Next Article in Journal
Comparison of Proclarix, PSA Density and MRI-ERSPC Risk Calculator to Select Patients for Prostate Biopsy after mpMRI
Previous Article in Journal
Use of Glycoproteins—Prostate-Specific Membrane Antigen and Galectin-3 as Primary Tumor Markers and Therapeutic Targets in the Management of Metastatic Prostate Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Insignificant Correlation between Androgen Deprivation Therapy and Incidence of Dementia Using an Extension Survival Cox Hazard Model and Propensity-Score Matching Analysis in a Retrospective, Population-Based Prostate Cancer Registry

1
National Cancer Control Institute, National Cancer Center, Goyang 10408, Korea
2
Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang 10408, Korea
3
Department of Urology, Urological Cancer Center, Research Institute and Hospital of National Cancer Center, Goyang 10408, Korea
4
Health Insurance Policy Research Institute, National Health Insurance Service, Wonju 26464, Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(11), 2705; https://doi.org/10.3390/cancers14112705
Submission received: 26 April 2022 / Revised: 25 May 2022 / Accepted: 27 May 2022 / Published: 30 May 2022
(This article belongs to the Topic Application of Big Medical Data in Precision Medicine)

Abstract

:

Simple Summary

This study shows the insignificant effect of the duration of androgen-deprivation therapy on the incidence of dementia in patients with prostate cancer from population-based data. We found that, despite an overall lower incidence of dementia in the androgen-deprivation-therapy group compared to the non-therapy group, there were no significant correlations between androgen-deprivation therapy and the prevalence of individual dementia subtypes in patients with prostate cancer. This demonstrates that patients with old age, obesity, regional SEER stage, a history of cerebrovascular disease, and a high Charlson Comorbidity Index were at increased risk for dementia.

Abstract

This study aims to evaluate the effect of androgen-deprivation therapy (ADT) on the incidence of dementia, after considering the time-dependent survival in patients with prostate cancer (PC) using a Korean population-based cancer registry database. After excluding patients with cerebrovascular disease and dementia before or within the 3-month-ADT and those with surgical castration, 9880 (19.3%) patients were matched into ADT and non-ADT groups using propensity-score matching (PSM) among 51,206 patients registered between 2006 and 2013. To define the significant relationship between ADT duration and the incidence of dementia, the extension Cox proportional hazard model was used with p-values < 0.05 regarded as statistically significant. The mean age and survival time were 67.3 years and 4.33 (standard deviation [SD] 2.16) years, respectively. A total of 2945 (9.3%) patients developed dementia during the study period, including Parkinson’s (11.0%), Alzheimer’s (42.6%), vascular (18.2%), and other types of dementia (28.2%). Despite PSM, the PC-treatment subtypes, survival rate, and incidence of dementia significantly differed between the ADT and non-ADT groups (p < 0.05), whereas the rate of each dementia subtype did not significantly differ (p = 0.069). A multivariate analysis for dementia incidence showed no significance of ADT type or use duration among patients with PC (p > 0.05), whereas old age, obesity, regional SEER stage, a history of cerebrovascular disease, and a high Charlson Comorbidity Index were significant factors for dementia (p < 0.05). Insignificant correlation was observed between ADT and the incidence of dementia based on the extension survival model with PSM among patients with PC.

1. Introduction

Worldwide, newly diagnosed patients with prostate cancer (PC) starting androgen-deprivation therapy (ADT) were estimated to be approximately 50% of all PC patients (25,000 patients annually) [1,2]. The incidence and types of ADT-related complications have also been found to increase proportionally to the exposure time to ADT, since ADT use has become the standard treatment for both androgen-dependent and androgen-independent PC, and its duration has increased from 12–24 months in the past to 3–5 years currently with other new agents added [2,3,4,5].
A recently emerging and highly debated issue on ADT-related adverse events is the possibility of impaired cognitive dysfunction, such as dementia [6], due to the irreversible, multifactorial, detrimental, exposure time-dependent, and general performance status-dependent impacts of ADT on the central nervous system, shown in both preclinical ex vivo and in vivo studies [7,8,9] and clinical non-randomized studies, especially for the Alzheimer’s dementia [6,10]. Without large-scale, long-term follow-up randomized controlled studies, the association between ADT and dementia incidence cannot be determined, although some multicentric/population-based studies or baseline controlled studies with different groups using matching methodology could be utilized to draw a conclusion [8,10,11].
As the prognostic outcome of PC and the incidence of dementia associated with ADT exposure are time-dependent factors [12], it is important to consider time-dependent survival variables in a multivariate analysis when investigating ADT as a risk factor for survival and dementia prevalence. Indeed, these survival variables might have had an influence on the contradictory inferences obtained by previous studies [8,10,11,12,13]. Therefore, using data from a Korean population-based cancer registry, this study aimed to evaluate the effect of ADT on the incidence of dementia in patients with PC, after considering time-dependent extension survival methodology and after eliminating the influences of inherent selection biases, by applying an extension survival model and propensity-score matching (PSM) on PC cohorts with and without ADT use.

2. Materials and Methods

2.1. Population-Based Cancer Registry Database

The analytic methodology for the anonymized the Korean National Health Insurance System of Statistics (NHIS) database has been described in detail in our previous epidemiological articles [4,13]. This study used cohorts with cancer incidence and mortality data from 2006 to 2013 in the NHIS combined Korean Central Cancer Registry, in which accurate recording of cancer registration data followed a standardized cancer diagnosis dating system. Hence, the possibility of multiple diagnosis dates in previously diagnosed patients was excluded, and only accurately classified existing and new patients were included.

2.2. Study Sample

A total of 51,206 patients with PC between 2006 and 2013 were included based on the International Classification of Diseases, Tenth Edition (ICD-10) diagnosis code C61, and followed-up until the end of 2014. Patients with a history of dementia or cerebrovascular disease prior to PC diagnosis (N = 15720, 30.7%) were excluded. Patients within 3-months of PC diagnosis (N = 1039, 2.0%); within 3-months of initiating ADT (N = 372, 0.7%); with less than 3-months of ADT history (N = 2179, 4.3%); and a history of surgical castration (N = 325, 0.6%) for a total of 34,075 patients with complete medical records, including 16,827 (49.4%) patients treated with ADT, were included in the study.
During the study period, a total of 2945 (8.60%) patients with PC with a newly developed diagnosis of dementia were enrolled, based on the ICD-9 and ICD-10 codes for dementia and cognitive dysfunction (F00~03 and G309). The dementia subtypes were classified into Alzheimer’s dementia (F00 or G309), vascular dementia (F01), Parkinson’s dementia (F02), other non-Parkinson’s dementia (F02), and unspecified dementia (F03). Furthermore, patients were enrolled only if they had a longer than 3-month history of dementia medication use and had been diagnosed more than twice within 6 months following the initial diagnosis by a neurologist, to exclude elderly patients taking prophylactic dementia medications without a diagnosis of dementia.
Other clinico-pathological parameters including underlying disease, body mass index (BMI), Charlson Comorbidity Index (CCI), and medication use were selected according to the methods used in our previous epidemiological studies [4,14]. Only patients using statin and aspirin as preventive medical therapy for cardiovascular disease were considered. SEER (Surveillance, Epidemiology, and End Results) stage classification was used to determine the stage of PC (local, regional, distant, and unknown). The primary therapeutic strategies for PC treatment included surgery, radiation, and chemotherapy, which were coded under EDI-CD.
During the study period, a total of 84 hormonal agents approved by the Korean FDA, including gonadotrophin-releasing hormone (GnRH) agents (goserelin, leuprorelin, and triptorelin acetate) and anti-androgen agents (flutamide and bicalutamide), were used for ADT. The finally enrolled ADT group comprised patients treated with ADT for at least 3 months.

2.3. Propensity-Score Matching Analysis

PSM of 1:1 was performed between the ADT and non-ADT groups to prevent significant baseline differences and resulted in a final enrollment of 9880 patients. To characterize the insignificant differences between ADT and non-ADT groups in PC, a 1249-paired PSM 1:1 was performed. The matched variables, which were significantly different between ADT and non-ADT groups, were age at cancer diagnosis, BMI, SEER stage, a history of smoking, underlying disease (hypertension, diabetes mellitus, and cardiovascular disease), CCI, and the medication (aspirin and statin) used at the time of the study period.
After matching, a test for homogeneity between the groups was performed using a chi-squared or Mc Nemar’s test for categorical data, a paired t-test for continuous data, and analysis of variance (ANOVA) to explain the effect of each confounding variable on the incidence of dementia. Lastly, a PSM of 1:1 was performed according to the disease states to analyze the relationship between the use of ADT and the incidence of dementia. To adjust for confounding variables, we performed regression analysis to test the difference in the resulting variables.

2.4. Extended Model Analysis

An extended survival model [9] was developed for the duration and type of the ADT variables to control for treatment duration and type that affects time-dependent outcome variable. The survival intervals were divided into 30 days and included in the survival period of each group according to the duration and type of the ADT at each survival interval.

2.5. Multivariate Cox Analysis

The paired t-test and Chi-square or Mc Nemar’s test were used to determine the differences between groups. A multivariate Cox analysis was used to define the significant effect of ADT and its duration on dementia incidence during the study period. From the starting date of ADT, hazard ratios (HRs) and 95% confidence interval (CI) values for the occurrence of dementia and death events among the ADT groups were analyzed using Cox proportional hazard models.
To adjust the Cox proportional models, age, BMI, a history of smoking, SEER stage, medication (aspirin and statin), underlying diseases (diabetes, hypertension, and other cardiovascular diseases), and the CCI were used. Two-sided p-values < 0.05 were considered statistically significant. All statistical analyses were performed using SAS (release 9.4, SAS Institute Inc., Cary, NC, USA).

3. Results

3.1. Overall Patients’ Baseline Characteristics

The mean age, ADT duration, and survival time of the 9880 included patients were 67.3 (SD 6.3) years-old, 960.8 (range, 736.1–741.0) days, and 4.33 (SD 2.16) years, respectively, with a 3.53% of overall death rate, including 2.0% of PC-specific deaths during a mean follow-up time of 4.3 (SD 2.1) years (Table 1). Among 592 (6.0%) patients diagnosed with dementia during the study period, Alzheimer’s dementia (N = 252, 42.6%) was the most prevalent subtype, followed by vascular dementia (N = 108, 18.2%), Parkinson’s disease (N = 65, 11.0%), and other non-Parkinson’s dementia (N = 167, 28.2%). Other clinic-pathological information, including SEER stage and therapeutic modalities, are described in Table 1.

3.2. Comparative Results between Matched Cohort of ADT and Non-ADT Groups

The 1:1 matched comparative results between ADT and non-ADT groups demonstrated that the overall dementia incidence was significantly different (ADT, N = 270 [45.6%] vs. non-ADT, N = 322 [54.4%], p = 0.025), whereas the incidences of each individual dementia subtype were not different between groups (p = 0.069), in which the vascular dementia rate was higher in the non-ADT group (21.74%) than the ADT group (14.07%) (Table 1).
Although there were no baseline differences in SEER stage, BMI, diagnosed year of cancer, year of dementia diagnosis, underlying diseases, medication, and smoking history between the ADT and non-ADT groups after PSM, there were still significant baseline differences, with significantly higher rates in the ADT group of cancer treatment within 6 months from cancer diagnosis (83.9% vs. 49.6% in non-ADT), chemotherapy (42.0% vs. 10.6%), radiation therapy (16.5% vs. 5.0%), and death (5.4% vs. 1.7%), including PC-specific death (65.7% vs. 15.9%) than in the non-ADT group, which had a significantly higher rate of surgical treatment (46.8%) than the ADT group (28.9%) (p < 0.05, Table 1).

3.3. Multivariate Results of Dementia Incidence

In the multivariate analysis among the 1:1 PS-matched patients (total 9880) with PC, ADT duration and ADT therapeutic types were not significant risk factors for dementia incidence (p > 0.05), whereas old age (HR > 1.0), obese BMI (25~30, HR 0.676, CI 0.545–0.838), regional SEER stage (HR 0.720, CI 0.578–0.897), presence of cardiovascular disease (HR 3.083, CI 2.543–3.737), and high CCI (HR 1.479 for CCI 1 and HR 1.961 for CCI2+) were significant risk factors for dementia (p < 0.05, Table 2).

3.4. Comparative Results between Dementia and Non-Dementia Groups

In the comparison between 1:1 PS-matched groups (2498 patients) with dementia and without dementia by SEER stage, BMI, diagnosed year of cancer, year of dementia diagnosis, underlying diseases, medication, and smoking history (Table 3), the dementia group had a significantly longer interval between cancer diagnosis and treatment initiation within 6 months (59.8% vs. 38.35%), and lower rates of primary treatment, such as surgery/chemotherapy/radiation (1.0/26.2/2.7% vs. 26.2/27.0/3.7%), higher rate of death (12.4% vs. 7.5%), shorter duration of ADT use (843.9 vs. 1243.0 days) and shorter time interval from cancer diagnosis to ADT initiation (89.4 vs. 143.2 days) (p < 0.05).

3.5. Multivariate Results of Dementia Incidence According to SEER Tumor Staging

In the stratified comparison according to SEER staging, Table 4 and Table 5 show the multivariate analysis of risk factors for dementia among local/regional and distant/unknown SEER stage patients after 1:1 PSM. The results showed that old age (HR 1.965–2.860), obesity BMI (HR0.711, CI 0.564–0.896), high CCI (HR 1.1468–1.716), and ADT duration between 3–24 months (HR 0.224-0.410) were significant risk factors for dementia among local/regional SEER staged patients (p < 0.05, Table 4); age > 75 year-old (HR 7.995, CI 2.159–29.604), underweight BMI (HR 3.425, CI 1.421–8.258), and high CCI (HR 1.919 for CCI 1 and HR 2.201 for CC2+) were significant risk factors among distant/unknown SEER stage patients (p < 0.05, Table 5).

4. Discussion

ADT is a standardized therapy for PC, and it blocks the androgen effects resulting in a rapid drop of testosterone concentration, leading to andropause to prevent from PC progression. However, ADT had a detrimental exposure-dependent effect on general performance status, including impaired cognitive dysfunction, such as the development of dementia [15]. Some contradictory results denied the insignificantly direct correlation of ADT exposure time to dementia incidence in PC [16,17,18].
This study used an extended survival methodology to evaluate the time-dependent influences of dementia by the use of ADT and also concluded that a significantly direct relationship between ADT and its duration and dementia incidence have not been shown, especially for Alzheimer’s dementia in a Korean population cohort. This study considered the time-dependent effect of extended exposure of ADT on dementia and disease burden states by the categorized SEER staging.
We applied strict diagnostic indications of dementia, and eliminated the effect of the inherent bias of baseline differences between ADT and non-ADT cohorts by PSM, even after considering all the adjusted major known etiologies of cognitive dysfunction and all the disease burden states according to SEER staging, especially in an Asian population-based cancer registry [7,16,19,20,21,22,23]. In the absence of randomized clinical trials, our analytic method is the best approach to demonstrate the insignificant relationship between ADT exposure and dementia in real-world clinical settings.
Contrary to previous studies showing a significant direct relationship between ADT and dementia incidence, the insignificant relationship between ADT exposure and the incidence of dementia we observed in this study might be explained by multiple aspects, such as the different characteristics of the cohorts, geographical prevalence of dementia in different ethnical backgrounds, and their enrollment criteria. This study applied different selection criteria of dementia compared to other similar population-based studies [15,16,17]. We enrolled patients with a more than 3-month prescription history of dementia medication and who were diagnosed more than twice within at least 6 months from the first diagnosis. Those who were taking prophylactic dementia medications were excluded from the analysis.
Other reasons for the insignificant results could be the different ethnicity and geographical locations of the cohorts [7,15,21]. Hence, the study cohorts likely differed in terms of immunological profiles [8], social background and lifestyles [18,24], the regional prevalence rate of dementia [25,26], and different ADT-dosing policies based on ethnicity-related anthropometric characteristics and androgen-physiologic profiles [27]. Previous studies using Asian cohorts showed a similarly insignificant relationship between ADT and dementia prevalence, especially with ADT-related Alzheimer’s disease. Asians reportedly have a lower rate of Alzheimer’s dementia and higher rate of non-Alzheimer’s disease, such as vascular dementia, compared with those seen in Caucasians and African Americans [24,28,29] due to different familial and social lifestyles, dietary habits, lifespans, and genetic/immunological backgrounds [25,26,27].
Koreans are part of the East Asian population, which includes the populations of Japan, China, and Taiwan. These countries culturally possess strong family bonds based on the Han-ethnic and Confucianism culture, a highly hierarchic male-dominant social lifestyle with higher socioeconomic burden in males, geographically overcrowded urban lifestyle [25], and a nationally obligatory Bacille Calmette–Guerin vaccination program [9], which could account for the reduced risks of Alzheimer’s dementia but increased risks of non-Alzheimer’s dementia, such as vascular dementia [24,29]. This study also showed an insignificant difference in the rate of Alzheimer’s dementia between the ADT (42.96%) and non-ADT (42.24%) groups, whereas a higher rate of vascular dementia was found in the non-ADT group (14.07% ADT vs. 21.74% non-ADT, Table 1) [16].
This study compared the dementia and non-dementia groups to identify any differential characteristics among the patients (Table 3), revealing the significant importance of tumor characteristics and their relationship to the patients’ general performance status in PC cohorts, which affects the dementia incidence. Since the patients’ general condition significantly affected the incidence of dementia, the tumor burden and extent were important factors for not only general performance and nutritional state but also mental health, including cognitive function [7,19,20,30].
The dementia group had a significantly poorer general condition (underweight BMI, higher CCI, and higher rates of hypertension/diabetes mellitus/aspirin use), higher tumor extent (higher rate of distant staging and lower rate of regional staging, and higher rate of early initiation of cancer treatment), and higher rates of no ADT treatment and death. They were also older compared to the non-dementia group. Therefore, a thorough dementia screening program in collaboration with other specialists, including neurologists, nutritionists, psychiatrists, and rehabilitation medicine doctors, should be considered for high-risk PC patients based on these factors.
Other reasons for the contradictory results compared to previous studies could be the ADT duration and study period [9,16,20]. The study period was between 2006 and 2013, during which time most of the patients had ADT histories of less than 3 years, since ADT had been approved by the national insurance system as a first-line treatment only for advanced and metastatic PC cases until biochemical recurrence. After that point, second-line systemic therapies were started without ADT due to the transformation of hormone sensitive PC to hormone-resistant, hormone refractory PC.
Currently, ADT has become the mainstay treatment and is continuously used in patients with PC until either death or remission, with the newly introduced secondary and chemotherapeutic agents offering prolonged overall survival [12,15]. Thus the number of patients using ADT > 3 years has increased, suggesting there might be a significant relationship between ADT and dementia incidence in this group. Comparable results have been shown by previous studies comparing the effects of similar ethnicities [22,27], dietary lifestyle [31], and psycho-social environments [3] on the incidence of dementia.
The multivariate analyses in Table 4 and Table 5 showed that ADT was a significant risk factor of dementia only for the localized/regional SEER staging groups, whereas it was not for the distant/unknown SEER staging cohorts. Further enrollment of localized/regional SEER staged patients with PC might reveal a significant relationship between ADT and dementia incidence, because the survival time of this cohort have been prolonged more recently with new therapeutic modalities.
Despite the fact that PSM methodology and extended survival analysis were used to adjust for inherent influencing parameters on dementia incidence, this study had certain limitations: it was a retrospective study that utilized health-claims data, and not all variables influencing dementia, such as exercise, severity and extent of cardiovascular disease, bisphosphonate, follow-up duration, and systemic therapies, such as chemotherapy and anticoagulant agents (cilostazol or clopidogrel), were included in the multivariate analysis. In particular, the significant cardiovascular disease for risk factors of the dementia in this study has also been a well-known factor for both ADT and dementia [19,20].
Further concrete evaluation of the severity and extent of cardiovascular disease might help to eliminate the confounding effect of the cardiovascular disease on the cognitive dysfunction. In addition, a strict definition of dementia used in this study and shorter follow-up duration compared with the survival time for PC might eliminate all the possible minor dementia in the analysis as well as elimination of follow-up duration, radiation, and chemotherapy as matching variables. Further study with more concrete adjusting the extent of cardiovascular disease should be considered. However, this was the first population-based retrospective study to use two statistical modalities to control the inherent cohort bias and time-dependent bias to demonstrate the relationship between ADT duration and type and dementia incidence in a PC cohort. Further, current second generation of hormonal agents with ADT should also be considered in future studies.

5. Conclusions

This study showed an insignificant correlation between ADT and the incidence of dementia using an extension survival Cox hazard model and PSM analyses. Further prospective trials with large-sized cohorts should be performed to evaluate the true effect of ADT on dementia incidence in PC during the current new therapeutic era of PC.

Author Contributions

Conceptualization, S.-H.K., S.H.K. and Y.A.K., Data acquisition, Study Performance, S.H.K. and J.Y.J.; Interpretation of data, Writing—Reviewing and Editing, and Supervision S.-H.K., S.H.K. and Y.A.K.; and Statistical analysis, Y.A.K., M.S.Y. and J.H.B. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the National Cancer Center, Grant (No. NCC2111080-2 and NCC2211880-1).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board (IRB) of the National Cancer Center and Cancer Research Institute in Korea (IRB no. NCC2015-0217).

Informed Consent Statement

The requirement for informed consent was waived because of the anonymous nature of the cancer registry public data from the Korean National Health Insurance System (NHIS) and the Korea National Cancer Incidence Database of the Korean Central Cancer Registry database. All methods were performed in accordance with the relevant guidelines and regulations from the NHIS.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hassanipour-Azgomi, S.; Mohammadian-Hafshejani, A.; Ghoncheh, M.; Towhidi, F.; Jamehshorani, S.; Salehiniya, H. Incidence and mortality of prostate cancer and their relationship with the Human Development Index worldwide. Prostate Int. 2016, 4, 118–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Shahinian, V.B.; Kuo, Y.-F.; Freeman, J.L.; Orihuela, E.; Goodwin, J.S. Increasing use of gonadotropin-releasing hormone agonists for the treatment of localized prostate carcinoma. Cancer 2005, 103, 1615–1624. [Google Scholar] [CrossRef] [PubMed]
  3. Ritch, C.A.-O.; Cookson, M. Recent trends in the management of advanced prostate cancer. F1000Research 2018, 7, F1000 Faculty Rev-1513. [Google Scholar] [CrossRef] [Green Version]
  4. Shin, H.R.; Won, J.Y.; Jung, W.K.; Kong, J.K.; Yim, H.S.; Lee, K.J.; Noh, I.H.; Lee, K.J.; Pisani, P.; Park, G.H. Nationwide cancer incidence in Korea, 1999~2001; first result using the national cancer incidence database. Cancer Res. Treat. 2005, 37, 325–331. [Google Scholar] [CrossRef] [Green Version]
  5. Maggi, M.; Salciccia, S.; Del Giudice, F.; Busetto, G.M.; Falagario, U.G.; Carrieri, G.; Ferro, M.; Porreca, A.; Di Pi-erro, G.B.; Fasulo, V.; et al. A systematic review and meta-analysis of randomized controlled trials with novel hormonal therapies for non-metastatic castration-resistant prostate cancer: An update from mature overall survival data. Front. Oncol. 2021, 11, 700258. [Google Scholar] [CrossRef]
  6. Maki, P.M.; Sundermann, E. Hormone therapy and cognitive function. Hum. Reprod. Update 2009, 15, 667–681. [Google Scholar] [CrossRef] [Green Version]
  7. Leranth, C.; Petnehazy, O.; MacLusky, N.J. Gonadal hormones affect spine synaptic density in the CA1 hippocampal subfield of male rats. J. Neurosci. 2003, 23, 1588–1592. [Google Scholar] [CrossRef] [Green Version]
  8. Nead, K.T.; Sinha, S.; Nguyen, P.L. Androgen deprivation therapy for prostate cancer and dementia risk: A systematic review and meta-analysis. Prostate Cancer Prostatic Dis. 2017, 20, 259–264. [Google Scholar] [CrossRef]
  9. Hong, J.-H.; Huang, C.-Y.; Chang, C.-H.; Muo, C.-H.; Jaw, F.-S.; Lu, Y.-C.; Chung, C.-J. Different androgen deprivation therapies might have a differential impact on cognition—An analysis from a population-based study using time-dependent exposure model. Cancer Epidemiol. 2020, 64, 101657. [Google Scholar] [CrossRef]
  10. Nead, K.T.; Gaskin, G.; Chester, C.; Swisher-McClure, S.; Leeper, N.J.; Shah, N.H. Association between androgen deprivation therapy and risk of dementia. JAMA Oncol. 2017, 3, 49–55. [Google Scholar] [CrossRef]
  11. Gofrit, O.N.; Bercovier, H.; Klein, B.Y.; Cohen, I.R.; Ben-Hur, T.; Greenblatt, C.L. Can immunization with Bacillus Calmette-Guerin (BCG) protect against Alzheimer’s disease? Med. Hypotheses 2019, 123, 95–97. [Google Scholar] [CrossRef] [PubMed]
  12. Mdzinarishvili, T.; Gleason, M.X.; Kinarsky, L.; Sherman, S. Extension of cox proportional hazard model for estimation of interrelated age-period-cohort effects on cancer survival. Cancer Inform. 2011, 10, 31–44. [Google Scholar] [CrossRef] [PubMed]
  13. Baik, S.H.; Kury, F.S.P.; McDonald, C.J. Risk of Alzheimer’s Disease Among Senior Medicare Beneficiaries Treated with Androgen Deprivation Therapy for Prostate Cancer. J. Clin. Oncol. 2017, 35, 3401–3409. [Google Scholar] [CrossRef] [PubMed]
  14. Jung, K.W.; Won, Y.J.; Kong, H.J.; Lee, E.S. Prediction of Cancer Incidence and Mortality in Korea, 2019. Cancer Res. Treat. 2019, 51, 431–437. [Google Scholar] [CrossRef]
  15. Martinsson, L.; Lundström, S.; Sundelöf, J. Quality of end-of-life care in patients with dementia compared to patients with cancer: A population-based register study. PLoS ONE 2018, 13, e0201051. [Google Scholar] [CrossRef] [Green Version]
  16. Chung, S.D.; Lin, H.C.; Tsai, M.C.; Kao, L.T.; Huang, C.Y.; Chen, K.C. Androgen deprivation therapy did not increase the risk of Alzheimer’s and Parkinson's disease in patients with prostate cancer. Andrology 2016, 4, 481–485. [Google Scholar] [CrossRef] [Green Version]
  17. Kao, L.T.; Lin, H.C.; Chung, S.D.; Huang, C.Y. No increased risk of dementia in patients receiving androgen deprivation therapy for prostate cancer: A 5-year follow-up study. Asian J. Androl. 2017, 19, 414–417. [Google Scholar] [CrossRef]
  18. Khosrow-Khavar, F.; Rej, S.; Yin, H.; Aprikian, A.; Azoulay, L. Androgen Deprivation Therapy and the Risk of Dementia in Patients with Prostate Cancer. J. Clin. Oncol. 2017, 35, 201–207. [Google Scholar] [CrossRef]
  19. Muniyan, S.; Xi, L.; Datta, K.; Das, A.; Teply, B.A.; Batra, S.K.; Kukreja, R.C. Cardiovascular risks and toxicity—The Achilles heel of androgen deprivation therapy in prostate cancer patients. Biochim. Biophys. Acta Rev. Cancer 2020, 1874, 188383. [Google Scholar] [CrossRef]
  20. Sun, L.; Parikh, R.B.; Hubbard, R.A.; Cashy, J.; Takvorian, S.U.; Vaughn, D.J.; Robinson, K.W.; Narayan, V.; Ky, B. Assessment and Management of Cardiovascular Risk Factors Among US Veterans with Prostate Cancer. JAMA Netw. Open 2021, 4, e210070. [Google Scholar] [CrossRef]
  21. Kim, J.H.; Lee, B.; Han, D.H.; Chung, K.J.; Jeong, I.G.; Chung, B.I. Discrepancies on the association between androgen deprivation therapy for prostate cancer and subsequent dementia: Meta-analysis and meta-regression. Oncotarget 2017, 8, 73087–73097. [Google Scholar] [CrossRef] [Green Version]
  22. Sun, M.; Cole, A.P.; Hanna, N.; Mucci, L.A.; Berry, D.L.; Basaria, S.; Ahern, D.K.; Kibel, A.S.; Choueiri, T.K.; Trinh, Q.D. Cognitive Impairment in Men with Prostate Cancer Treated with Androgen Deprivation Therapy: A Systematic Review and Meta-Analysis. J. Urol. 2018, 199, 1417–1425. [Google Scholar] [CrossRef] [PubMed]
  23. Tae, B.S.; Jeon, B.J.; Shin, S.H.; Choi, H.; Bae, J.H.; Park, J.Y. Correlation of Androgen Deprivation Therapy with Cognitive Dysfunction in Patients with Prostate Cancer: A Nationwide Population-Based Study Using the National Health Insurance Service Database. Cancer Res. Treat. 2019, 51, 593–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Venketasubramanian, N.; Sahadevan, S.; Kua, E.H.; Chen, C.P.; Ng, T.P. Interethnic differences in dementia epidemiology: Global and Asia-Pacific perspectives. Dement. Geriatr. Cogn. Disord. 2010, 30, 492–498. [Google Scholar] [CrossRef] [PubMed]
  25. Russ, T.C.; Batty, G.D.; Hearnshaw, G.F.; Fenton, C.; Starr, J.M. Geographical variation in dementia: Systematic review with meta-analysis. Int. J. Epidemiol. 2012, 41, 1012–1032. [Google Scholar] [CrossRef] [PubMed]
  26. Yuchi, W.; Sbihi, H.; Davies, H.; Tamburic, L.; Brauer, M. Road proximity, air pollution, noise, green space and neurologic disease incidence: A population-based cohort study. Environ. Health 2020, 19, 8. [Google Scholar] [CrossRef] [Green Version]
  27. Matthews, K.A.; Xu, W.; Gaglioti, A.H.; Holt, J.B.; Croft, J.B.; Mack, D.; McGuire, L.C. Racial and ethnic estimates of Alzheimer’s disease and related dementias in the United States (2015–2060) in adults aged ≥65 years. Alzheimers Dement. 2019, 15, 17–24. [Google Scholar] [CrossRef]
  28. Ellis, L.; Nyborg, H. Racial/ethnic variations in male testosterone levels: A probable contributor to group differences in health. Steroids 1992, 57, 72–75. [Google Scholar] [CrossRef]
  29. Gofrit, O.N.; Klein, B.Y.; Cohen, I.R.; Ben-Hur, T.; Greenblatt, C.L.; Bercovier, H. Bacillus Calmette-Guérin (BCG) therapy lowers the incidence of Alzheimer’s disease in bladder cancer patients. PLoS ONE 2019, 14, e0224433. [Google Scholar] [CrossRef] [Green Version]
  30. McHugh, D.J.; Root, J.C.; Nelson, C.J.; Morris, M.J. Androgen-deprivation therapy, dementia, and cognitive dysfunction in men with prostate cancer: How much smoke and how much fire. Cancer 2018, 124, 1326–1334. [Google Scholar] [CrossRef]
  31. Banda, Y.; Kvale, M.N.; Hoffmann, T.J.; Hesselson, S.E.; Ranatunga, D.; Tang, H.; Sabatti, C.; Croen, L.A.; Dispensa, B.P.; Henderson, M.; et al. Characterizing race/ethnicity and genetic ancestry for 100,000 subjects in the genetic epidemiology research on adult health and aging (GERA) cohort. Genetics 2015, 200, 1285–1295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Comparison of demographics between ADT and non-ADT groups among patients with prostate cancer after 1:1 propensity score matching adjusted variables for age, BMI, smoking, diagnosed year of cancer and dementia, SEER stage, underlying diseases, medications, and Charlson comorbidity index.
Table 1. Comparison of demographics between ADT and non-ADT groups among patients with prostate cancer after 1:1 propensity score matching adjusted variables for age, BMI, smoking, diagnosed year of cancer and dementia, SEER stage, underlying diseases, medications, and Charlson comorbidity index.
Total (N = 9880)ADT_Group
(N = 4940)
Non-ADT Group
(N = 4940)
p-Value
n%N%n%
Age group (year-old)9880 4940 4940
    50–64303773.73155431.46148368.540.1254
    65–7457700.22287558.20289541.80
    75-107326.0551110.3456289.66
BMI (missing = 4084) (kg/cm2)
    Underweight (<18.5)2212.241132.291082.190.9516
    Normal (18.5~22.9)309431.31154831.34154631.30
    Overweight (23~24.9)288629.21142728.89145929.53
    Obesity (25~29.9)344934.91173835.18171134.63
    Severe obesity. (30~)2302.331142.301162.35
Smoking (Missing = 11,099)
    Ex-smoker8788.894398.894398.891
    current smoker123412.4961712.4961712.49
    non smoker776878.62388478.62388478.62
Year of cancer diagnosis
    2006–2009376438.10188638.18187838.020.8684
    2010–2013611661.90305461.82306261.98
SEER
    Local598260.55299160.55299160.551
    Regional284228.77142128.77142128.77
    Distant440.44220.45220.44
    Unknown101210.2450610.2450610.24
Interval time between cancer diagnosis and treatment initiation within 6 months659666.76414483.89245249.64<0.0001
Underlying disease
    Hypertension490049.60245049.60245049.601
    Diabetes255025.81127525.81127525.811
    Cardiovascular disease9209.314609.314609.311
Medication
    Aspirin380238.48190138.48190138.481
    Statin238824.17119424.17119424.171
Treatment type
    Surgical Treatment373737.82142728.89231046.76<0.0001
    Chemotherapy265026.82210842.6754210.97<0.0001
    Radiation107510.8884517.112304.66<0.0001
    No treatment241824.4856011.33185837.61<0.0001
Pathology
    adenocarcinoma822083.20408682.71413483.680.1965
    no adeno166016.8085417.2980616.32
Charlson comorbidity index
    0434844.01217444.01217444.011
    1426643.18213343.18213343.18
    2+126612.8163312.8163312.81
Survival(death)3493.532655.36841.70<0.0001
Cancer death
    Prostate cancer18753.5817465.661315.48<0.0001
    other cancer3911.17176.422226.19
    Other12335.257427.924958.33
Survival time(year, mean [SD])4.33 (2.16)4.31 (2.15)4.34 (2.16)0.5926
Dementia59210027045.632254.40.025
    Parkinson6510.983011.113510.870.0691
    Alzheimer25242.5711642.9613642.24
    Vascular dementia10818.243814.087021.74
    Other dementia16728.218631.858125.15
ADT treatment NA
    only GnRH agonist3406.883446.88---
    only Antiandrogen70614.2976014.29---
    Both389478.83401378.83---
Duration of ADT (days, mean (SD)) NA
    Total960.81(736.11)960.81(736.11)
    only Antiandrogen711.39(617.76)711.39(617.76)--
    only GnRH agonist692.37(506.89)692.37(506.89)--
    Both1029.47(757.41)1029.47(757.41)--
ADT Duration (days, mean (SD)) -
    No ADT----
    3 ≤ ADT < 6130.45(28.90)130.45(28.90)--
    6 ≤ ADT < 12268.82(53.84)268.82(53.84)--
    12 ≤ ADT < 24528.75(104.34)528.75(104.34)--
    24 ≤ ADT1486.90(626.09)1486.90(626.09)--
Interval time from cancer diagnosis to ADT start(days, mean(SD), median) NA
    Total186.45(370.44)186.45(370.44)
    only Antiandrogen319.47(496.64)319.47(496.64)--
    only GnRH agonist148.66(321.02)148.66(321.02)--
    Both343.14 (474.36)343.14 (474.36)--
NA, not applicable; ADT, androgen-deprivation therapy; SEER, Surveillance, Epidemiology, and End Results; SD, standard deviation; GnRH, Gonadotrophin-releasing hormone; and BMI, body mass index.
Table 2. Multivariate cox regression of risk factors for dementia among overall patients after propensity-score matching and extended survival analysis.
Table 2. Multivariate cox regression of risk factors for dementia among overall patients after propensity-score matching and extended survival analysis.
Univariate Multivariate
CharacteristicsHR95% CIp-ValueaHR95% CIp-Value
Age group (year-old)
   50 ≤ Age < 651 1
   65 ≤ Age ≤ 752.1031.6862.622<0.00011.7971.4262.265<0.0001
   75 < Age3.1442.3894.137<0.00012.3921.7813.213<0.0001
BMI (kg/cm2)
   Normal (18.5~22.9)1 1
   Underweight (<18.5)1.4530.9462.2320.08771.2350.7861.9390.3598
   Overweight (23~24.9)0.8220.6751.0020.05220.8860.7221.0860.2431
   Obesity (25~)0.6170.5040.756<0.00010.6760.5450.8380.0004
   Severe obesity. (30~)0.5850.3111.1030.09750.6550.3461.2410.1943
Smoking
   non smoker1
   current smoker0.9280.7231.190.5548
   ex smoker0.730.5291.0080.0562
SEER
   Local1 1
   Regional0.6350.5170.780<0.00010.7200.5780.8970.0033
   Distant1.8220.7544.4010.18241.9380.7994.7040.1434
   Unknown1.1900.9331.5170.16131.1450.8891.4750.2939
Medication
   Aspirin (Ref. = no)1.2241.0411.4410.01471.0100.8241.2380.9255
   Statin (Ref. = no)0.9790.8101.1830.6018
Underlying disease
   CVD3.4232.8434.122<0.00013.0832.5433.737<0.0001
   Hypertension (Ref. = no)1.2361.0511.4520.01041.0580.8611.2990.5937
   Diabetes mellitus (Ref. = no)1.2301.03114670.02140.9020.7311.1140.3387
Charlson comorbidity index
   01 1
   11.5501.2901.862<0.00011.4791.2171.798<0.0001
   2+2.1801.7322.745<0.00011.9611.5102.547<0.0001
ADT type
   No ADT1 1
   only Antiandrogen0.7380.5171.0520.09310.6200.2911.3190.2144
   only GnRH agonist1.0750.7031.6430.73850.8930.3932.0280.7865
   Both1.0580.8851.2640.53610.8120.3961.6660.5701
ADT Duration(month)
   No ADT1 1
   3 ≤ ADT < 60.6490.4191.0080.05410.7660.3341.7590.5302
   6 ≤ ADT < 120.9570.71.3080.78071.1930.5512.5830.6539
   12 ≤ ADT < 240.7610.5751.0060.05480.9220.4321.9690.8345
   24 ≤ ADT1.5951.2741.997<0.00011.8470.8813.8730.1045
NA, not applicable; ADT, androgen-deprivation therapy; SEER, Surveillance, Epidemiology, and End Results; SD, standard deviation; GnRH, Gonadotrophin-releasing hormone; and BMI, body mass index.
Table 3. Comparison of the demographic characteristics between dementia and non-dementia groups among patients with prostate cancer after 1:1 propensity score matching adjusted variables for age, SEER stage, diagnosed year, aspirin use, statin use, BMI, smoking, hypertension, diabetes mellitus, and cardiovascular disease.
Table 3. Comparison of the demographic characteristics between dementia and non-dementia groups among patients with prostate cancer after 1:1 propensity score matching adjusted variables for age, SEER stage, diagnosed year, aspirin use, statin use, BMI, smoking, hypertension, diabetes mellitus, and cardiovascular disease.
Total (N = 2498)Dementia Group
(N = 1249)
Non-Dementia Group
(N = 1249)
p-Value
n%n%n%
Age group (year-old)
   50–6437515.0119015.2118514.810.5583
   65–74141256.5369365.4471957.57
   75-71128.4636689.7134527.62
BMI (missing = 4084) (kg/cm2)
   Underweight (<18.5)833.32423.36413.280.9968
   Normal (18.5~22.9)93137.2746837.4746337.07
   Overweight (23~24.9)72128.8636228.9835928.74
   Obesity (25~)72228.9035728.5836529.22
   Severe obesity (30~)411.64201.60211.68
Year of cancer diagnosis
   2006–2009135954.4069955.9666052.840.1172
   2010–2013113945.6055044.0458947.16
Smoking (Missing = 11,341)
   Ex-smoker1967.85987.85987.851
   current smoker35014.0117514.0117514.01
   non smoker195278.1497678.1497678.14
SEER
   Local146869.5773458.7773458.771
   Regional50423.8925220.1825220.18
   Distant1386.54695.52695.52
   Unknown38818.3919415.5319415.53
Interval time between cancer diagnosis and treatment initiation within 6 months122649.0874759.8147938.35<0.0001
Underlying disease
   Hypertension138062.6769062.6769062.671
   Diabetes82237.3341137.3341137.331
Medication
   Aspirin111444.6055744.6055744.601
   no Aspirin138455.4069255.4069255.40
   Statin63625.4631825.4631825.461
   no Statin186274.5493174.5493174.54
Treatment type
   Surgical Treatment43716.9690.9542826.23<0.0001
   Chemotherapy68826.7124726.1744127.02<0.0001
   Radiation853.30252.65603.680.0001
   Hormonal therapy136653.0366370.2370343.080.1079
Pathology
   adenocarcinoma105082.07103783.03101381.100.2107
   no adenocarcinoma44817.9321216.9723618.90
Charlson comorbidity index
   082432.9941232.9941232.991
   1115446.2057746.2057746.20
   2+52020.8226020.8226020.82
Survival(death)25810.3316112.44977.50<0.0001
Cancer death
   Prostate cancer16463.579559.016971.130.1426
   other cancer228.53159.3277.22
   Other7227.915131.682121.65
Survival year (mean, SD)5.06(2.75)5.11(2.10)5.01(2.05)0.2034
Dementia
   Parkinson14711.7714711.77--
   Alzheimer53342.6753342.67--
   Vascular dementia21417.1321417.13--
   Other dementia35528.4235528.42--
ADT treatment
   only GnRH agonist1385.52655.20735.840.2844
   only Antiandrogen833.32453.60383.04
   Both114545.8455344.2859247.40
   No ADT113245.3258646.9254643.71
Duration of ADT (days, mean (SD))
   Total1049.28(756.35)843.86(613.88)1243.00(824.34)<0.0001
   only Antiandrogen735.86(625.31)640.72(542.19)820.56(683.56)0.0876
   only GnRH agonist849.06(622.95)711.06(485.17)1012.47(727.69)0.0332
   Both1101.57(768.40)878.55(625.77)1309.89(828.94)<0.0001
ADT duration (days, mean (SD))
   No treatment----
   3–6 months125.73 (28.77)126.25(27.22)124.89(31.53)0.8226
   6–12 months270.24 (55.79)273.85(52.67)264.96(60.00)0.2730
   12–24 months531.79 (102.10)531.79(106.7)531.79(95.65)0.9999
   ≤24 months1533.30 (628.15)1337.40(506.5)1668.20(667.6)<0.0001
Interval time from cancer diagnosis to ADT start (days, mean (SD))
   Total117.04(293.25)89.35(227.21)143.16(342.24)0.0006
   only Antiandrogen248.36(503.14)116.00(267.39)366.21(623.09)0.0023
   only GnRH agonist261.52(170.00)117.62(194.10)232.03(315.42)0.0564
   Both97.38(254.40)83.92(224.59)109.95(279.00)0.0813
NA, not applicable; ADT, androgen-deprivation therapy; SEER, Surveillance, Epidemiology, and End Results; SD, standard deviation; GnRH, Gonadotrophin-releasing hormone; and BMI, body mass index.
Table 4. Multivariate cox regression of risk factors for dementia among local/regional SEER staging patients after propensity-score matching.
Table 4. Multivariate cox regression of risk factors for dementia among local/regional SEER staging patients after propensity-score matching.
UnivariateMultivariate
ParameterHazard Ratio95% Hazard Ratio
Confidence Limits
Pr > ChiSqHazard Ratio95% Hazard Ratio
Confidence Limits
Pr > ChiSq
Age group (year-old)
   50–64ref ref
   65–742.1551.5622.973<0.00011.9651.4222.717<0.0001
   75-2.0021.4632.74<0.00012.8601.9054.293<0.0001
BMI (kg/cm2)
   Normal (18.5~22.9)ref ref
   Underweight (<18.5)1.3670.8062.3190.24621.3060.7682.2190.3246
   Overweight (23~24.9)0.8490.6761.0670.16010.8550.6801.0750.1799
   Obesity (25~29.9)0.720.5730.9030.00450.7110.5640.8960.0039
   Severe obesity. (30~)0.7910.4181.4980.47160.7690.4051.4610.4229
Smoking
   non smokerref
   Ex-smoker0.7210.51.040.797
   current smoker0.9240.6951.2290.5863
Aspirin (ref = no)1.3071.0871.570.00431.1120.8941.3840.3403
Statin (ref = no)1.020.8261.2590.8563
HTN (ref = no)1.3561.1281.6310.00121.1780.9441.4710.1469
Diabetes (ref = no)1.1390.931.3960.2089
Charlson comorbidity index(CCI)
   0ref ref
   11.5491.2641.899<0.00011.4681.1941.8040.0003
   2+1.8491.42.442<0.00011.7161.2912.2810.0002
ADT TYPE
   No treatmentref
   only Antiandrogen0.7340.4991.0780.1151
   only GnRH agonist0.9420.5681.5620.8158
   Both0.960.7831.1780.6985
ADT duration
   No treatmentref ref
   3–6 months0.6310.3891.0240.06240.3880.2240.6730.0008
   6–12 months1.0220.7331.4260.89740.6260.4100.9550.0296
   12–24 months0.7350.5331.0140.06070.4510.2980.6830.0002
   ≤24 months1.30.9931.7020.05610.8030.5521.1680.2506
Table 5. Multivariate cox regression of risk factors for dementia among distant/unknown SEER staging patients after propensity-score matching.
Table 5. Multivariate cox regression of risk factors for dementia among distant/unknown SEER staging patients after propensity-score matching.
UnivariateMultivariate
ParameterHazard Ratio95% Hazard Ratio
Confidence Limits
Pr > ChiSqHazard Ratio95% Hazard Ratio
Confidence Limits
Pr > ChiSq
Age group (year-old)
   50–64ref ref
   65–744.5031.3614.9120.01384.0251.20813.4090.2736
   75–6.7792.1221.6780.00137.9952.15929.6040.0013
BMI (kg/cm2)
   Normal (18.5~22.9)ref ref
   Underweight (<18.5)2.9731.2437.110.01433.4251.4218.2580.0061
   Overweight (23~24.9)0.7060.3921.2710.24550.7820.4311.4210.4196
   Obesity (25~29.9)0.9850.5961.6290.9541.0610.6321.7810.8226
   Severe obesity. (30~)0.4130.0563.0230.38390.4350.0593.1940.4129
Smoking
   non smokerref
   Ex-smoker0.7810.2472.4750.6748
   current smoker0.2850.090.9040.033
Aspirin (ref = no)1.2040.7781.8620.4051
Statin (ref = no)1.6481.0282.6430.03811.1720.6931.9840.0159
HTN (ref = no)1.7711.152.7280.00951.4180.8642.3250.5847
Diabetes (ref = no)1.5961.0062.5320.0471.0800.6384.2720.4268
Charlson comorbidity index(CCI)
   0ref ref
   12.0941.2353.5530.00611.9191.1123.3130.0193
   2+2.5371.3754.680.00292.2011.1344.2720.0197
ADT TYPE
   No treatmentref
   only Antiandrogen1.1540.4073.2760.7879
   only GnRH agonist2.1880.7716.2130.1413
   Both1.8151.132.9150.0137
ADT duration
   No treatmentref ref
   3–6 months0.660.1492.9140.58320.3000.0611.4670.1370
   6–12 months0.8770.3552.1680.77650.3920.1361.1320.0836
   12–24 months1.3880.7232.6640.32430.6010.2561.4130.2433
   ≤24 months2.7721.5434.980.00061.2770.5712.8590.5516
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kim, Y.A.; Kim, S.-H.; Joung, J.Y.; Yang, M.S.; Back, J.H.; Kim, S.H. The Insignificant Correlation between Androgen Deprivation Therapy and Incidence of Dementia Using an Extension Survival Cox Hazard Model and Propensity-Score Matching Analysis in a Retrospective, Population-Based Prostate Cancer Registry. Cancers 2022, 14, 2705. https://doi.org/10.3390/cancers14112705

AMA Style

Kim YA, Kim S-H, Joung JY, Yang MS, Back JH, Kim SH. The Insignificant Correlation between Androgen Deprivation Therapy and Incidence of Dementia Using an Extension Survival Cox Hazard Model and Propensity-Score Matching Analysis in a Retrospective, Population-Based Prostate Cancer Registry. Cancers. 2022; 14(11):2705. https://doi.org/10.3390/cancers14112705

Chicago/Turabian Style

Kim, Young Ae, Su-Hyun Kim, Jae Young Joung, Min Soo Yang, Joung Hwan Back, and Sung Han Kim. 2022. "The Insignificant Correlation between Androgen Deprivation Therapy and Incidence of Dementia Using an Extension Survival Cox Hazard Model and Propensity-Score Matching Analysis in a Retrospective, Population-Based Prostate Cancer Registry" Cancers 14, no. 11: 2705. https://doi.org/10.3390/cancers14112705

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop