World J Mens Health. 2024;42:e34. Forthcoming. English.
Published online Mar 14, 2024.
Copyright © 2024 Korean Society for Sexual Medicine and Andrology
Original Article

Secondary Cancer after Androgen Deprivation Therapy in Prostate Cancer: A Nationwide Study

Jae Heon Kim,1 Gi Hwan Bae,2 Jaehun Jung,2,3 and Tae Il Noh4
    • 1Department of Urology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea.
    • 2Artificial Intelligence and Big-Data Convergence Center, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea.
    • 3Department of Preventive Medicine, Gachon University College of Medicine, Incheon, Korea.
    • 4Department of Urology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea.
Received August 21, 2023; Revised October 15, 2023; Accepted December 05, 2023.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Purpose

Androgen signaling is associated with various secondary cancer, which could be promising for potential treatment using androgen deprivation therapy (ADT). This study investigated whether ADT use was associated with secondary cancers other than prostate cancer in a nationwide population-based cohort.

Materials and Methods

A total, 278,434 men with newly diagnosed prostate cancer between January 1, 2002 and December 31, 2017 were identified. After applying the exclusion criteria, 170,416 men were enrolled. The study cohort was divided into ADT and non-ADT groups by individual matching followed by propensity score matching (PSM). Study outcomes were incidence of all male cancers. Cox proportional hazard regression models were used to estimate adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) of events.

Results

During a median follow-up of 4.5 years, a total of 11,059 deaths (6,329 in the ADT group and 4,730 in the non-ADT group) after PSM were found. After PSM, the overall all-cause of secondary cancer incidence risk of the ADT group was higher than that of the non-ADT group (HR: 1.312, 95% CI: 1.23–1.36; adjusted HR: 1.344, 95% CI: 1.29–1.40). The ADT group showed higher risk of overall brain and other central nervous system (CNS) cancer-specific incidence than the non-ADT group (adjusted HR: 1.648, 95% CI: 1.21–2.24). The ADT group showed lower risks of overall cancer-specific incidence for stomach, colon/rectum, liver/inflammatory bowel disease (IBD), gall bladder/extrahepatic bile duct, lung, bladder, and kidney cancers than the non-ADT group. When the duration of ADT was more than 2 years of ADT, the ADT group showed higher risk of cancer-specific incidence for brain and other CNS cancers but lower risk of cancer-specific incidence for liver/IBD and lung cancers than the non-ADT group.

Conclusions

This study demonstrates that ADT could affect cancer-specific incidence for various cancers.

Keywords
Cohort studies; Prostatic neoplasms; Receptors, androgen; Survival analysis

INTRODUCTION

Androgens play diverse roles in the body, encompassing the development of male secondary sex characteristics, erythropoiesis, body metabolism, bone density, and libido [1]. Normal androgen activity relies on androgens signaling through their receptor, the androgen receptor (AR), leading to the transcription of AR target genes.

Recent research has increasingly focused on androgens due to their involvement in various diseases, including metabolic syndrome, diabetes, cardiovascular disease, osteoporosis, dementia, and, notably, late onset hypogonadism in males [2]. This emphasis has also extended to oncological disorders, spurring a multitude of studies that investigate the role of AR expression in prostate cancer and other malignancies, such as breast cancer, lung cancer, and hepatobiliary cancers [3, 4]. Initial epidemiological observations revealed significant differences in cancer incidence and survival between sexes [5, 6], suggesting possible pathophysiological roles of androgens or AR expression, besides other sex hormones.

AR signaling is involved in various cancer types, similar to EGFR, HER2/Neu, AKT/mTOR/PI3K, and Wnt pathways [7, 8, 9, 10]. Traditionally, the central role of AR expression in the development and progression of prostate cancer is well-established [11]. Furthermore, the expression of AR target genes is highly relevant to androgen deprivation therapy (ADT) status [9]. Beyond prostate cancer, the involvement of AR in other malignancies has also been investigated, offering prospects for potential treatments involving antiandrogen therapies [12]. Recent studies also suggest that AR, as a prognostic marker in breast cancer survival, can affect estrogen receptor-dependent transcription [13, 14]. Consequently, strategies for treating breast cancer have shifted towards blocking AR signaling [15], with ongoing clinical trials, including phase 2 and phase 3 trials [4]. Investigations have hinted at a possible role of AR expression in cancer growth and drug resistance, not limited to prostate or breast cancer [3, 4]. However, few clinical studies have directly linked AR expression to cancer development, often due to challenges in accurately measuring AR expression and interpreting androgen activity based on serum testosterone levels.

The American Heart Association, in an observational study, linked ADT to an increased risk of cardiovascular events [16]. Therefore, researchers are now studying how ADT affects various diseases, with a particular focus on cardiovascular risks. Some new research suggests that ADT may harm the blood vessels by causing oxidative stress [17]. While ADT might impact cancer incidence by affecting cell growth and death through androgen signaling pathways, there isn't much ongoing research on this aspect.

Our study is based on the hypothesis that androgen signaling is associated with the development of secondary cancers, which holds promise for potential treatment using ADT in various malignancies. In this study, we investigated the pleiotropic role of androgen deprivation in different cancers, except for gynecological cancer.

MATERIALS AND METHODS

1. Database and ethics

In South Korea, the National Health Insurance System (NHIS) is a mandatory universal health coverage system that covers over 95% of the Korean population. National Health Insurance Services data are generated from health insurance claims as part of the reimbursement process for healthcare services under the NHIS in Korea. These data included age, sex, healthcare utilization (clinic, hospital, and emergency department visits), diagnoses coded according to the International Classification of Diseases and Related Health Problems, 10th edition (ICD-10), as well as prescriptions and procedures covered by the NHIS.

The study complied with the Declaration of Helsinki. The Institutional Review Board of Gachon University Gil Medical Center approved the research protocol (GCIRB2020-128). Informed consent was deemed unnecessary, given that data from all subjects were anonymized by NHIS in keeping with data protection recommendations.

2. Study cohorts

Claims data between January 1, 2002 and May 31, 2017 were retrieved from the NHIS database. A total of 218,203 men with prostate cancer (C61·0) were identified between January 1, 2008 and December 31, 2017, after randomly selecting 50% from the 556,868 men with prostate cancer (C61·0) during same period. Among these, 218,203 male patients had newly diagnosed prostate cancer. Patients with a previous history of cancer, patients under 40 years old, those with missing data, and those who initiated ADT more than 2 years after prostate cancer diagnosis were excluded to eliminate the possibility that ADT was not due to primary prostate cancer. Finally, a total of 170,416 patients were included for analysis in the present study (Fig. 1). The study cohort was divided into ADT and non-ADT groups. The ADT group comprised 15,672 subjects. The non-ADT group comprised 41,734 subjects. The cumulative dose of ADT was calculated as the sum of the action periods for each ADT preparation, and the duration of ADT was classified into three years.

Fig. 1
Deposition flow. ADT: androgen deprivation therapy, CCI: Charlson comorbidity index.

3. Outcomes and covariates

The ICD-10 codes were used to identify diagnoses. The primary outcome was overall all cause incidence of secondary cancer. The secondary outcome was cancer-specific incidence beside prostate cancer after ADT (Supplement Table 1). Death event was investigated at least 6 months after ADT exposure (ranging from 6 months to 2 years or more than 2 years). The ADT group was defined as the use of at least two times of prescription of gonadotropin-releasing hormone (GnRH) agonist or antagonist or antiandrogen drugs (Supplement Table 2) after the diagnosis of prostate cancer or bilateral orchiectomy after the diagnosis of prostate cancer. ADT included the administration of GnRH agonists (leuprolide, goserelin, and triptorelin), oral antiandrogens (cyproterone acetate, flutamide, and bicalutamide), bilateral orchiectomy and estrogens (estramustine), and undergoing bilateral orchiectomy. Covariates included 5-alpha-reductase inhibitors (Finasteride, Dutasteride) and new signaling androgen inhibitors (Enzalutamide, Abiraterone).

4. Statistical analysis

Baseline characteristics were compared according to ADT exposure. Continuous variables are expressed as means±standard deviation. Since multiple testing was performed between subgroups, we set a robust cutoff of p<0.01 for statistical significance. Before propensity score matching (PSM), individual matching by age was performed to reduce sample size of the non-ADT group (1:3 perfect matching). PSM was performed by age, Charlson comorbidity index (CCI), income level, health care utilization, region (metropolitan vs. non-metropolitan), and follow-up time.

After PSM, Cox proportional hazards models were used to determine the risk of death according to ADT and the incidence of all-cause secondary cancer. Individual cancer-specific incidence was reported in terms of hazard ratios (HRs) and 95% confidence intervals (CIs). Adjusted HR was defined considering significant variables during PSM. SAS version 9.4 (SAS Institute Inc.) and R version 3.5.2 (R Foundation for Statistical Computing) software programs were used for all statistical analyses. Cox proportional hazards included prematching covariates such as age, CCI, medications, and chemotherapy, as well as post-matching factors like income level, medical service utilization, medications, and chemotherapy.

RESULTS

1. Study characteristics

A total of 170,416 men between January 1, 2002 and December 31, 2017 were enrolled. After perfect matching, the ADT group included 15,672 (age, 72.76±8.29 years) and the non-ADT group included 41,734 (age, 71.73±8.12 years). After prostate-specific antigen (PSA) matching, the ADT group included 15,381 (age, 72.8±8.07 years) and the non-ADT group included 15,381 (age, 72.8±8.07 years). Before PSM matching, age at diagnosis, age distribution, CCI, income level, residence area (metropolitan/non-metropolitan), and medical service utilization showed significant differences between ADT and non-ADT groups. After PS matching, all covariates except income level and inpatient medical service utilization were balanced between ADT and non-ADT groups (Table 1).

Table 1
Characteristics of patients before and after propensity score matching

2. Overall incidence of secondary cancer between ADT and non-ADT groups

During a median follow-up of 4.5 years, a total of 18,069 deaths (6,467 in the ADT group and 11,632 in the non-ADT group) were found. After PSA matching, there were a total of 11,059 developments of secondary cancer (6,329 in the ADT group and 4,730 in the non-ADT group) (Table 2 and Supplement Table 3). After PSM, the ADT group had higher overall all-cause of secondary cancer risk than the non-ADT group (HR: 1.312, 95% CI: 1.26–1.36; adjusted HR: 1.344, 95% CI: 1.29–1.40). Survival analysis showed difference in cumulative incidence of secondary cancer between ADT and non-ADT groups (Fig. 2). Overall all-cause of secondary cancer risk of the ADT group according to duration of ADT exposure showed a decreasing pattern. However, it was still higher than that of the non-ADT group (adjusted HR: 1.764, 95% CI: 1.53–2.03 after exposure for five months, adjusted HR: 1.336, 95% CI: 1.27–1.41 after exposure for 2 years) (Table 3).

Fig. 2
Overall all cause of secondary cancer incidence before (A) and after (B) propensity score matching. ADT: androgen deprivation therapy.

Table 2
Overall all cause of secondary cancer incidence and individual overall cancer-specific incidence

Table 3
Individual cancer-specific incidence according to duration of androgen deprivation therapy exposure

3. Cancer-specific incidence between ADT and non-ADT groups

Table 2 and Supplement Table 3 show individual cancer-specific incidence risk of the ADT group compared with the non-ADT group. Overall brain and other central nervous system (CNS) cancer-specific incidence of the ADT group was higher risk than that of the non-ADT group both before and after PSM (HR: 1.656, 95% CI: 1.22–2.25; adjusted HR: 1.648, 95% CI: 1.21–2.24) (Table 2). The ADT group showed lower risk of overall cancer-specific incidence of stomach, colon/rectum, liver/inflammatory bowel disease (IBD), gall bladder/extrahepatic bile duct, lung, bladder, and kidney cancers that the non-ADT group (adjusted HRs: 0.826 [95% CI: 0.74–0.93], 0.695 [95% CI: 0.63–0.76], 0.542 [95% CI: 0.49–0.60], 0.752 [95% CI: 0.61–0.93], 0.697 [95% CI: 0.64–0.76], 0.767 [95% CI: 0.69–0.85], and 0.583 [95% CI: 0.35–0.98], respectively) (Table 2).

When the duration of ADT was between 6 months and 2 years, cancer-specific incidence risk for brain and other CNS cancers of the ADT group was higher risk than that of the non-ADT group both before and after PSM (HR: 2.721, 95% CI: 1.37–5.41; adjusted HR: 2.683, 95% CI: 1.35–5.34) (Table 3). However, in the same duration range, the ADT group showed lower risk of cancer-specific incidence for liver/IBD, lung, and kidney cancers than the non-ADT group (adjusted HR: 0.633, 95% CI: 0.51–0.78; 0.724, 95% CI: 0.59–0.88; and 0.235, 95% CI: 0.07–0.83; respectively) (Table 3). When the duration was more than 2 years, the ADT group showed higher risk cancer-specific incidence risk for brain and other CNS cancers than the non-ADT group in both before and after PSM (HR: 1.589, 95% CI: 1.04–2.44; adjusted HR: 1.575, 95% CI: 1.03–2.42) (Table 3). However, in the same duration range, the ADT group showed lower risk of cancer-specific incidence for liver/IBD and lung cancers than the non-ADT group (adjusted HR: 0.853, 95% CI: 0.74–0.98 and adjusted HR: 0.878, 95% CI: 0.77–0.99, respectively) (Table 3). Survival analysis showed different patterns among brain and other CNS cancer, liver/IBD cancer, and lung cancer (Fig. 3). Individual cancer-specific incidence according to the duration of ADT before PSM can be seen in Supplement Table 4.

Fig. 3
Overall cancer-specific incidence after propensity score matching. Brain and central nervous system cancers (A), liver and intrahepatic ductal cancers (B), and lung cancer (C). ADT: androgen deprivation therapy.

DISCUSSION

Our findings indicate a positive association between ADT in the treatment of prostate cancer and the risk of developing brain and other CNS cancers, while revealing an inverse association with the risk of liver, colorectal, and lung cancers. To the best of our knowledge, this is the first clinical observation study suggesting that ADT could be a risk factor for various cancer types.

As the role of AR-driven transcriptional activities in prostate cancer has been well-established, recent studies have investigated its role in transcription and genomic stability across various cancers, with positive results [18]. However, the expression and activation of the AR in various cancers are not yet fully understood due to heterogeneous results attributable to clinical factors such as tumor type and stage [3, 19].

Recently, Hu et al [12] have described the role of AR in the development and outcome of various tumor types by pan-cancer analysis of the Cancer Genome Atlas (TCGA)-Clinical Data Resource. They reported that correlations of AR expression with incidence and survival varied across tumor types. AR expression was positively associated with stomach cancer and low-grade glioma. In contrast, it showed inverse associations with kidney cancer, acute myeloid leukemia, liver cancer, ovarian cancer, and melanoma [12]. Although this pan-cancer analysis was a precedent at the genomic level, its data have limitations for clinical applications due to inconsistent expression profiles between mRNA and protein levels. Moreover, the TCGA data lacked lung cancer data. Such data have been upgraded since 2014. Furthermore, interpretation of the TCGA clinical data is onerous due to challenges associated with analyzing and managing the complexity of huge data sets. Although we could not assess the expression at the genomic level, our findings represent the first real clinical data that could provide useful insight for future preclinical and clinical investigations regarding the role of AR in cancer therapy using ADT.

Ongoing clinical trials that are exploring AR-focused therapies for various cancers including hepatocellular cancer, bladder cancer, pancreatic cancer, AR-positive salivary cancer, renal cell cancer, and mantle cell lymphoma could provide further insight into the AR’s involvement in malignancies [3, 4]. Such clinical trials have been the basis of the hypothesis that AR expression could be associated with cancer incidence and aggravation not only in prostate cancer, but also in other cancer types. While our study couldn't directly establish this association, it did reveal intriguing patterns, including a potential inverse link with stomach cancer. AR expression has previously been associated with the incidence of liver cancer including hepatocellular carcinoma [6, 20], with several studies pointing to associations with worse clinical stages and poor outcomes [6, 21, 22]. Moreover, many studies have shown a positive association between AR signaling and increased risk of hepatitis B and C, known risk factors for hepatocellular cancer [23]. AR expression has been confirmed in both normal human lung cells and lung cancer cells [24, 25]. AR stimulation by androgens can stimulate the growth of small cell and non-small cell lung cancer lines in vitro [26]. Recently, Grant et al [27] have reported that androgen pathway blockade can significantly improve the survival of lung cancer patients.

Inconsistent findings between previous studies, clinical trials, and our observations suggest that there should be focus on the dual or complex action of AR depending on clinical cancer stage. These discrepancies could be due to small sampling sizes and different analysis protocols employed in previous studies. Most studies, including our current study, could not show clinical staging, tumor size, tumor complexity, or intervention protocols. Thus, such inconsistent findings draw the focus towards a possible dual function of AR according to clinical stage. However, our study could show cumulative risk of AR expression by ADT in secondary cancer risk.

The role of AR is different between early stages of cancer development and advanced progression stages in non-prostate cancers, especially in hepatocellular carcinoma and lung cancer, as implied by earlier evidence on this topic. Currently, there are no definitive results pointing to a role of AR as a cancer promoter, protective effector, or suppressor of cancer progression across cancer types and stages [3, 12]. Recently, Ma et al [21] have reported that AR expression is upregulated only in tumors smaller than 3 cm and that AR expression is absent in advanced and severe liver cancer tissues. They also demonstrated the importance of AR using an in vivo model of L-ARKO mice lacking hepatic AR treated with a carcinogen. The absence of hepatic AR resulted in poor survival, increased aggravation, and metastasis. It suggests that the key mechanism involves suppression of NF-κB and p38 phosphorylation. Regarding lung cancer, Nishio et al [28] have reported that patients with advanced or recurrent non-small cell lung cancer treated with an epidermal growth factor receptor tyrosine kinase inhibitor show lower androgen levels after treatment. This implies that a low androgen level could be a negative prognostic marker after treatment [28]. Grant et al [27] have also reported that high Ki67 and low AR expression are related to a higher risk of death and recurrence of non-small cell lung cancer.

Although our study showed an association between AR and secondary cancer development using ADT in a large cohort, several limitations should be taken into account for a better understanding of our results. First, our study did not directly measure serum testosterone level or AR expression. However, direct measurement of serum testosterone has been shown to be less reliable due to its wide range normal value and its dependence on the biological cycle [29]. Moreover, direct measurement of AR expression is expensive and complex. It is only possible in an experimental setting. However, our previous studies [30, 31] have shown that PSA with normal range could be a surrogate marker for serum testosterone using meta-analysis. It also could be used a predictive marker for subclinical and clinical cardiovascular diseases, indicative of androgen activity. Furthermore, our data could not present clinical staging, complicating objective interpretation, or relevance of clinical results. However, the clinical stage of prostate cancer itself could net affect the risk of secondary cancer development. This study population consisted of Korean men. It is well known that androgen activity is affected by ethnicity. Therefore, our findings might not be generalizable to other age groups, populations with a higher prevalence of comorbidities, or other race/ethnic groups. In addition, we could not explain the higher overall incidence of secondary cancer in the ADT group than in the non-ADT group. Lastly, despite adjustments for various medical service utilizations, this study, which used health insurance claims data, could not fully account for differences in follow-up intensity based on ADT status.

It is observed that ADT may be related to an increased or decreased risk of specific malignancies. The variations in risk suggest that the influence of ADT on different types of cancers is complex and multifaceted. While we must interpret these results cautiously, our study suggests that AR expression may play a role in cancer development and progression, with the impact varying based on the specific cancer type. In the future, rigorous randomized controlled trials (RCTs) and studies on mRNA and AR protein levels are crucial to understand the relationship between various ADT types and secondary cancer. Furthermore, the need for extended real-world evidence assessing the effects of ADT types on secondary cancers is paramount.

CONCLUSIONS

Our study demonstrated a positive association between the use of ADT in the treatment of prostate cancer and the risk of developing brain and other CNS cancers. Simultaneously, it revealed an inverse association between ADT and the risk of liver, colorectal, and lung cancers. This study supports the dual role of AR in various cancer types. Further studies including RCTs and genomic analysis are needed to elucidate the relationship between ADT and pan-cancer clinical features.

Supplementary Materials

Supplementary materials can be found via https://doi.org/10.5534/wjmh.230237

Supplement Table 1

Cancer codes for analysis

Click here to view.(70K, pdf)

Supplement Table 2

Pharmaceutical codes of study drugs

Click here to view.(65K, pdf)

Supplement Table 3

Overall all cause of secondary cancer incidence and individual overall cancer-specific incidence before propensity score matching

Click here to view.(67K, pdf)

Supplement Table 4

Individual cancer-specific incidence according to duration of androgen deprivation exposure before propensity score matching

Click here to view.(69K, pdf)

Notes

Conflict of Interest:The authors have nothing to disclose.

Funding:This study was supported by Soonchunhyang University Research Fund, National Research Foundation of Korea grant funded by the Korea government (NRF-2021R1A5A2030333), and grants from the Korea University College of Medicine. The sponsor of the study was not involved in the study design, data analysis, data interpretation, writing of the report, or the decision to submit the study results for publication.

Author Contribution:

  • Conceptualization: JHK, JJ.

  • Data preparation and analysis: GHB, JJ.

  • Writing – original draft: JHK, TIN.

  • All authors have contributed in study design and revising the draft.

Acknowledgements

None.

Data Sharing Statement

The data that support the findings of this study are available from National Health Insurance System (NHIS) upon reasonable request. Although there are no patient privacy or safety concern, the data belongs to NHIS and could be allowed for the data accessing after IRB approval and NHIS permission.

References

    1. Heemers HV, Tindall DJ. Androgen receptor (AR) coregulators: a diversity of functions converging on and regulating the AR transcriptional complex. Endocr Rev 2007;28:778–808.
    1. Grossmann M, Ng Tang Fui M, Cheung AS. Late-onset hypogonadism: metabolic impact. Andrology 2020;8:1519–1529.
    1. Antonarakis ES. AR signaling in human malignancies: prostate cancer and beyond. Cancers (Basel) 2018;10:22
    1. Schweizer MT, Yu EY. AR-signaling in human malignancies: prostate cancer and beyond. Cancers (Basel) 2017;9:7
    1. Fabris C, Toniutto P, Falleti E, Fontanini E, Cussigh A, Bitetto D, et al. MTHFR C677T polymorphism and risk of HCC in patients with liver cirrhosis: role of male gender and alcohol consumption. Alcohol Clin Exp Res 2009;33:102–107.
    1. Kalra M, Mayes J, Assefa S, Kaul AK, Kaul R. Role of sex steroid receptors in pathobiology of hepatocellular carcinoma. World J Gastroenterol 2008;14:5945–5961.
    1. Drake JM, Graham NA, Lee JK, Stoyanova T, Faltermeier CM, Sud S, et al. Metastatic castration-resistant prostate cancer reveals intrapatient similarity and interpatient heterogeneity of therapeutic kinase targets. Proc Natl Acad Sci U S A 2013;110:E4762–E4769.
    1. Hsieh AC, Liu Y, Edlind MP, Ingolia NT, Janes MR, Sher A, et al. The translational landscape of mTOR signalling steers cancer initiation and metastasis. Nature 2012;485:55–61.
    1. Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, Mosquera JM, et al. Integrative clinical genomics of advanced prostate cancer. Cell 2015;161:1215–1228.
      Erratum in: Cell 2015;162:454.
    1. Yeh S, Lin HK, Kang HY, Thin TH, Lin MF, Chang C. From HER2/Neu signal cascade to androgen receptor and its coactivators: a novel pathway by induction of androgen target genes through MAP kinase in prostate cancer cells. Proc Natl Acad Sci U S A 1999;96:5458–5463.
    1. Aurilio G, Cimadamore A, Mazzucchelli R, Lopez-Beltran A, Verri E, Scarpelli M, et al. Androgen receptor signaling pathway in prostate cancer: from genetics to clinical applications. Cells 2020;9:2653
    1. Hu C, Fang D, Xu H, Wang Q, Xia H. The androgen receptor expression and association with patient's survival in different cancers. Genomics 2020;112:1926–1940.
    1. Peters AA, Buchanan G, Ricciardelli C, Bianco-Miotto T, Centenera MM, Harris JM, et al. Androgen receptor inhibits estrogen receptor-alpha activity and is prognostic in breast cancer. Cancer Res 2009;69:6131–6140.
    1. Robinson JL, Macarthur S, Ross-Innes CS, Tilley WD, Neal DE, Mills IG, et al. Androgen receptor driven transcription in molecular apocrine breast cancer is mediated by FoxA1. EMBO J 2011;30:3019–3027. [doi: 10.1038/emboj.2011.216]
    1. Fioretti FM, Sita-Lumsden A, Bevan CL, Brooke GN. Revising the role of the androgen receptor in breast cancer. J Mol Endocrinol 2014;52:R257–R265.
    1. Levine GN, D'Amico AV, Berger P, Clark PE, Eckel RH, Keating NL, et al. 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. Teoh JY, Tian XY, Wong CY, Lau CW, Cheng CK, Tang VW, et al. Endothelial dysfunction after androgen deprivation therapy and the possible underlying mechanisms. Prostate 2022;82:13–25.
    1. Karanika S, Karantanos T, Li L, Corn PG, Thompson TC. DNA damage response and prostate cancer: defects, regulation and therapeutic implications. Oncogene 2015;34:2815–2822.
    1. Gu S, Papadopoulou N, Gehring EM, Nasir O, Dimas K, Bhavsar SK, et al. Functional membrane androgen receptors in colon tumors trigger pro-apoptotic responses in vitro and reduce drastically tumor incidence in vivo. Mol Cancer 2009;8:114
    1. Vizoso FJ, Rodriguez M, Altadill A, González-Diéguez ML, Linares A, González LO, et al. Liver expression of steroid hormones and apolipoprotein D receptors in hepatocellular carcinoma. World J Gastroenterol 2007;13:3221–3227.
    1. Ma WL, Hsu CL, Yeh CC, Wu MH, Huang CK, Jeng LB, et al. Hepatic androgen receptor suppresses hepatocellular carcinoma metastasis through modulation of cell migration and anoikis. Hepatology 2012;56:176–185.
    1. Ma WL, Lai HC, Yeh S, Cai X, Chang C. Androgen receptor roles in hepatocellular carcinoma, fatty liver, cirrhosis and hepatitis. Endocr Relat Cancer 2014;21:R165–R182.
    1. White DL, Tavakoli-Tabasi S, Kuzniarek J, Pascua R, Ramsey DJ, El-Serag HB. Higher serum testosterone is associated with increased risk of advanced hepatitis C-related liver disease in males. Hepatology 2012;55:759–768.
    1. Beattie CW, Hansen NW, Thomas PA. Steroid receptors in human lung cancer. Cancer Res 1985;45:4206–4214.
    1. Kaiser U, Hofmann J, Schilli M, Wegmann B, Klotz U, Wedel S, et al. Steroid-hormone receptors in cell lines and tumor biopsies of human lung cancer. Int J Cancer 1996;67:357–364.
    1. Maasberg M, Rotsch M, Jaques G, Enderle-Schmidt U, Weehle R, Havemann K. Androgen receptors, androgen-dependent proliferation, and 5 alpha-reductase activity of small-cell lung cancer cell lines. Int J Cancer 1989;43:685–691.
    1. Grant L, Banerji S, Murphy L, Dawe DE, Harlos C, Myal Y, et al. Androgen receptor and Ki67 expression and survival outcomes in non-small cell lung cancer. Horm Cancer 2018;9:288–294.
    1. Nishio M, Ohyanagi F, Horiike A, Ishikawa Y, Satoh Y, Okumura S, et al. Gefitinib treatment affects androgen levels in non-small-cell lung cancer patients. Br J Cancer 2005;92:1877–1880.
    1. Rosner W, Auchus RJ, Azziz R, Sluss PM, Raff H. Position statement: utility, limitations, and pitfalls in measuring testosterone: an Endocrine Society position statement. J Clin Endocrinol Metab 2007;92:405–413.
    1. Chang Y, Kim JH, Noh JW, Cho YS, Park HJ, Joo KJ, et al. Prostate-specific antigen within the reference range, subclinical coronary atherosclerosis, and cardiovascular mortality. Circ Res 2019;124:1492–1504.
    1. Kim DK, Noh JW, Chang Y, Lee HY, Park JJ, Ryu S, et al. Association between prostate-specific antigen and serum testosterone: a systematic review and meta-analysis. Andrology 2020;8:1194–1213.

Metrics
Share
Figures

1 / 3

Tables

1 / 3

PERMALINK