Skip to main content
Top
Published in: Reviews in Endocrine and Metabolic Disorders 6/2022

Open Access 24-06-2022 | Prostate Cancer

Androgens, aging, and prostate health

Authors: Karin Welén, Jan-Erik Damber

Published in: Reviews in Endocrine and Metabolic Disorders | Issue 6/2022

Login to get access

Abstract

Due to late onset hypogonadism (LOH), there is an increased usage of testosterone replacement therapy (TRT) in the aging male population. Since prostate is a target organ for androgens and anti-androgenic strategies are used to treat and palliate benign prostate hyperplasia (BPH) and prostate cancer (PC), the prevalence of both increases with age, the possible influence of TRT on prostate health becomes highly relevant. The present review summarizes existing data on the associations between endogenous hormone concentrations and prostate growth and concludes that circulating concentrations of androgens do not appear to be associated with the risks of development of BPH or initiation or progression of PC. The explanation for these findings relates to an apparent insensitivity of prostatic tissue to changes of testosterone concentrations within the physiological range.
Literature
1.
go back to reference Nguyen CP, et al. Testosterone and “age-related hypogonadism”--FDA concerns. N Engl J Med. 2015;373(8):689–91.CrossRef Nguyen CP, et al. Testosterone and “age-related hypogonadism”--FDA concerns. N Engl J Med. 2015;373(8):689–91.CrossRef
2.
go back to reference Morgentaler A, et al. Fundamental concepts regarding testosterone deficiency and treatment: International expert consensus resolutions. Mayo Clin Proc. 2016;91(7):881–96.CrossRef Morgentaler A, et al. Fundamental concepts regarding testosterone deficiency and treatment: International expert consensus resolutions. Mayo Clin Proc. 2016;91(7):881–96.CrossRef
3.
go back to reference Rizk PJ, et al. Testosterone therapy improves erectile function and libido in hypogonadal men. Curr Opin Urol. 2017;27(6):511–5.CrossRef Rizk PJ, et al. Testosterone therapy improves erectile function and libido in hypogonadal men. Curr Opin Urol. 2017;27(6):511–5.CrossRef
4.
go back to reference Yassin A, et al. Testosterone therapy in men with hypogonadism prevents progression from prediabetes to type 2 diabetes: Eight-year data from a registry study. Diabetes Care. 2019;42(6):1104–11.CrossRef Yassin A, et al. Testosterone therapy in men with hypogonadism prevents progression from prediabetes to type 2 diabetes: Eight-year data from a registry study. Diabetes Care. 2019;42(6):1104–11.CrossRef
5.
go back to reference Garnick MB. Testosterone replacement therapy faces FDA scrutiny. JAMA. 2015;313(6):563–4.CrossRef Garnick MB. Testosterone replacement therapy faces FDA scrutiny. JAMA. 2015;313(6):563–4.CrossRef
6.
go back to reference Berry SJ, et al. The development of human benign prostatic hyperplasia with age. J Urol. 1984;132(3):474–9.CrossRef Berry SJ, et al. The development of human benign prostatic hyperplasia with age. J Urol. 1984;132(3):474–9.CrossRef
7.
go back to reference Kok ET, et al. Risk factors for lower urinary tract symptoms suggestive of benign prostatic hyperplasia in a community based population of healthy aging men: the Krimpen Study. J Urol. 2009;181(2):710–6.CrossRef Kok ET, et al. Risk factors for lower urinary tract symptoms suggestive of benign prostatic hyperplasia in a community based population of healthy aging men: the Krimpen Study. J Urol. 2009;181(2):710–6.CrossRef
8.
go back to reference Madersbacher S, Sampson N, Culig Z. Pathophysiology of benign prostatic hyperplasia and benign prostatic enlargement: A mini-review. Gerontology. 2019;65(5):458–64.CrossRef Madersbacher S, Sampson N, Culig Z. Pathophysiology of benign prostatic hyperplasia and benign prostatic enlargement: A mini-review. Gerontology. 2019;65(5):458–64.CrossRef
9.
go back to reference Prajapati A, et al. Prostate stem cells in the development of benign prostate hyperplasia and prostate cancer: emerging role and concepts. Biomed Res Int. 2013;2013:107954.CrossRef Prajapati A, et al. Prostate stem cells in the development of benign prostate hyperplasia and prostate cancer: emerging role and concepts. Biomed Res Int. 2013;2013:107954.CrossRef
10.
go back to reference Guess HA, et al. Cumulative prevalence of prostatism matches the autopsy prevalence of benign prostatic hyperplasia. Prostate. 1990;17(3):241–6.CrossRef Guess HA, et al. Cumulative prevalence of prostatism matches the autopsy prevalence of benign prostatic hyperplasia. Prostate. 1990;17(3):241–6.CrossRef
11.
go back to reference Sanda MG, et al. Genetic susceptibility of benign prostatic hyperplasia. J Urol. 1994;152(1):115–9.CrossRef Sanda MG, et al. Genetic susceptibility of benign prostatic hyperplasia. J Urol. 1994;152(1):115–9.CrossRef
12.
go back to reference Sanda MG, et al. Clinical and biological characteristics of familial benign prostatic hyperplasia. J Urol. 1997;157(3):876–9.CrossRef Sanda MG, et al. Clinical and biological characteristics of familial benign prostatic hyperplasia. J Urol. 1997;157(3):876–9.CrossRef
13.
go back to reference Wen S, et al. Stromal androgen receptor roles in the development of normal prostate, benign prostate hyperplasia, and prostate cancer. Am J Pathol. 2015;185(2):293–301.CrossRef Wen S, et al. Stromal androgen receptor roles in the development of normal prostate, benign prostate hyperplasia, and prostate cancer. Am J Pathol. 2015;185(2):293–301.CrossRef
14.
go back to reference Parsons JK. Benign prostatic hyperplasia and male lower urinary tract symptoms: Epidemiology and risk factors. Curr Bladder Dysfunct Rep. 2010;5(4):212–8.CrossRef Parsons JK. Benign prostatic hyperplasia and male lower urinary tract symptoms: Epidemiology and risk factors. Curr Bladder Dysfunct Rep. 2010;5(4):212–8.CrossRef
15.
go back to reference Kristal AR, et al. Serum steroid and sex hormone-binding globulin concentrations and the risk of incident benign prostatic hyperplasia: results from the prostate cancer prevention trial. Am J Epidemiol. 2008;168(12):1416–24.CrossRef Kristal AR, et al. Serum steroid and sex hormone-binding globulin concentrations and the risk of incident benign prostatic hyperplasia: results from the prostate cancer prevention trial. Am J Epidemiol. 2008;168(12):1416–24.CrossRef
16.
go back to reference Parsons JK, et al. Prospective study of serum dihydrotestosterone and subsequent risk of benign prostatic hyperplasia in community dwelling men: The Rancho Bernardo Study. J Urol. 2010;184(3):1040–4.CrossRef Parsons JK, et al. Prospective study of serum dihydrotestosterone and subsequent risk of benign prostatic hyperplasia in community dwelling men: The Rancho Bernardo Study. J Urol. 2010;184(3):1040–4.CrossRef
17.
go back to reference Trifiro MD, et al. Serum sex hormones and the 20-year risk of lower urinary tract symptoms in community-dwelling older men. BJU Int. 2010;105(11):1554–9.CrossRef Trifiro MD, et al. Serum sex hormones and the 20-year risk of lower urinary tract symptoms in community-dwelling older men. BJU Int. 2010;105(11):1554–9.CrossRef
18.
go back to reference Nicholson TM, Ricke WA. Androgens and estrogens in benign prostatic hyperplasia: past, present and future. Differentiation. 2011;82(4–5):184–99.CrossRef Nicholson TM, Ricke WA. Androgens and estrogens in benign prostatic hyperplasia: past, present and future. Differentiation. 2011;82(4–5):184–99.CrossRef
19.
go back to reference Haghsheno MA, et al. Lower urinary tract symptoms are associated with low levels of serum serotonin, high levels of adiponectin and fasting glucose, and benign prostatic enlargement. Scand J Urol. 2015;49(2):155–61.CrossRef Haghsheno MA, et al. Lower urinary tract symptoms are associated with low levels of serum serotonin, high levels of adiponectin and fasting glucose, and benign prostatic enlargement. Scand J Urol. 2015;49(2):155–61.CrossRef
20.
go back to reference Hammarsten J, Hogstedt B. Hyperinsulinaemia as a risk factor for developing benign prostatic hyperplasia. Eur Urol. 2001;39(2):151–8.CrossRef Hammarsten J, Hogstedt B. Hyperinsulinaemia as a risk factor for developing benign prostatic hyperplasia. Eur Urol. 2001;39(2):151–8.CrossRef
21.
go back to reference Nandeesha H, et al. Hyperinsulinemia and dyslipidemia in non-diabetic benign prostatic hyperplasia. Clin Chim Acta. 2006;370(1–2):89–93.CrossRef Nandeesha H, et al. Hyperinsulinemia and dyslipidemia in non-diabetic benign prostatic hyperplasia. Clin Chim Acta. 2006;370(1–2):89–93.CrossRef
22.
go back to reference Wang W, Bergh A, Damber JE. Chronic inflammation in benign prostate hyperplasia is associated with focal upregulation of cyclooxygenase-2, Bcl-2, and cell proliferation in the glandular epithelium. Prostate. 2004;61(1):60–72.CrossRef Wang W, Bergh A, Damber JE. Chronic inflammation in benign prostate hyperplasia is associated with focal upregulation of cyclooxygenase-2, Bcl-2, and cell proliferation in the glandular epithelium. Prostate. 2004;61(1):60–72.CrossRef
23.
go back to reference Rastrelli G, et al. Testosterone and benign prostatic hyperplasia. Sex Med Rev. 2019;7(2):259–71.CrossRef Rastrelli G, et al. Testosterone and benign prostatic hyperplasia. Sex Med Rev. 2019;7(2):259–71.CrossRef
24.
go back to reference Malik S, et al. Impact of the metabolic syndrome on mortality from coronary heart disease, cardiovascular disease, and all causes in United States adults. Circulation. 2004;110(10):1245–50.CrossRef Malik S, et al. Impact of the metabolic syndrome on mortality from coronary heart disease, cardiovascular disease, and all causes in United States adults. Circulation. 2004;110(10):1245–50.CrossRef
25.
go back to reference De Nunzio C, et al. The correlation between metabolic syndrome and prostatic diseases. Eur Urol. 2012;61(3):560–70.CrossRef De Nunzio C, et al. The correlation between metabolic syndrome and prostatic diseases. Eur Urol. 2012;61(3):560–70.CrossRef
26.
go back to reference Hammarsten J, et al. Components of the metabolic syndrome-risk factors for the development of benign prostatic hyperplasia. Prostate Cancer Prostatic Dis. 1998;1(3):157–62.CrossRef Hammarsten J, et al. Components of the metabolic syndrome-risk factors for the development of benign prostatic hyperplasia. Prostate Cancer Prostatic Dis. 1998;1(3):157–62.CrossRef
27.
go back to reference DiBello JR, et al. Prevalence of metabolic syndrome and its components among men with and without clinical benign prostatic hyperplasia: a large, cross-sectional, UK epidemiological study. BJU Int. 2016;117(5):801–8.CrossRef DiBello JR, et al. Prevalence of metabolic syndrome and its components among men with and without clinical benign prostatic hyperplasia: a large, cross-sectional, UK epidemiological study. BJU Int. 2016;117(5):801–8.CrossRef
28.
go back to reference Devaraj S, Singh U, Jialal I. Human C-reactive protein and the metabolic syndrome. Curr Opin Lipidol. 2009;20(3):182–9.CrossRef Devaraj S, Singh U, Jialal I. Human C-reactive protein and the metabolic syndrome. Curr Opin Lipidol. 2009;20(3):182–9.CrossRef
29.
go back to reference Penna G, et al. Seminal plasma cytokines and chemokines in prostate inflammation: interleukin 8 as a predictive biomarker in chronic prostatitis/chronic pelvic pain syndrome and benign prostatic hyperplasia. Eur Urol. 2007;51(2):524–33 discussion 533.CrossRef Penna G, et al. Seminal plasma cytokines and chemokines in prostate inflammation: interleukin 8 as a predictive biomarker in chronic prostatitis/chronic pelvic pain syndrome and benign prostatic hyperplasia. Eur Urol. 2007;51(2):524–33 discussion 533.CrossRef
30.
go back to reference Crawford ED, et al. Baseline factors as predictors of clinical progression of benign prostatic hyperplasia in men treated with placebo. J Urol. 2006;175(4):1422–6.CrossRef Crawford ED, et al. Baseline factors as predictors of clinical progression of benign prostatic hyperplasia in men treated with placebo. J Urol. 2006;175(4):1422–6.CrossRef
31.
go back to reference Schenk JM, et al. Biomarkers of systemic inflammation and risk of incident, symptomatic benign prostatic hyperplasia: results from the prostate cancer prevention trial. Am J Epidemiol. 2010;171(5):571–82.CrossRef Schenk JM, et al. Biomarkers of systemic inflammation and risk of incident, symptomatic benign prostatic hyperplasia: results from the prostate cancer prevention trial. Am J Epidemiol. 2010;171(5):571–82.CrossRef
32.
go back to reference Rohrmann S, et al. Serum sex steroid hormones and lower urinary tract symptoms in Third National Health and Nutrition Examination Survey (NHANES III). Urology. 2007;69(4):708–13.CrossRef Rohrmann S, et al. Serum sex steroid hormones and lower urinary tract symptoms in Third National Health and Nutrition Examination Survey (NHANES III). Urology. 2007;69(4):708–13.CrossRef
33.
go back to reference Traish AM. Adverse health effects of testosterone deficiency (TD) in men. Steroids. 2014;88:106–16.CrossRef Traish AM. Adverse health effects of testosterone deficiency (TD) in men. Steroids. 2014;88:106–16.CrossRef
34.
go back to reference van Deursen JM. The role of senescent cells in ageing. Nature. 2014;509(7501):439–46.CrossRef van Deursen JM. The role of senescent cells in ageing. Nature. 2014;509(7501):439–46.CrossRef
35.
go back to reference Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585–621.CrossRef Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585–621.CrossRef
36.
go back to reference Chen YQ, et al. Testosterone ameliorates vascular aging via the Gas6/Axl signaling pathway. Aging. 2020;12(16):16111–25.CrossRef Chen YQ, et al. Testosterone ameliorates vascular aging via the Gas6/Axl signaling pathway. Aging. 2020;12(16):16111–25.CrossRef
37.
go back to reference Coppe JP, et al. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118.CrossRef Coppe JP, et al. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118.CrossRef
38.
go back to reference Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci. 2014;69(Suppl 1):S4–9.CrossRef Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci. 2014;69(Suppl 1):S4–9.CrossRef
39.
go back to reference Choi J, et al. Expression of senescence-associated beta-galactosidase in enlarged prostates from men with benign prostatic hyperplasia. Urology. 2000;56(1):160–6.CrossRef Choi J, et al. Expression of senescence-associated beta-galactosidase in enlarged prostates from men with benign prostatic hyperplasia. Urology. 2000;56(1):160–6.CrossRef
40.
go back to reference Jiang S, Song CS, Chatterjee B. Stimulation of prostate cells by the senescence phenotype of epithelial and stromal cells: Implication for benign prostate hyperplasia. FASEB Bioadv. 2019;1(6):353–63. Jiang S, Song CS, Chatterjee B. Stimulation of prostate cells by the senescence phenotype of epithelial and stromal cells: Implication for benign prostate hyperplasia. FASEB Bioadv. 2019;1(6):353–63.
41.
go back to reference Fiard G, et al. Cellular senescence as a possible link between prostate diseases of the ageing male. Nat Rev Urol. 2021;18(10):597–610.CrossRef Fiard G, et al. Cellular senescence as a possible link between prostate diseases of the ageing male. Nat Rev Urol. 2021;18(10):597–610.CrossRef
42.
go back to reference Sung H, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–49.CrossRef Sung H, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–49.CrossRef
43.
go back to reference Gandaglia G, et al. Epidemiology and prevention of prostate cancer. Eur Urol Oncol. 2021;4(6):877–92 Gandaglia G, et al. Epidemiology and prevention of prostate cancer. Eur Urol Oncol. 2021;4(6):877–92
44.
go back to reference Welch HG, Albertsen PC. Prostate cancer diagnosis and treatment after the introduction of prostate-specific antigen screening: 1986–2005. J Natl Cancer Inst. 2009;101(19):1325–9.CrossRef Welch HG, Albertsen PC. Prostate cancer diagnosis and treatment after the introduction of prostate-specific antigen screening: 1986–2005. J Natl Cancer Inst. 2009;101(19):1325–9.CrossRef
45.
go back to reference Bell KJ, et al. Prevalence of incidental prostate cancer: A systematic review of autopsy studies. Int J Cancer. 2015;137(7):1749–57.CrossRef Bell KJ, et al. Prevalence of incidental prostate cancer: A systematic review of autopsy studies. Int J Cancer. 2015;137(7):1749–57.CrossRef
46.
go back to reference Gacci M, et al. Meta-analysis of metabolic syndrome and prostate cancer. Prostate Cancer Prostatic Dis. 2017;20(2):146–55.CrossRef Gacci M, et al. Meta-analysis of metabolic syndrome and prostate cancer. Prostate Cancer Prostatic Dis. 2017;20(2):146–55.CrossRef
47.
go back to reference Rivera-Izquierdo M, et al. Obesity as a Risk Factor for Prostate Cancer Mortality: A Systematic Review and Dose-Response Meta-Analysis of 280,199 Patients. Cancers (Basel). 2021;13(16):4169. Rivera-Izquierdo M, et al. Obesity as a Risk Factor for Prostate Cancer Mortality: A Systematic Review and Dose-Response Meta-Analysis of 280,199 Patients. Cancers (Basel). 2021;13(16):4169.
48.
go back to reference Michels N, et al. Chronic inflammation towards cancer incidence: A systematic review and meta-analysis of epidemiological studies. Crit Rev Oncol Hematol. 2021;157:103177.CrossRef Michels N, et al. Chronic inflammation towards cancer incidence: A systematic review and meta-analysis of epidemiological studies. Crit Rev Oncol Hematol. 2021;157:103177.CrossRef
49.
go back to reference Damber JE. Endocrine therapy for prostate cancer. Acta Oncol. 2005;44(6):605–9.CrossRef Damber JE. Endocrine therapy for prostate cancer. Acta Oncol. 2005;44(6):605–9.CrossRef
50.
go back to reference Ohlson N, et al. Cell proliferation and apoptosis in prostate tumors and adjacent non-malignant prostate tissue in patients at different time-points after castration treatment. Prostate. 2005;62(4):307–15.CrossRef Ohlson N, et al. Cell proliferation and apoptosis in prostate tumors and adjacent non-malignant prostate tissue in patients at different time-points after castration treatment. Prostate. 2005;62(4):307–15.CrossRef
51.
go back to reference Westin P, et al. Castration therapy rapidly induces apoptosis in a minority and decreases cell proliferation in a majority of human prostatic tumors. Am J Pathol. 1995;146(6):1368–75. Westin P, et al. Castration therapy rapidly induces apoptosis in a minority and decreases cell proliferation in a majority of human prostatic tumors. Am J Pathol. 1995;146(6):1368–75.
52.
go back to reference Cunha GR, et al. Hormone-induced morphogenesis and growth: role of mesenchymal-epithelial interactions. Recent Prog Horm Res. 1983;39:559–98. Cunha GR, et al. Hormone-induced morphogenesis and growth: role of mesenchymal-epithelial interactions. Recent Prog Horm Res. 1983;39:559–98.
53.
go back to reference Wu CT, et al. Increased prostate cell proliferation and loss of cell differentiation in mice lacking prostate epithelial androgen receptor. Proc Natl Acad Sci U S A. 2007;104(31):12679–84.CrossRef Wu CT, et al. Increased prostate cell proliferation and loss of cell differentiation in mice lacking prostate epithelial androgen receptor. Proc Natl Acad Sci U S A. 2007;104(31):12679–84.CrossRef
54.
go back to reference Boyle P, et al. Endogenous and exogenous testosterone and the risk of prostate cancer and increased prostate-specific antigen (PSA) level: A meta-analysis. BJU Int. 2016;118(5):731–41.CrossRef Boyle P, et al. Endogenous and exogenous testosterone and the risk of prostate cancer and increased prostate-specific antigen (PSA) level: A meta-analysis. BJU Int. 2016;118(5):731–41.CrossRef
55.
go back to reference Thompson IM, et al. The influence of finasteride on the development of prostate cancer. N Engl J Med. 2003;349(3):215–24.CrossRef Thompson IM, et al. The influence of finasteride on the development of prostate cancer. N Engl J Med. 2003;349(3):215–24.CrossRef
56.
go back to reference Thompson IM Jr, et al. Long-term survival of participants in the prostate cancer prevention trial. N Engl J Med. 2013;369(7):603–10.CrossRef Thompson IM Jr, et al. Long-term survival of participants in the prostate cancer prevention trial. N Engl J Med. 2013;369(7):603–10.CrossRef
57.
go back to reference Andriole GL, et al. Effect of dutasteride on the risk of prostate cancer. N Engl J Med. 2010;362(13):1192–202.CrossRef Andriole GL, et al. Effect of dutasteride on the risk of prostate cancer. N Engl J Med. 2010;362(13):1192–202.CrossRef
58.
go back to reference Bonde Miranda T, et al. 5alpha-reductase inhibitors and risk of prostate cancer death. J Urol. 2020;204(4):714–9.CrossRef Bonde Miranda T, et al. 5alpha-reductase inhibitors and risk of prostate cancer death. J Urol. 2020;204(4):714–9.CrossRef
59.
go back to reference Lane BR, et al. Low testosterone and risk of biochemical recurrence and poorly differentiated prostate cancer at radical prostatectomy. Urology. 2008;72(6):1240–5.CrossRef Lane BR, et al. Low testosterone and risk of biochemical recurrence and poorly differentiated prostate cancer at radical prostatectomy. Urology. 2008;72(6):1240–5.CrossRef
60.
go back to reference Lee JK, et al. Preoperative serum sex hormone-binding globulin level is an independent predictor of biochemical outcome after radical prostatectomy. Med (Baltim). 2015;94(28):e1185.CrossRef Lee JK, et al. Preoperative serum sex hormone-binding globulin level is an independent predictor of biochemical outcome after radical prostatectomy. Med (Baltim). 2015;94(28):e1185.CrossRef
61.
go back to reference Roder MA, et al. Serum testosterone level as a predictor of biochemical failure after radical prostatectomy for localized prostate cancer. BJU Int. 2012;109(4):520–4.CrossRef Roder MA, et al. Serum testosterone level as a predictor of biochemical failure after radical prostatectomy for localized prostate cancer. BJU Int. 2012;109(4):520–4.CrossRef
62.
go back to reference Yamamoto S, et al. Preoperative serum testosterone level as an independent predictor of treatment failure following radical prostatectomy. Eur Urol. 2007;52(3):696–701.CrossRef Yamamoto S, et al. Preoperative serum testosterone level as an independent predictor of treatment failure following radical prostatectomy. Eur Urol. 2007;52(3):696–701.CrossRef
63.
go back to reference Patel A. Does Baseline Serum Testosterone Influence Androgen Deprivation Therapy Outcomes in Hormone Naive Patients with Advanced Prostate Cancer? J Urol. 2021;205(3):806–11.CrossRef Patel A. Does Baseline Serum Testosterone Influence Androgen Deprivation Therapy Outcomes in Hormone Naive Patients with Advanced Prostate Cancer? J Urol. 2021;205(3):806–11.CrossRef
64.
go back to reference Roach M 3rd. Baseline serum testosterone in men treated with androgen deprivation therapy and radiotherapy for localized prostate cancer. Int J Radiat Oncol Biol Phys. 2010;78(5):1314–22.CrossRef Roach M 3rd. Baseline serum testosterone in men treated with androgen deprivation therapy and radiotherapy for localized prostate cancer. Int J Radiat Oncol Biol Phys. 2010;78(5):1314–22.CrossRef
65.
go back to reference Morgentaler A, Traish AM. Shifting the paradigm of testosterone and prostate cancer: the saturation model and the limits of androgen-dependent growth. Eur Urol. 2009;55(2):310–20.CrossRef Morgentaler A, Traish AM. Shifting the paradigm of testosterone and prostate cancer: the saturation model and the limits of androgen-dependent growth. Eur Urol. 2009;55(2):310–20.CrossRef
66.
go back to reference Ho SM, et al. Androgen receptor levels and androgen contents in the prostate lobes of intact and testosterone-treated Noble rats. J Androl. 1985;6(5):279–90.CrossRef Ho SM, et al. Androgen receptor levels and androgen contents in the prostate lobes of intact and testosterone-treated Noble rats. J Androl. 1985;6(5):279–90.CrossRef
67.
go back to reference Arnold JT, et al. Comparative effects of DHEA vs. testosterone, dihydrotestosterone, and estradiol on proliferation and gene expression in human LNCaP prostate cancer cells. Am J Physiol Endocrinol Metab. 2005;288(3):E573-84.CrossRef Arnold JT, et al. Comparative effects of DHEA vs. testosterone, dihydrotestosterone, and estradiol on proliferation and gene expression in human LNCaP prostate cancer cells. Am J Physiol Endocrinol Metab. 2005;288(3):E573-84.CrossRef
68.
go back to reference Bologna M, et al. Finasteride dose-dependently reduces the proliferation rate of the LnCap human prostatic cancer cell line in vitro. Urology. 1995;45(2):282–90.CrossRef Bologna M, et al. Finasteride dose-dependently reduces the proliferation rate of the LnCap human prostatic cancer cell line in vitro. Urology. 1995;45(2):282–90.CrossRef
69.
go back to reference Banerjee PP, et al. Age- and lobe-specific responses of the brown Norway rat prostate to androgen. Biol Reprod. 1994;51(4):675–84.CrossRef Banerjee PP, et al. Age- and lobe-specific responses of the brown Norway rat prostate to androgen. Biol Reprod. 1994;51(4):675–84.CrossRef
70.
go back to reference Robaire B, et al. Interactions of testosterone and estradiol-17 beta on the reproductive tract of the male rat. Biol Reprod. 1979;21(2):455–63.CrossRef Robaire B, et al. Interactions of testosterone and estradiol-17 beta on the reproductive tract of the male rat. Biol Reprod. 1979;21(2):455–63.CrossRef
71.
go back to reference Wright AS, et al. Androgen-induced regrowth in the castrated rat ventral prostate: role of 5alpha-reductase. Endocrinology. 1999;140(10):4509–15.CrossRef Wright AS, et al. Androgen-induced regrowth in the castrated rat ventral prostate: role of 5alpha-reductase. Endocrinology. 1999;140(10):4509–15.CrossRef
72.
go back to reference Bhasin S, et al. The effects of supraphysiologic doses of testosterone on muscle size and strength in normal men. N Engl J Med. 1996;335(1):1–7.CrossRef Bhasin S, et al. The effects of supraphysiologic doses of testosterone on muscle size and strength in normal men. N Engl J Med. 1996;335(1):1–7.CrossRef
73.
go back to reference Bhasin S, et al. Testosterone dose-response relationships in healthy young men. Am J Physiol Endocrinol Metab. 2001;281(6):E1172-81.CrossRef Bhasin S, et al. Testosterone dose-response relationships in healthy young men. Am J Physiol Endocrinol Metab. 2001;281(6):E1172-81.CrossRef
74.
go back to reference Cooper CS, et al. Effect of exogenous testosterone on prostate volume, serum and semen prostate specific antigen levels in healthy young men. J Urol. 1998;159(2):441–3.CrossRef Cooper CS, et al. Effect of exogenous testosterone on prostate volume, serum and semen prostate specific antigen levels in healthy young men. J Urol. 1998;159(2):441–3.CrossRef
75.
go back to reference Mohr BA, et al. Are serum hormones associated with the risk of prostate cancer? Prospective results from the Massachusetts Male Aging Study. Urology. 2001;57(5):930–5.CrossRef Mohr BA, et al. Are serum hormones associated with the risk of prostate cancer? Prospective results from the Massachusetts Male Aging Study. Urology. 2001;57(5):930–5.CrossRef
76.
go back to reference Monath JR, et al. Physiologic variations of serum testosterone within the normal range do not affect serum prostate-specific antigen. Urology. 1995;46(1):58–61.CrossRef Monath JR, et al. Physiologic variations of serum testosterone within the normal range do not affect serum prostate-specific antigen. Urology. 1995;46(1):58–61.CrossRef
77.
go back to reference Page ST, et al. Dihydrotestosterone administration does not increase intraprostatic androgen concentrations or alter prostate androgen action in healthy men: a randomized-controlled trial. J Clin Endocrinol Metab. 2011;96(2):430–7.CrossRef Page ST, et al. Dihydrotestosterone administration does not increase intraprostatic androgen concentrations or alter prostate androgen action in healthy men: a randomized-controlled trial. J Clin Endocrinol Metab. 2011;96(2):430–7.CrossRef
78.
go back to reference Thirumalai A, et al. Stable intraprostatic dihydrotestosterone in healthy medically castrate men treated with exogenous testosterone. J Clin Endocrinol Metab. 2016;101(7):2937–44.CrossRef Thirumalai A, et al. Stable intraprostatic dihydrotestosterone in healthy medically castrate men treated with exogenous testosterone. J Clin Endocrinol Metab. 2016;101(7):2937–44.CrossRef
80.
go back to reference Oefelein MG, et al. Reassessment of the definition of castrate levels of testosterone: implications for clinical decision making. Urology. 2000;56(6):1021–4.CrossRef Oefelein MG, et al. Reassessment of the definition of castrate levels of testosterone: implications for clinical decision making. Urology. 2000;56(6):1021–4.CrossRef
81.
go back to reference Jennbacken K, et al. N-cadherin increases after androgen deprivation and is associated with metastasis in prostate cancer. Endocr Relat Cancer. 2010;17(2):469–79.CrossRef Jennbacken K, et al. N-cadherin increases after androgen deprivation and is associated with metastasis in prostate cancer. Endocr Relat Cancer. 2010;17(2):469–79.CrossRef
82.
go back to reference Kleeberger W, et al. Roles for the stem cell associated intermediate filament Nestin in prostate cancer migration and metastasis. Cancer Res. 2007;67(19):9199–206.CrossRef Kleeberger W, et al. Roles for the stem cell associated intermediate filament Nestin in prostate cancer migration and metastasis. Cancer Res. 2007;67(19):9199–206.CrossRef
83.
go back to reference Wei Q, et al. Global analysis of differentially expressed genes in androgen-independent prostate cancer. Prostate Cancer Prostatic Dis. 2007;10(2):167–74.CrossRef Wei Q, et al. Global analysis of differentially expressed genes in androgen-independent prostate cancer. Prostate Cancer Prostatic Dis. 2007;10(2):167–74.CrossRef
84.
go back to reference Bhasin S, et al. Reference ranges for testosterone in men generated using liquid chromatography tandem mass spectrometry in a community-based sample of healthy nonobese young men in the Framingham Heart Study and applied to three geographically distinct cohorts. J Clin Endocrinol Metab. 2011;96(8):2430–9.CrossRef Bhasin S, et al. Reference ranges for testosterone in men generated using liquid chromatography tandem mass spectrometry in a community-based sample of healthy nonobese young men in the Framingham Heart Study and applied to three geographically distinct cohorts. J Clin Endocrinol Metab. 2011;96(8):2430–9.CrossRef
85.
go back to reference Gray A, et al. Age, disease, and changing sex hormone levels in middle-aged men: Results of the Massachusetts Male Aging Study. J Clin Endocrinol Metab. 1991;73(5):1016–25.CrossRef Gray A, et al. Age, disease, and changing sex hormone levels in middle-aged men: Results of the Massachusetts Male Aging Study. J Clin Endocrinol Metab. 1991;73(5):1016–25.CrossRef
86.
go back to reference Harman SM, et al. Longitudinal effects of aging on serum total and free testosterone levels in healthy men. Baltimore Longitudinal Study of Aging. J Clin Endocrinol Metab. 2001;86(2):724–31.CrossRef Harman SM, et al. Longitudinal effects of aging on serum total and free testosterone levels in healthy men. Baltimore Longitudinal Study of Aging. J Clin Endocrinol Metab. 2001;86(2):724–31.CrossRef
87.
go back to reference Brazhnik K, et al. Single-cell analysis reveals different age-related somatic mutation profiles between stem and differentiated cells in human liver. Sci Adv. 2020;6(5):eaax2659.CrossRef Brazhnik K, et al. Single-cell analysis reveals different age-related somatic mutation profiles between stem and differentiated cells in human liver. Sci Adv. 2020;6(5):eaax2659.CrossRef
88.
go back to reference Hopkins JF, et al. Mitochondrial mutations drive prostate cancer aggression. Nat Commun. 2017;8(1):656.CrossRef Hopkins JF, et al. Mitochondrial mutations drive prostate cancer aggression. Nat Commun. 2017;8(1):656.CrossRef
89.
go back to reference Conboy IM, et al. Notch-mediated restoration of regenerative potential to aged muscle. Science. 2003;302(5650):1575–7.CrossRef Conboy IM, et al. Notch-mediated restoration of regenerative potential to aged muscle. Science. 2003;302(5650):1575–7.CrossRef
90.
go back to reference Molofsky AV, et al. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature. 2006;443(7110):448–52.CrossRef Molofsky AV, et al. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature. 2006;443(7110):448–52.CrossRef
91.
go back to reference Crowell PD, et al. Expansion of luminal progenitor cells in the aging mouse and human prostate. Cell Rep. 2019;28(6):1499-510 e6.CrossRef Crowell PD, et al. Expansion of luminal progenitor cells in the aging mouse and human prostate. Cell Rep. 2019;28(6):1499-510 e6.CrossRef
92.
go back to reference Guo W, et al. Single-cell transcriptomics identifies a distinct luminal progenitor cell type in distal prostate invagination tips. Nat Genet. 2020;52(9):908–18.CrossRef Guo W, et al. Single-cell transcriptomics identifies a distinct luminal progenitor cell type in distal prostate invagination tips. Nat Genet. 2020;52(9):908–18.CrossRef
94.
go back to reference Crowell PD, et al. Distinct cell-types in the prostate share an aging signature suggestive of metabolic reprogramming. Am J Clin Exp Urol. 2020;8(4):140–51. Crowell PD, et al. Distinct cell-types in the prostate share an aging signature suggestive of metabolic reprogramming. Am J Clin Exp Urol. 2020;8(4):140–51.
95.
go back to reference Jiang J, et al. The role of prostatitis in prostate cancer: meta-analysis. PLoS ONE. 2013;8(12):e85179.CrossRef Jiang J, et al. The role of prostatitis in prostate cancer: meta-analysis. PLoS ONE. 2013;8(12):e85179.CrossRef
96.
go back to reference Perletti G, et al. The association between prostatitis and prostate cancer. Systematic review and meta-analysis. Arch Ital Urol Androl. 2017;89(4):259–65.CrossRef Perletti G, et al. The association between prostatitis and prostate cancer. Systematic review and meta-analysis. Arch Ital Urol Androl. 2017;89(4):259–65.CrossRef
97.
go back to reference Klein EA, et al. Vitamin E and the risk of prostate cancer: The Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA. 2011;306(14):1549–56.CrossRef Klein EA, et al. Vitamin E and the risk of prostate cancer: The Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA. 2011;306(14):1549–56.CrossRef
98.
go back to reference Platz EA, et al. A prospective study of chronic inflammation in benign prostate tissue and risk of prostate cancer: Linked PCPT and SELECT cohorts. Cancer Epidemiol Biomarkers Prev. 2017;26(10):1549–57.CrossRef Platz EA, et al. A prospective study of chronic inflammation in benign prostate tissue and risk of prostate cancer: Linked PCPT and SELECT cohorts. Cancer Epidemiol Biomarkers Prev. 2017;26(10):1549–57.CrossRef
99.
go back to reference Mohamad NV, et al. The relationship between circulating testosterone and inflammatory cytokines in men. Aging Male. 2019;22(2):129–40.CrossRef Mohamad NV, et al. The relationship between circulating testosterone and inflammatory cytokines in men. Aging Male. 2019;22(2):129–40.CrossRef
100.
go back to reference Trigunaite A, Dimo J, Jorgensen TN. Suppressive effects of androgens on the immune system. Cell Immunol. 2015;294(2):87–94.CrossRef Trigunaite A, Dimo J, Jorgensen TN. Suppressive effects of androgens on the immune system. Cell Immunol. 2015;294(2):87–94.CrossRef
101.
go back to reference Morrison C, Thornhill J, Gaffney E. The connective tissue framework in the normal prostate, BPH and prostate cancer: analysis by scanning electron microscopy after cellular digestion. Urol Res. 2000;28(5):304–7.CrossRef Morrison C, Thornhill J, Gaffney E. The connective tissue framework in the normal prostate, BPH and prostate cancer: analysis by scanning electron microscopy after cellular digestion. Urol Res. 2000;28(5):304–7.CrossRef
102.
go back to reference Provenzano PP, et al. Collagen density promotes mammary tumor initiation and progression. BMC Med. 2008;6:11.CrossRef Provenzano PP, et al. Collagen density promotes mammary tumor initiation and progression. BMC Med. 2008;6:11.CrossRef
103.
go back to reference Cunha GR, Donjacour AA, Sugimura Y. Stromal-epithelial interactions and heterogeneity of proliferative activity within the prostate. Biochem Cell Biol. 1986;64(6):608–14.CrossRef Cunha GR, Donjacour AA, Sugimura Y. Stromal-epithelial interactions and heterogeneity of proliferative activity within the prostate. Biochem Cell Biol. 1986;64(6):608–14.CrossRef
104.
go back to reference Jennbacken K, et al. The prostatic environment suppresses growth of androgen-independent prostate cancer xenografts: an effect influenced by testosterone. Prostate. 2009;69(11):1164–75.CrossRef Jennbacken K, et al. The prostatic environment suppresses growth of androgen-independent prostate cancer xenografts: an effect influenced by testosterone. Prostate. 2009;69(11):1164–75.CrossRef
105.
go back to reference Begley L, et al. CXCL12 overexpression and secretion by aging fibroblasts enhance human prostate epithelial proliferation in vitro. Aging Cell. 2005;4(6):291–8.CrossRef Begley L, et al. CXCL12 overexpression and secretion by aging fibroblasts enhance human prostate epithelial proliferation in vitro. Aging Cell. 2005;4(6):291–8.CrossRef
106.
go back to reference Hammarsten J, et al. A stage-dependent link between metabolic syndrome components and incident prostate cancer. Nat Rev Urol. 2018;15(5):321–33.CrossRef Hammarsten J, et al. A stage-dependent link between metabolic syndrome components and incident prostate cancer. Nat Rev Urol. 2018;15(5):321–33.CrossRef
107.
go back to reference McGrowder DA, Jackson LA, Crawford TV. Prostate cancer and metabolic syndrome: is there a link? Asian Pac J Cancer Prev. 2012;13(1):1–13.CrossRef McGrowder DA, Jackson LA, Crawford TV. Prostate cancer and metabolic syndrome: is there a link? Asian Pac J Cancer Prev. 2012;13(1):1–13.CrossRef
108.
go back to reference Guay AT. The emerging link between hypogonadism and metabolic syndrome. J Androl. 2009;30(4):370–6.CrossRef Guay AT. The emerging link between hypogonadism and metabolic syndrome. J Androl. 2009;30(4):370–6.CrossRef
109.
go back to reference Fall K, et al. Diabetes mellitus and prostate cancer risk; a nationwide case-control study within PCBaSe Sweden. Cancer Epidemiol Biomarkers Prev. 2013;22(6):1102–9.CrossRef Fall K, et al. Diabetes mellitus and prostate cancer risk; a nationwide case-control study within PCBaSe Sweden. Cancer Epidemiol Biomarkers Prev. 2013;22(6):1102–9.CrossRef
110.
go back to reference Tsilidis KK, et al. Diabetes mellitus and risk of prostate cancer in the European Prospective Investigation into Cancer and Nutrition. Int J Cancer. 2015;136(2):372–81.CrossRef Tsilidis KK, et al. Diabetes mellitus and risk of prostate cancer in the European Prospective Investigation into Cancer and Nutrition. Int J Cancer. 2015;136(2):372–81.CrossRef
111.
go back to reference Kim YJ, et al. The association between metabolic syndrome and prostate-specific antigen levels. Int J Urol. 2008;15(10):905–9.CrossRef Kim YJ, et al. The association between metabolic syndrome and prostate-specific antigen levels. Int J Urol. 2008;15(10):905–9.CrossRef
112.
go back to reference Xu H, et al. Diabetes mellitus and prostate cancer risk of different grade or stage: a systematic review and meta-analysis. Diabetes Res Clin Pract. 2013;99(3):241–9.CrossRef Xu H, et al. Diabetes mellitus and prostate cancer risk of different grade or stage: a systematic review and meta-analysis. Diabetes Res Clin Pract. 2013;99(3):241–9.CrossRef
113.
go back to reference Bianchini F, Kaaks R, Vainio H. Overweight, obesity, and cancer risk. Lancet Oncol. 2002;3(9):565–74.CrossRef Bianchini F, Kaaks R, Vainio H. Overweight, obesity, and cancer risk. Lancet Oncol. 2002;3(9):565–74.CrossRef
114.
go back to reference O’Malley RL, Taneja SS. Obesity and prostate cancer. Can J Urol. 2006;13(Suppl 2):11–7. O’Malley RL, Taneja SS. Obesity and prostate cancer. Can J Urol. 2006;13(Suppl 2):11–7.
115.
go back to reference Calle EE, et al. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med. 2003;348(17):1625–38.CrossRef Calle EE, et al. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med. 2003;348(17):1625–38.CrossRef
116.
go back to reference Pischon T, et al. Body size and risk of prostate cancer in the European prospective investigation into cancer and nutrition. Cancer Epidemiol Biomarkers Prev. 2008;17(11):3252–61.CrossRef Pischon T, et al. Body size and risk of prostate cancer in the European prospective investigation into cancer and nutrition. Cancer Epidemiol Biomarkers Prev. 2008;17(11):3252–61.CrossRef
117.
go back to reference Rodriguez C, et al. Body mass index, height, and prostate cancer mortality in two large cohorts of adult men in the United States. Cancer Epidemiol Biomarkers Prev. 2001;10(4):345–53. Rodriguez C, et al. Body mass index, height, and prostate cancer mortality in two large cohorts of adult men in the United States. Cancer Epidemiol Biomarkers Prev. 2001;10(4):345–53.
118.
go back to reference Chau CH, et al. Serum markers, obesity and prostate cancer risk: results from the prostate cancer prevention trial. Endocr Relat Cancer. 2022;29(2):99–109. Chau CH, et al. Serum markers, obesity and prostate cancer risk: results from the prostate cancer prevention trial. Endocr Relat Cancer. 2022;29(2):99–109.
119.
go back to reference Wang S, et al. Circulating IGF-1 promotes prostate adenocarcinoma via FOXO3A/BIM signaling in a double-transgenic mouse model. Oncogene. 2019;38(36):6338–53.CrossRef Wang S, et al. Circulating IGF-1 promotes prostate adenocarcinoma via FOXO3A/BIM signaling in a double-transgenic mouse model. Oncogene. 2019;38(36):6338–53.CrossRef
120.
go back to reference Laurent V, et al. Periprostatic adipocytes act as a driving force for prostate cancer progression in obesity. Nat Commun. 2016;7:10230.CrossRef Laurent V, et al. Periprostatic adipocytes act as a driving force for prostate cancer progression in obesity. Nat Commun. 2016;7:10230.CrossRef
121.
go back to reference Guerard A, et al. The chemokine receptor CCR3 is potentially involved in the homing of prostate cancer cells to bone: Implication of bone-marrow adipocytes. Int J Mol Sci. 2021;22(4):1994. Guerard A, et al. The chemokine receptor CCR3 is potentially involved in the homing of prostate cancer cells to bone: Implication of bone-marrow adipocytes. Int J Mol Sci. 2021;22(4):1994.
122.
go back to reference Carter JL, Coletti RJ, Harris RP. Quantifying and monitoring overdiagnosis in cancer screening: a systematic review of methods. BMJ. 2015;350:g7773.CrossRef Carter JL, Coletti RJ, Harris RP. Quantifying and monitoring overdiagnosis in cancer screening: a systematic review of methods. BMJ. 2015;350:g7773.CrossRef
123.
go back to reference Hugosson J, et al. A 16-yr follow-up of the European randomized study of screening for prostate cancer. Eur Urol. 2019;76(1):43–51.CrossRef Hugosson J, et al. A 16-yr follow-up of the European randomized study of screening for prostate cancer. Eur Urol. 2019;76(1):43–51.CrossRef
124.
go back to reference Schroder FH, et al. Screening and prostate cancer mortality: Results of the European Randomised Study of Screening for Prostate Cancer (ERSPC) at 13 years of follow-up. Lancet. 2014;384(9959):2027–35.CrossRef Schroder FH, et al. Screening and prostate cancer mortality: Results of the European Randomised Study of Screening for Prostate Cancer (ERSPC) at 13 years of follow-up. Lancet. 2014;384(9959):2027–35.CrossRef
125.
go back to reference Andriole GL, et al. Prostate cancer screening in the randomized Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial: Mortality results after 13 years of follow-up. J Natl Cancer Inst. 2012;104(2):125–32.CrossRef Andriole GL, et al. Prostate cancer screening in the randomized Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial: Mortality results after 13 years of follow-up. J Natl Cancer Inst. 2012;104(2):125–32.CrossRef
126.
go back to reference Grenabo Bergdahl A, et al. Role of magnetic resonance imaging in prostate cancer screening: A pilot study within the Goteborg randomised screening trial. Eur Urol. 2016;70(4):566–73.CrossRef Grenabo Bergdahl A, et al. Role of magnetic resonance imaging in prostate cancer screening: A pilot study within the Goteborg randomised screening trial. Eur Urol. 2016;70(4):566–73.CrossRef
127.
go back to reference Gronberg H, et al. Prostate cancer diagnostics using a combination of the Stockholm3 blood test and multiparametric magnetic resonance imaging. Eur Urol. 2018;74(6):722–8.CrossRef Gronberg H, et al. Prostate cancer diagnostics using a combination of the Stockholm3 blood test and multiparametric magnetic resonance imaging. Eur Urol. 2018;74(6):722–8.CrossRef
128.
go back to reference Nordstrom T, et al. Prostate cancer screening using a combination of risk-prediction, MRI, and targeted prostate biopsies (STHLM3-MRI): a prospective, population-based, randomised, open-label, non-inferiority trial. Lancet Oncol. 2021;22(9):1240–9.CrossRef Nordstrom T, et al. Prostate cancer screening using a combination of risk-prediction, MRI, and targeted prostate biopsies (STHLM3-MRI): a prospective, population-based, randomised, open-label, non-inferiority trial. Lancet Oncol. 2021;22(9):1240–9.CrossRef
129.
go back to reference Hernandez J, Thompson IM. Prostate-specific antigen: a review of the validation of the most commonly used cancer biomarker. Cancer. 2004;101(5):894–904.CrossRef Hernandez J, Thompson IM. Prostate-specific antigen: a review of the validation of the most commonly used cancer biomarker. Cancer. 2004;101(5):894–904.CrossRef
130.
go back to reference Rhoden EL, Morgentaler A. Testosterone replacement therapy in hypogonadal men at high risk for prostate cancer: results of 1 year of treatment in men with prostatic intraepithelial neoplasia. J Urol. 2003;170(6 Pt 1):2348–51.CrossRef Rhoden EL, Morgentaler A. Testosterone replacement therapy in hypogonadal men at high risk for prostate cancer: results of 1 year of treatment in men with prostatic intraepithelial neoplasia. J Urol. 2003;170(6 Pt 1):2348–51.CrossRef
131.
go back to reference Amadi C, Green KI, Odum EP. Is serum PSA a predictor of male hypogonadism? Testing the hypothesis. Arch Endocrinol Metab. 2021;65(2):144–51. Amadi C, Green KI, Odum EP. Is serum PSA a predictor of male hypogonadism? Testing the hypothesis. Arch Endocrinol Metab. 2021;65(2):144–51.
132.
go back to reference Rastrelli G, et al. Serum PSA as a predictor of testosterone deficiency. J Sex Med. 2013;10(10):2518–28.CrossRef Rastrelli G, et al. Serum PSA as a predictor of testosterone deficiency. J Sex Med. 2013;10(10):2518–28.CrossRef
133.
go back to reference Morgentaler A, Rhoden EL. Prevalence of prostate cancer among hypogonadal men with prostate-specific antigen levels of 4.0 ng/mL or less. Urology. 2006;68(6):1263–7.CrossRef Morgentaler A, Rhoden EL. Prevalence of prostate cancer among hypogonadal men with prostate-specific antigen levels of 4.0 ng/mL or less. Urology. 2006;68(6):1263–7.CrossRef
134.
go back to reference Coward RM, Simhan J, Carson CC 3rd. Prostate-specific antigen changes and prostate cancer in hypogonadal men treated with testosterone replacement therapy. BJU Int. 2009;103(9):1179–83. rd. . ).CrossRef Coward RM, Simhan J, Carson CC 3rd. Prostate-specific antigen changes and prostate cancer in hypogonadal men treated with testosterone replacement therapy. BJU Int. 2009;103(9):1179–83. rd. . ).CrossRef
135.
go back to reference Salonia A, et al. European association of urology guidelines on sexual and reproductive health–2021 update: Male sexual dysfunction. Eur Urol. 2021;80(3):333–57.CrossRef Salonia A, et al. European association of urology guidelines on sexual and reproductive health–2021 update: Male sexual dysfunction. Eur Urol. 2021;80(3):333–57.CrossRef
136.
go back to reference Banerjee PP, et al. Androgen action in prostate function and disease. Am J Clin Exp Urol. 2018;6(2):62–77. Banerjee PP, et al. Androgen action in prostate function and disease. Am J Clin Exp Urol. 2018;6(2):62–77.
137.
go back to reference Marberger M, et al. Relationship among serum testosterone, sexual function, and response to treatment in men receiving dutasteride for benign prostatic hyperplasia. J Clin Endocrinol Metab. 2006;91(4):1323–8.CrossRef Marberger M, et al. Relationship among serum testosterone, sexual function, and response to treatment in men receiving dutasteride for benign prostatic hyperplasia. J Clin Endocrinol Metab. 2006;91(4):1323–8.CrossRef
138.
go back to reference Cui Y, Zhang Y. The effect of androgen-replacement therapy on prostate growth: a systematic review and meta-analysis. Eur Urol. 2013;64(5):811–22.CrossRef Cui Y, Zhang Y. The effect of androgen-replacement therapy on prostate growth: a systematic review and meta-analysis. Eur Urol. 2013;64(5):811–22.CrossRef
139.
go back to reference Shigehara K, et al. Androgen replacement therapy contributes to improving lower urinary tract symptoms in patients with hypogonadism and benign prostate hypertrophy: a randomised controlled study. Aging Male. 2011;14(1):53–8.CrossRef Shigehara K, et al. Androgen replacement therapy contributes to improving lower urinary tract symptoms in patients with hypogonadism and benign prostate hypertrophy: a randomised controlled study. Aging Male. 2011;14(1):53–8.CrossRef
140.
go back to reference Debruyne FM, et al. Testosterone treatment is not associated with increased risk of prostate cancer or worsening of lower urinary tract symptoms: prostate health outcomes in the Registry of Hypogonadism in Men. BJU Int. 2017;119(2):216–24.CrossRef Debruyne FM, et al. Testosterone treatment is not associated with increased risk of prostate cancer or worsening of lower urinary tract symptoms: prostate health outcomes in the Registry of Hypogonadism in Men. BJU Int. 2017;119(2):216–24.CrossRef
141.
go back to reference Santella C, et al. Testosterone replacement therapy and the risk of prostate cancer in men with late-onset hypogonadism. Am J Epidemiol. 2019;188(9):1666–73.CrossRef Santella C, et al. Testosterone replacement therapy and the risk of prostate cancer in men with late-onset hypogonadism. Am J Epidemiol. 2019;188(9):1666–73.CrossRef
142.
go back to reference Loeb S, et al. Testosterone replacement therapy and risk of favorable and aggressive prostate cancer. J Clin Oncol. 2017;35(13):1430–6.CrossRef Loeb S, et al. Testosterone replacement therapy and risk of favorable and aggressive prostate cancer. J Clin Oncol. 2017;35(13):1430–6.CrossRef
143.
go back to reference Mottet N, et al. EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer–2020 update. Part 1: Screening, diagnosis, and local treatment with curative intent. Eur Urol. 2021;79(2):243–62.CrossRef Mottet N, et al. EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer–2020 update. Part 1: Screening, diagnosis, and local treatment with curative intent. Eur Urol. 2021;79(2):243–62.CrossRef
144.
go back to reference Ory J, et al. Testosterone therapy in patients with treated and untreated prostate cancer: impact on oncologic outcomes. J Urol. 2016;196(4):1082–9.CrossRef Ory J, et al. Testosterone therapy in patients with treated and untreated prostate cancer: impact on oncologic outcomes. J Urol. 2016;196(4):1082–9.CrossRef
145.
go back to reference Morgentaler A, et al. Testosterone therapy in men with untreated prostate cancer. J Urol. 2011;185(4):1256–60.CrossRef Morgentaler A, et al. Testosterone therapy in men with untreated prostate cancer. J Urol. 2011;185(4):1256–60.CrossRef
146.
go back to reference Kacker R, et al. Can testosterone therapy be offered to men on active surveillance for prostate cancer? Preliminary results. Asian J Androl. 2016;18(1):16–20.CrossRef Kacker R, et al. Can testosterone therapy be offered to men on active surveillance for prostate cancer? Preliminary results. Asian J Androl. 2016;18(1):16–20.CrossRef
147.
go back to reference Kim M, Byun SS, Hong SK. Testosterone replacement therapy in men with untreated or treated prostate cancer: Do we have enough evidences? World J Mens Health. 2021;39(4):705–23.CrossRef Kim M, Byun SS, Hong SK. Testosterone replacement therapy in men with untreated or treated prostate cancer: Do we have enough evidences? World J Mens Health. 2021;39(4):705–23.CrossRef
148.
go back to reference Kaufman JM, Graydon RJ. Androgen replacement after curative radical prostatectomy for prostate cancer in hypogonadal men. J Urol. 2004;172(3):920–2.CrossRef Kaufman JM, Graydon RJ. Androgen replacement after curative radical prostatectomy for prostate cancer in hypogonadal men. J Urol. 2004;172(3):920–2.CrossRef
149.
go back to reference Khera M, et al. Testosterone replacement therapy following radical prostatectomy. J Sex Med. 2009;6(4):1165–70.CrossRef Khera M, et al. Testosterone replacement therapy following radical prostatectomy. J Sex Med. 2009;6(4):1165–70.CrossRef
150.
go back to reference Pastuszak AW, et al. Testosterone replacement therapy in patients with prostate cancer after radical prostatectomy. J Urol. 2013;190(2):639–44.CrossRef Pastuszak AW, et al. Testosterone replacement therapy in patients with prostate cancer after radical prostatectomy. J Urol. 2013;190(2):639–44.CrossRef
151.
go back to reference Sarosdy MF. Testosterone replacement for hypogonadism after treatment of early prostate cancer with brachytherapy. Cancer. 2007;109(3):536–41.CrossRef Sarosdy MF. Testosterone replacement for hypogonadism after treatment of early prostate cancer with brachytherapy. Cancer. 2007;109(3):536–41.CrossRef
152.
go back to reference Pastuszak AW, et al. Testosterone replacement therapy in the setting of prostate cancer treated with radiation. Int J Impot Res. 2013;25(1):24–8.CrossRef Pastuszak AW, et al. Testosterone replacement therapy in the setting of prostate cancer treated with radiation. Int J Impot Res. 2013;25(1):24–8.CrossRef
153.
go back to reference Heemers HV, Tindall DJ. Androgen receptor (AR) coregulators: a diversity of functions converging on and regulating the AR transcriptional complex. Endocr Rev. 2007;28(7):778–808.CrossRef Heemers HV, Tindall DJ. Androgen receptor (AR) coregulators: a diversity of functions converging on and regulating the AR transcriptional complex. Endocr Rev. 2007;28(7):778–808.CrossRef
154.
go back to reference Gim HJ, et al. Conformational dynamics of androgen receptors bound to agonists and antagonists. Sci Rep. 2021;11(1):15887.CrossRef Gim HJ, et al. Conformational dynamics of androgen receptors bound to agonists and antagonists. Sci Rep. 2021;11(1):15887.CrossRef
155.
go back to reference Denmeade SR, et al. Prostate-specific antigen-activated thapsigargin prodrug as targeted therapy for prostate cancer. J Natl Cancer Inst. 2003;95(13):990–1000.CrossRef Denmeade SR, et al. Prostate-specific antigen-activated thapsigargin prodrug as targeted therapy for prostate cancer. J Natl Cancer Inst. 2003;95(13):990–1000.CrossRef
156.
go back to reference Gustavsson H, Welen K, Damber JE. Transition of an androgen-dependent human prostate cancer cell line into an androgen-independent subline is associated with increased angiogenesis. Prostate. 2005;62(4):364–73.CrossRef Gustavsson H, Welen K, Damber JE. Transition of an androgen-dependent human prostate cancer cell line into an androgen-independent subline is associated with increased angiogenesis. Prostate. 2005;62(4):364–73.CrossRef
157.
go back to reference Joly-Pharaboz MO, et al. Androgen inhibits the growth of carcinoma cell lines established from prostate cancer xenografts that escape androgen treatment. J Steroid Biochem Mol Biol. 2008;111(1–2):50–9.CrossRef Joly-Pharaboz MO, et al. Androgen inhibits the growth of carcinoma cell lines established from prostate cancer xenografts that escape androgen treatment. J Steroid Biochem Mol Biol. 2008;111(1–2):50–9.CrossRef
158.
go back to reference Bhasin S, Jasuja R. Selective androgen receptor modulators as function promoting therapies. Curr Opin Clin Nutr Metab Care. 2009;12(3):232–40.CrossRef Bhasin S, Jasuja R. Selective androgen receptor modulators as function promoting therapies. Curr Opin Clin Nutr Metab Care. 2009;12(3):232–40.CrossRef
Metadata
Title
Androgens, aging, and prostate health
Authors
Karin Welén
Jan-Erik Damber
Publication date
24-06-2022
Publisher
Springer US
Published in
Reviews in Endocrine and Metabolic Disorders / Issue 6/2022
Print ISSN: 1389-9155
Electronic ISSN: 1573-2606
DOI
https://doi.org/10.1007/s11154-022-09730-z

Other articles of this Issue 6/2022

Reviews in Endocrine and Metabolic Disorders 6/2022 Go to the issue

ReviewPaper

Preface

Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.