where its efficacy has not been directly compared to castration.

References

[1] Scher HI, Sawyers CL. Biology of progressive, castration-resistant prostate cancer: directed therapies targeting the androgen-receptor signaling axis. J Clin Oncol 2005;23: 8253–61.

[2] Smith MR, Goode M, Zietman AL, McGovern FJ, Lee H, Finkelstein JS. Bicalutamide monotherapy versus leupro-lide monotherapy for prostate cancer: effects on bone mineral density and body composition. J Clin Oncol 2004;22:2546–53.

[3] D’Amico AV, Manola J, Loffredo M, Renshaw AA, Della-Croce A, Kantoff PW. 6-Month androgen suppression plus radiation therapy vs radiation therapy alone for patients with clinically localized prostate cancer: a randomized controlled trial. JAMA 2004;292:821–7.

[4] Loblaw DA, Mendelson DS, Talcott JA, et al. American Society of Clinical Oncology recommendations for the initial hormonal management of androgen-sensitive metastatic, recurrent, or progressive prostate cancer. J Clin Oncol 2004;22:2927–41.

References

Related documents

The 137 Cs activity is calculated using a depletion code, and the following data: reaction rate values from the TRIPOLI-4® simulations, initial isotopes concentrations, and a

nlveau du Consel I vls-à-vls de l'ensemble du paquet en questlon notamment la dêlégatlon al lemande malntlent des réserves de fond. vls-à-vls des dlfférentes

Study of phytochemical and diuretic potential of methanol and aqueous extract of aerial parts of Phyla nodiflora Linn. Anti inflammatory and anti neoceptive

Found that increasing the PLGA polymer concentration in the organic phase resulting in increases the particle size and increasing the concentration of goserelin acetate in

(1) It uses the scans’ sequence numbers to make sure that the point pairs are selected randomly from different scans, avoiding the unreasonable situation of detecting the target’s

[82] Access to cloud computing services in traditional classified environments and in modern mobile environments provides numerous opportunities to gain visibility

In this manner, in this task, the component focuses are removed from each image obstruct as piece highlights and the element focuses ought to be hearty to

The impact of maternal postnatal depression on men’s experiences of fathering: A qualitative study of British fathers.. Leah Beestin a , Siobhan Hugh-Jones a *, Brendan Gough

projects to apply project management techniques to them.... When to Use

Table 1 compares the semantic dependency pars- ing performance of SPIGOT to all five baselines. , 2017 ) is a state-of-the-art variant of NEURBOPARSER that is trained using

LQR algorithm is used for the control processes.The results showed that the input current to the magnetic circuit plays an important role regarding the dynamic

The correlation matrix analysis between working experience in organization with medical staff absenteeism and turnover based on above correlation table 1 the results shows

relationship a tripartite one or even go so far as to designate the insurer a “client” of the insured’s attorney are much more likely to allow an insurer to bring a

In Conclusion can be said that MapTalk can be a valuable instrument to support decision making within collaborative spatial planning processes, but that positioning of the

SCHOEN and BROWN (1993) showed that if marker gene data are available for many populations of a species, signifi- cant gains in the number of neutral alleles retained in

We isolated and characterized three genes, crcA, apE and crcB, which when present in high copy confer camphor resistance on a cell and suppress mutations in

Finally, we propose a new model that can leverage remittances while serving immigrant communities by: (a) marketing a full array of financial services to immigrant remitters and

Berdasarkan pembahasan hasil analisis kemampuan pemecahan masalah siswa kelas XI MIA 2 SMA Islam Sunan Gunung Jati yang telah menempuh materi fluida statis, peneliti

Sebaran lokasi tangkapan perikanan skala kecil di Desa Berakit dan Malang Rapat yang memanfaatkan keberadaan ekosistem lamun sebagai tempat mencari ikan dan biota laut

Washington Blvd., Ogden, UT 84404 walkert@owatc.edu Vchair, Portfolio Mgr, Wells Capital Mgmt... Washington Blvd., Ogden, UT 84404 walkert@owatc.edu Vchair, Portfolio

• Basel II: Under Basel II, banks are currently required to have a capital rate of the first category 4%, with 2% of the core capital ratio of the first category. The

By considering an unobserved binary variable, we graphically investigate how randomized trials can also stochastically answer the question, &#34;What would be the effect of treatment

The goal of this thesis is to address problems that lie at the interface of statistical processing of data, online learning and network science with a focus on developing