Cardiovascular outcomes in patients with locally advanced and metastatic prostate cancer treated with luteinising-hormone-releasing-hormone agonists or transdermal oestrogen: the randomised, phase 2 MRC PATCH trial (PR09)

Ruth E Langley, Fay H Cafferty, Abdulla A Alhasso, Stuart D Rosen, Subramanian Kanaga Sundaram, Suzanne C Freeman, Philip Pollock, Rachel C Jinks, Ian F Godsland, Roger Kockelbergh, Noel W Clarke, Howard G Kynaston, Mahesh Kb Parmar, Paul D Abel, Ruth E Langley, Fay H Cafferty, Abdulla A Alhasso, Stuart D Rosen, Subramanian Kanaga Sundaram, Suzanne C Freeman, Philip Pollock, Rachel C Jinks, Ian F Godsland, Roger Kockelbergh, Noel W Clarke, Howard G Kynaston, Mahesh Kb Parmar, Paul D Abel

Abstract

Background: Luteinising-hormone-releasing-hormone agonists (LHRHa) to treat prostate cancer are associated with long-term toxic effects, including osteoporosis. Use of parenteral oestrogen could avoid the long-term complications associated with LHRHa and the thromboembolic complications associated with oral oestrogen.

Methods: In this multicentre, open-label, randomised, phase 2 trial, we enrolled men with locally advanced or metastatic prostate cancer scheduled to start indefinite hormone therapy. Randomisation was by minimisation, in a 2:1 ratio, to four self-administered oestrogen patches (100 μg per 24 h) changed twice weekly or LHRHa given according to local practice. After castrate testosterone concentrations were reached (1·7 nmol/L or lower) men received three oestrogen patches changed twice weekly. The primary outcome, cardiovascular morbidity and mortality, was analysed by modified intention to treat and by therapy at the time of the event to account for treatment crossover in cases of disease progression. This study is registered with ClinicalTrials.gov, number NCT00303784.

Findings: 85 patients were randomly assigned to receive LHRHa and 169 to receive oestrogen patches. All 85 patients started LHRHa, and 168 started oestrogen patches. At 3 months, 70 (93%) of 75 receiving LHRHa and 111 (92%) of 121 receiving oestrogen had achieved castrate testosterone concentrations. After a median follow-up of 19 months (IQR 12-31), 24 cardiovascular events were reported, six events in six (7·1%) men in the LHRHa group (95% CI 2·7-14·9) and 18 events in 17 (10·1%) men in the oestrogen-patch group (6·0-15·6). Nine (50%) of 18 events in the oestrogen group occurred after crossover to LHRHa. Mean 12-month changes in fasting glucose concentrations were 0·33 mmol/L (5·5%) in the LHRHa group and -0·16 mmol/L (-2·4%) in the oestrogen-patch group (p=0·004), and for fasting cholesterol were 0·20 mmol/L (4·1%) and -0·23 mmol/L (-3·3%), respectively (p<0·0001). Other adverse events reported by 6 months included gynaecomastia (15 [19%] of 78 patients in the LHRHa group vs 104 [75%] of 138 in the oestrogen-patch group), hot flushes (44 [56%] vs 35 [25%]), and dermatological problems (10 [13%] vs 58 [42%]).

Interpretation: Parenteral oestrogen could be a potential alternative to LHRHa in management of prostate cancer if efficacy is confirmed. On the basis of our findings, enrolment in the PATCH trial has been extended, with a primary outcome of progression-free survival.

Funding: Cancer Research UK, MRC Clinical Trials Unit.

Copyright © 2013 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile Randomisation was 2:1 (oestrogen patches:LHRHa). LHRHa=luteinising-hormone-releasing-hormone agonists. OP=oestrogen patches. *One patient did not start treatment, withdrew soon after randomisation, and did not attend any trial visits. †Men with oestradiol concentraions of 250 pmol/L or lower were assumed not to be using OP and were excluded. ‡Includes a small number of individuals (maximum 3) who had been assumed not to be receiving treatment at an earlier time (eg, owing to low oestradiol concentrations, reported difficulty in using OP, or missed appointments).
Figure 2
Figure 2
Changes in fasting glucose (A) and total cholesterol (B) concentrations in patients still receiving treatment at 6 and 12 months Patients were not receiving additional therapy. Boxes indicate median and IQR, whiskers indicate 1·5×IQR, and dots indicate outlying values. LHRHa=luteinising-hormone-releasing-hormone agonists. OP=oestrogen patches.

References

    1. Cancer Research UK Worldwide cancer statistics. (accessed Jan 29, 2013).
    1. Shahinian VB, Kuo YF, Gilbert SM. Reimbursement policy and androgen-deprivation therapy for prostate cancer. N Engl J Med. 2010;363:1822–1832.
    1. Pagliarulo V, Bracarda S, Eisenberger MA. Contemporary role of androgen deprivation therapy for prostate cancer. Eur Urol. 2012;61:11–25.
    1. Freedland SJ, Eastham J, Shore N. Androgen deprivation therapy and estrogen deficiency induced adverse effects in the treatment of prostate cancer. Prostate Cancer Prostatic Dis. 2009;12:333–338.
    1. Lee H, McGovern K, Finkelstein JS, Smith MR. Changes in bone mineral density and body composition during initial and long-term gonadotropin-releasing hormone agonist treatment for prostate carcinoma. Cancer. 2005;104:1633–1637.
    1. Wadhwa VK, Weston R, Parr NJ. Frequency of zoledronic acid to prevent further bone loss in osteoporotic patients undergoing androgen deprivation therapy for prostate cancer. BJU Int. 2010;105:1082–1088.
    1. Smith MR, Lee WC, Brandman J, Wang Q, Botteman M, Pashos CL. Gonadotropin-releasing hormone agonists and fracture risk: a claims-based cohort study of men with nonmetastatic prostate cancer. J Clin Oncol. 2005;23:7897–7903.
    1. Abrahamsen B, Nielsen MF, Eskildsen P, Andersen JT, Walter S, Brixen K. Fracture risk in Danish men with prostate cancer: a nationwide register study. BJU Int. 2007;100:749–754.
    1. Shahinian VB, Kuo YF, Freeman JL, Goodwin JS. Risk of fracture after androgen deprivation for prostate cancer. N Engl J Med. 2005;352:154–164.
    1. Alibhai SM, Duong-Hua M, Sutradhar R. Impact of androgen deprivation therapy on cardiovascular disease and diabetes. J Clin Oncol. 2009;27:3452–3458.
    1. Saylor PJ, Smith MR. Metabolic complications of androgen deprivation therapy for prostate cancer. J Urol. 2009;181:1998–2006.
    1. Keating NL, O'Malley AJ, Smith MR. Diabetes and cardiovascular disease during androgen deprivation therapy for prostate cancer. J Clin Oncol. 2006;24:4448–4456.
    1. Keating NL, O'Malley AJ, Freedland SJ, Smith MR. Diabetes and cardiovascular disease during androgen deprivation therapy: observational study of veterans with prostate cancer. J Natl Cancer Inst. 2010;102:39–46.
    1. Levine GN, D'Amico AV, Berger P. Androgen-deprivation therapy in prostate cancer and cardiovascular risk: a science advisory from the American Heart Association, American Cancer Society, and American Urological Association: endorsed by the American Society for Radiation Oncology. Circulation. 2010;121:833–840.
    1. Byar DP. Proceedings: the Veterans Administration Cooperative Urological Research Group's studies of cancer of the prostate. Cancer. 1973;32:1126–1130.
    1. von Schoultz B, Carlstrom K, Collste L. Estrogen therapy and liver function—metabolic effects of oral and parenteral administration. Prostate. 1989;14:389–395.
    1. Henriksson P, Blomback M, Eriksson A, Stege R, Carlstrom K. Effect of parenteral oestrogen on the coagulation system in patients with prostatic carcinoma. Br J Urol. 1990;65:282–285.
    1. Ockrim J. Transdermal oestradiol therapy for the treatment of advanced prostate cancer. Imperial College; London: 2003. MD thesis.
    1. Langley RE, Godsland IF, Kynaston H. Early hormonal data from a multicentre phase II trial using transdermal oestrogen patches as first-line hormonal therapy in patients with locally advanced or metastatic prostate cancer. BJU Int. 2008;102:442–445.
    1. Hedlund PO, Ala-Opas M, Brekkan E. Parenteral estrogen versus combined androgen deprivation in the treatment of metastatic prostatic cancer—Scandinavian Prostatic Cancer Group (SPCG) study no. 5. Scand J Urol Nephrol. 2002;36:405–413.
    1. Ockrim JL, Lalani EN, Laniado ME, Carter SS, Abel PD. Transdermal estradiol therapy for advanced prostate cancer—forward to the past? J Urol. 2003;169:1735–1737.
    1. Blackard CE, Doe RP, Mellinger GT, Byar DP. Incidence of cardiovascular disease and death in patients receiving diethylstilbestrol for carcinoma of the prostate. Cancer. 1970;26:249–256.
    1. Van Hemelrijck M, Garmo H, Holmberg L. Absolute and relative risk of cardiovascular disease in men with prostate cancer: results from the population-based PCBaSe Sweden. J Clin Oncol. 2010;28:3448–3456.
    1. Tsai HK, D'Amico AV, Sadetsky N, Chen MH, Carroll PR. Androgen deprivation therapy for localized prostate cancer and the risk of cardiovascular mortality. J Natl Cancer Inst. 2007;99:1516–1524.
    1. Punnen S, Cooperberg MR, Sadetsky N, Carroll PR. Androgen deprivation therapy and cardiovascular risk. J Clin Oncol. 2011;29:3510–3516.
    1. Nguyen PL, Je Y, Schutz FA. Association of androgen deprivation therapy with cardiovascular death in patients with prostate cancer: a meta-analysis of randomized trials. JAMA. 2011;306:2359–2366.
    1. Norman G, Dean ME, Langley RE. Parenteral oestrogen in the treatment of prostate cancer: a systematic review. Br J Cancer. 2008;98:697–707.
    1. Bracamonte MP, Miller VM. Vascular effects of estrogens: arterial protection versus venous thrombotic risk. Trends Endocrinol Metab. 2001;12:204–209.
    1. Hedlund PO, Damber JE, Hagerman I. Parenteral estrogen versus combined androgen deprivation in the treatment of metastatic prostatic cancer: part 2. Final evaluation of the Scandinavian Prostatic Cancer Group (SPCG) study no. 5. Scand J Urol Nephrol. 2008;42:220–229.
    1. Hedlund PO, Johansson R, Damber JE. Significance of pretreatment cardiovascular morbidity as a risk factor during treatment with parenteral oestrogen or combined androgen deprivation of 915 patients with metastasized prostate cancer: evaluation of cardiovascular events in a randomized trial. Scand J Urol Nephrol. 2011;45:346–353.
    1. Smith MR, Finkelstein JS, McGovern FJ. Changes in body composition during androgen deprivation therapy for prostate cancer. J Clin Endocrinol Metab. 2002;87:599–603.
    1. Eri LM, Urdal P, Bechensteen AG. Effects of the luteinizing hormone-releasing hormone agonist leuprolide on lipoproteins, fibrinogen and plasminogen activator inhibitor in patients with benign prostatic hyperplasia. J Urol. 1995;154:100–104.
    1. Torimoto K, Samma S, Kagebayashi Y. The effects of androgen deprivation therapy on lipid metabolism and body composition in Japanese patients with prostate cancer. Jpn J Clin Oncol. 2011;41:577–581.
    1. Suzuki K, Nukui A, Hara Y, Morita T. Glucose intolerance during hormonal therapy for prostate cancer. Prostate Cancer Prostatic Dis. 2007;10:384–387.
    1. Dockery F, Bulpitt CJ, Agarwal S, Donaldson M, Rajkumar C. Testosterone suppression in men with prostate cancer leads to an increase in arterial stiffness and hyperinsulinaemia. Clin Sci (Lond) 2003;104:195–201.
    1. Smith MR. Insulin sensitivity during combined androgen blockade for prostate cancer. J Clin Endocrinol Metab. 2006;91:1305–1308.
    1. Smith MR, Lee H, McGovern F. Metabolic changes during gonadotropin-releasing hormone agonist therapy for prostate cancer: differences from the classic metabolic syndrome. Cancer. 2008;112:2188–2194.
    1. Henriksson P, Angelin B, Berglund L. Hormonal regulation of serum Lp (a) levels. Opposite effects after estrogen treatment and orchidectomy in males with prostatic carcinoma. J Clin Invest. 1992;89:1166–1171.
    1. Godsland IF. Oestrogens and insulin secretion. Diabetologia. 2005;48:2213–2220.

Source: PubMed

3
订阅