Analiză din lumea reală privind cancerul de prostată hormonosensibil metastatic de novo: experiența pe parcursul a trei ani a unui centru regional din România
Real-world analysis on de novo hormone-sensitive metastatic prostate cancer: three years’ experience of a regional center in Romania
Data primire articol: 28 Noiembrie 2025
Data acceptare articol: 09 Decembrie 2025
Editorial Group: MEDICHUB MEDIA
10.26416/OnHe.73.4.2025.11287
Descarcă pdf
Abstract
Introduction. We present the results of a retrospective study exploring the characteristics of patients with de novo metastatic hormone-sensitive metastatic prostate cancer (mHSPC). The main objective was to describe the treatment options and outcomes in current practice in patients with mHSPC treated at the Bihor County Emergency Clinical Hospital, Oradea, Romania. Materials and method. Data were collected from the charts of patients with de novo mHSPC registered at the Bihor County Emergency Clinical Hospital, Oradea, between January 2021 and December 2023. Statistics were descriptive. Results. Patient characteristics (n=88): median age 72 years old, median PS (performance status) 2, median baseline PSA (prostate specific antigen) 126 ng/mL (values available for 87/88 patients). Tumor characteristics: median Gleason score 8 (values available for 83/88 patients), most of the patients had bone metastases on diagnosis, approximately 60% presented high-volume disease, according to CHAARTED criteria. Eighty-eight patients were treated with androgen-deprivation therapy (ADT), 60 (68.22%) with monotherapy, six (6.81%) in combination with docetaxel, five (5.68%) with abiraterone, 13 (14.77%) with apalutamide, and four (4.54%) with the triple combination of ADT, docetaxel and abiraterone. Eight patients (9.09%) also received local radiotherapy after the initiation of ADT. Median PSA after six months of treatment was 0.465 ng/mL (values available for 62/88 patients). At data analysis (December 2024), the median time to progression to castration-resistant metastatic stage was 13 months, calculated for 82 of the patients, six patients were lost to follow-up, and 20 patients had not progressed to castration-resistant metastatic stage. Conclusions. This real-world analysis provided information about the characteristics of patients with de novo mHSPC and about the management of this pathology in the practice of a regional center in Romania. Considering the therapeutic advances, the identification of the treatment regimen appropriate to the patient profile is essential.
Keywords
de novo metastatic hormone-sensitive prostate cancerreal-world datatreatment patternstriple therapytreatment intensificationRezumat
Scop. Prezentăm rezultatele unui studiu retrospectiv, care a explorat caracteristicile pacienților cu cancer de prostată hormonosensibil metastatic (CPHSm) de novo. Principalul obiectiv a fost de a descrie opțiunile de tratament și rezultatele în practica curentă la pacienții cu CPHSm tratați în cadrul Spitalului Clinic Județean de Urgență Bihor, Oradea. Materiale şi metodă. Datele au fost colectate din fișele pacienților cu CPHSm de novo luați în evidență în Spitalul Clinic Județean de Urgență Bihor, Oradea, în perioada ianuarie 2021 – decembrie 2023. Statistica a fost descriptivă. Rezultate. Caracteristicile pacienților (n=88): vârsta mediană 72 de ani, SP (status de performanță) median 2, PSA (antigen specific prostatic) inițial median 126 ng/mL (valori disponibile pentru 87/88 de pacienți). Caracteristici tumorale: scor Gleason median 8 (valori disponibile pentru 83/88 de pacienți), majoritatea pacienților au avut metastaze osoase, aproximativ 60% au prezentat boală high-volume conform criteriilor CHAARTED. 88 de pacienți au fost tratați cu terapie de deprivare androgenică (TDA), 60 (68,22%) în monoterapie, şase (6,81%) în combinație cu docetaxel, cinci (5,68%) cu abirateronă, 13 (14,77%) cu apalutamidă și patru (4,54%) cu combinația triplă de TDA, docetaxel și abirateronă. Opt pacienți (9,09%) au beneficiat și de radioterapie locală, după inițierea TDA. PSA median după şase luni de tratament a fost de 0,465 ng/mL (valori disponibile pentru 62/88 de pacienți). La data analizei (decembrie 2024), timpul median până la progresia la stadiul metastatic rezistent la castrare a fost de 13 luni, calculat pentru 82 de pacienți, şase au fost pierduți din urmărire, iar 20 nu progresaseră la acest stadiu. Concluzii. Această analiză din lumea reală a adus informații despre caracteristicile pacienților cu CPHSm de novo și despre managementul acestei patologii în practica unui centru regional din România. În contextul progreselor terapeutice, alegerea unei scheme de tratament adaptate profilului fiecărui pacient este esențială.
Cuvinte Cheie
cancer de prostată hormonosensibil metastatic de novodate din lumea realătipare de tratamentterapie triplăintensificarea tratamentului1. Introduction
Prostate cancer is the second most frequently diagnosed malignancy among men worldwide, with an estimated 1.2 million new cases and 359,000 deaths annually(1). The clinical spectrum of prostate cancer ranges from localized disease to advanced metastatic forms, and therapeutic strategies have evolved considerably over the recent years, now encompassing active screening and surveillance, surgery, radiotherapy, hormone therapy, chemotherapy, targeted agents and bone health interventions(2,3). Despite these advances, metastatic hormone-sensitive prostate cancer (mHSPC) remains a significant unmet medical need, as patients with metastatic disease continue to experience poor outcomes and limited survival(3).
Since 2015, several clinical trials have transformed the management of mHSPC, demonstrating survival benefits for agents such as docetaxel(4,5), abiraterone(5,6), apalutamide(7), enzalutamide(8,9) and darolutamide(10), leading to their inclusion into international guidelines(11,12). More recently, triplet therapy and intensified androgen deprivation therapy (ADT) regimens, including combinations with abiraterone or darolutamide, have shown efficacy and safety in studies such as PEACE-1 and ARASENS, and are now recommended as standard of care(13-15). Radiotherapy has also emerged as a promising therapeutic pathway for patients with low metastatic burden, as evidenced by the HORRAD and STAMPEDE trials, as well as by real-world analyses(3,5,16).
However, the populations enrolled in clinical trials often differ substantially from those encountered in routine clinical practice. Trial participants typically have better performance status, fewer comorbidities and better organ function, which may limit the replication of trial findings to the broader patient population(3,17). In fact, only a small fraction of patients (estimated at 3% in the United States of America) participate in clinical trials, with underrepresentation of elderly, low-income and minority groups(18). Strict eligibility criteria further exclude patients with poor performance status or significant comorbidities, resulting in a gap between trial evidence and real-world practice(17).
Real-world data (RWD) analyses are increasingly recognized as essential for bridging this gap, providing insights into treatment patterns, effectiveness and safety in unselected patient populations(17,18). RWD can validate and expand upon clinical trial findings, inform regulatory decisions and guide the design of future studies, particularly by highlighting the challenges and limitations of current therapeutic approaches in everyday practice(19). Nevertheless, there remains a lack of published data regarding prostate cancer incidence, treatment patterns and risk factors specific to the Romanian population, hindering efforts to optimize care and inform policy.
In this context, our study aims to address these knowledge gaps by presenting real-world treatment patterns and early outcomes for patients with de novo mHSPC in a Romanian county-level referral hospital. By analyzing unselected patient cohorts and reporting on actual clinical practice, our findings offer a more realistic perspective on the management of mHSPC in Romania, with the potential to inform both local and national therapeutic strategies.
2. Materials and method
2.1. Study design and setting
A single-center, observational, retrospective cohort study was conducted at the Bihor County Emergency Clinical Hospital, Oradea, Romania, including patients diagnosed with de novo metastatic hormone-sensitive prostate cancer between 1 January 2021 and 31 December 2023. The data cutoff for this analysis was 31 December 2023. The study setting is a county-level referral institution providing multidisciplinary oncologic care through collaborative partnerships with urology, radiology, radiation oncology and palliative care services. Data were collected from anonymized files from the database of the hospital.
2.2. Study population and eligibility criteria
Eligible patients were adults (aged >18 years old) with histologically confirmed prostate adenocarcinoma and evidence of metastatic disease at initial diagnosis, as verified by medical imaging, including computed tomography (CT), magnetic resonance imaging (MRI) and bone scintigraphy when available. Patients presenting with metachronous metastatic disease, defined as relapse following prior localized therapy, were excluded from the analysis.
2.3. Patient characteristics and treatment variables
Baseline patient data included demographic information, Eastern Cooperative Oncology Group (ECOG) performance status, comorbidity profiles, Gleason/International Society of Urological Pathology (ISUP) grade, initial prostate-specific antigen (PSA) levels, TNM staging, sites of metastasis (classified as M1a, M1b, or M1c), and disease volume as defined by the CHAARTED criteria. Treatment-related variables encompassed the initial androgen deprivation therapy (ADT) modality (medical castration, surgical castration not performed), treatment intensification (including androgen receptor pathway inhibitor – ARPI, type and number of docetaxel cycles administered), administration of prostate-directed radiotherapy, and documented reasons for limiting treatment intensification (such as ECOG performance status, recent cardiovascular or cerebrovascular events, patient preference, or reimbursement constraints).
2.4. Study outcomes
The study was designed with three key objectives. The primary objective was to characterize real-world first-line treatment patterns for patients with de novo metastatic hormone-sensitive prostate cancer at a Romanian county-level referral hospital, encompassing therapies initiated at diagnosis and during follow-up, and stratified by clinical subgroups such as disease volume and risk category. The secondary objective was to evaluate early clinical outcomes associated with first-line treatment selection, including time to castration resistance (TTCR), prostate-specific antigen response rates, and imaging-based assessments where available. The third exploratory objective was to delineate diagnostic pathways and baseline patient characteristics pertinent to treatment decision-making, including disease volume, risk stratification and performance status.
2.5. Statistical analysis
Descriptive statistics were utilized to summarize patient demographics, disease characteristics and treatment patterns. Medians were reported where estimable; otherwise, event counts and follow-up status were detailed. Time-to-event outcomes were analyzed using institutional Kaplan-Meier survival estimates. Exploratory Cox proportional hazards models were employed to assess the potential prognostic impact of variables such as Gleason grade, metastatic category (M category), disease volume, comorbidity burden and treatment intensification. Given the limited sample size, these analyses are considered hypothesis-generating and should be interpreted with caution due to potential confounding. All descriptive statistical analyses were conducted using Microsoft Excel spreadsheet software. Kaplan-Meier survival analyses were performed using R software (R Foundation for Statistical Computing, Vienna, Austria).
2.6. Ethical statement
Due to the retrospective and observational, non-interventional design of the study, the requirement for patient informed consent was waived. All study procedures were conducted in accordance with the ethical standards set forth in the Declaration of Helsinki.
Results
3.1. Patient characteristics
A total of 286 individuals were diagnosed with prostate cancer at the regional center between January 2021 and December 2023, of whom 88 presented with de novo mHSPC. The median age among the de novo mHSPC cohort was 72 years old (range: 55-91). ECOG performance status was distributed as follows: eight patients with a score of 0; 25 with a score of 1; 18 with a score of 2; 34 with a score of 3; three patients with a score of 4; 11 patients had a recent major cardiovascular event. The median baseline PSA level was 126 ng/mL (data available for 87 patients; range: 5.4-2400 ng/mL).
3.2. Tumor characteristics
Tumor aggressiveness was evaluated using the Gleason grading system, with scores available for 83 patients. Of these, six patients had a Gleason score of 6, 25 had a score of 7, 22 had a score of 8, 22 had a score of 9, and eight patients had a score of 10. At baseline, bone metastases were identified in 68 patients, 12 exhibited extra-pelvic lymph node involvement, and eight had visceral metastases. According to CHAARTED criteria, approximately 70% of patients were classified as having high-volume disease, while 58% met high-risk criteria as defined by the LATITUDE trial. The summarized patient data can be found in Table 1.
3.3. Treatment patterns and outcomes
The median interval from biopsy to initial oncology consultation was 26.5 days (range: 8-91 days), and the median time from de novo mHSPC diagnosis to the initiation of therapy was one day (range: 1-80 days). All patients with de novo mHSPC received androgen deprivation therapy (one of the following: goserelin, triptorelin, leuprorelin) – 60 (68.2%) as monotherapy, six (6.81%) in combination with docetaxel (mean number of docetaxel cycles was 5.7), five (5.7%) with abiraterone, 13 (14.8%) with apalutamide, and four (4.5%) with the triple combination of ADT, docetaxel and abiraterone. Eight patients (9.1%) from the total cohort of 88 de novo mHSPC cases with low-volume disease underwent local radiotherapy, seven received ADT monotherapy, and one received ADT plus apalutamide. No surgical castration was performed, and first-generation antiandrogens (bicalutamide, flutamide) were not used. Median progression-free survival (mPFS) and TTCR for each treatment line can be found in Table 2. Six patients were lost to follow-up, and 20 had not progressed to the castration-resistant stage at the time of analysis; at the time of the data cutoff, 19 patients were deceased, all of them being on the ADT monotherapy treatment line. The median PSA level at six months post-treatment initiation was 0.465 ng/mL (data available for 62 patients; range: 0-442 ng/mL). The mPSA values at six months for each therapy line can be found in Table 3.
3.4. Kaplan-Meier analysis results
Kaplan-Meier survival analysis was conducted to evaluate the time to the development of metastatic castration-resistant prostate cancer (mCRPC) in the study cohort, with results stratified by several clinical and pathological variables.
When patients were grouped according to Gleason score categories (≤6; 7; ≥8), the analysis demonstrated a statistically significant difference in mCRPC-free survival among the groups. Patients with a Gleason score ≥8 exhibited the shortest median mCRPC-free survival, followed by those with a score of 7, while patients with a score ≤6 had the longest survival without progression to mCRPC (p=0.04). The distribution of events and the separation of survival curves indicated a clear association between higher Gleason scores and earlier onset of castration resistance (Figure 1).
When analyzing mCRPC-free survival by metastasis type (M1A, M1B, M1C), the Kaplan-Meier (KM) curves showed no statistically significant difference between the groups (log-rank p=0.65). Although prior boxplot summaries suggested a possible gradient (M1C > M1B > M1A) in time to progression, this visual difference was not supported by time-to-event statistics once censoring and follow-up time were properly accounted for in the KM model.
Six patients with M1B disease were excluded from the survival analysis because they were lost to follow-up before any evaluable event time (i.e., mcrpc_time missing). These censored cases could not contribute to the survival estimate, explaining the slightly lower M1B sample size (63 instead of 69). Boxplots summarized observed times only, while KM analysis correctly accounted for censored follow-up, providing a more accurate estimate of mCRPC-free survival probability.
Thus, even though the raw progression times appear shorter in M1C and M1B on simple plots, the KM log-rank test indicates that these differences are not statistically significant in this dataset (Figure 2).
The cohort was also analyzed based on metastatic disease volume at baseline, classified as low-volume or high-volume disease. Kaplan-Meier curves revealed that individuals with high-volume metastatic disease experienced a more rapid progression to mCRPC compared to those with low-volume disease. The difference in mCRPC-free survival between these two groups was statistically significant (p<0.001), with the high-volume group showing a steeper decline in survival probability over time (Figure 3).
Treatment regimens were compared by grouping patients who received ADT alone and those who received intensified regimens, which included ADT in combination with docetaxel and/or androgen receptor pathway inhibitors. The Kaplan-Meier analysis showed that the group treated with intensified therapy had a significantly longer duration of mCRPC-free survival compared to the ADT monotherapy group (p=0.038). The survival curves for these groups diverged early after treatment initiation and remained separated throughout the follow-up period. PSA response at six months post-treatment initiation was evaluated as a prognostic factor (Figure 4).
Patients with initial PSA≥20 ng/mL showed a trend toward shorter mCRPC-free survival compared with those below 20, but this did not reach statistical significance (p=0.093). This suggests that, while higher baseline PSA may be associated with earlier progression, the current sample size may limit the power to detect a significant effect (Figure 5).
Patients were stratified into three categories based on PSA levels at six months: ≤0.2 ng/mL, 0.2-4 ng/mL, and ≥4 ng/mL. The analysis demonstrated that patients with PSA levels ≤0.2 ng/mL at six months had the longest mCRPC-free survival, whereas those with PSA levels ≥4 ng/mL had the shortest survival (p<0.001). The intermediate group (0.2-4 ng/mL) showed survival outcomes between the other two groups. The survival curves were clearly separated and remained distinct throughout the observation period (Figure 6).
Finally, mCRPC-free survival was assessed according to LATITUDE risk classification, which categorizes patients as high-risk or low-risk based on specific clinical criteria. Patients classified as high-risk had a significantly shorter time to mCRPC compared to those in the low-risk group (p=0.002). The survival analysis showed a distinct difference in the timing of progression between these risk categories (Figure 7).
Across all subgroup analyses, the log-rank test was used to assess statistical significance, and the results consistently indicated that higher Gleason score, high-volume metastatic disease, absence of treatment intensification, higher PSA at six months and high-risk LATITUDE classification were each associated with reduced mCRPC-free survival.
Discussion
4.1. Overview
This single-center, retrospective cohort study describes real-world first-line management and early outcomes for patients with de novo mHSPC in a Romanian county referral hospital. The cohort was characterized by older age, higher disease burden and substantial comorbidity, reflecting a population that differs from those typically enrolled in phase III clinical trials. These findings are consistent with international real-world data, where delayed presentation to medical consultations and system-level constraints often limit the adoption of guideline-recommended treatment intensification(11,12,19-21).
4.2. Treatment patterns and barriers
A considerable proportion of patients received ADT monotherapy. This was influenced by several factors:
- Patient-level factors – frailty, high ECOG performance status, recent cardiovascular or cerebrovascular events limited eligibility for intensified therapy.
- System-level constraints – reimbursement issues and pandemic-related disruptions restricted access to novel agents and combination regimens(22,23).
- Disease biology and timing – advanced disease volume, high PSA levels and visceral involvement at presentation reduced the opportunity for clinical stabilization and treatment intensification.
These barriers are consistent with reports from other real-world settings, where older age, comorbidities and access limitations are associated with lower rates of treatment intensification(24-26).
4.3. Prognostic factors and treatment outcomes
Several independent risk factors for earlier progression to mCRPC were identified, including high Gleason grade (8-10), high-volume disease and major comorbidities. These findings align with previous studies demonstrating the adverse prognostic impact of these features(20,27,28). When comparing mCRPC-free survival across different metastasis types (M1A, M1B, M1C), our analysis did not reveal any statistically significant differences between the groups (log-rank p=0.65). While initial boxplot visualizations suggested a possible gradient in progression times (with M1C appearing to progress faster than M1B and M1A), these apparent differences were not confirmed by the Kaplan-Meier survival analysis, which properly accounted for censored data and variable follow-up durations. The exclusion of six M1B patients who were lost to follow-up before any evaluable event further underscores the importance of using time-to-event methods for accurate survival estimates. Overall, although raw data plots may hint at shorter progression times in certain subgroups, the more robust KM analysis indicates that these differences are not statistically significant in our current dataset. Treatment intensification with ADT plus an ARPI and/or docetaxel was associated with longer time to castration resistance, consistent with results from randomized trials such as CHAARTED, STAMPEDE, LATITUDE, TITAN, ARCHES, ENZAMET and triplet therapy(5-10,20,28). Although subgroup sizes were limited and follow-up is ongoing, the observed benefit supports the use of intensified regimens in eligible patients.
4.4. PSA levels as a prognostic marker
PSA levels at six months post-treatment initiation were a strong prognostic indicator. Patients achieving a PSA≤0.2 ng/mL had significantly longer mCRPC-free survival, while those with PSA≥4 ng/mL experienced earlier progression. This supports the use of early PSA response as a practical marker for prognosis and treatment monitoring(29-32).
Although patients with an initial PSA of 20 ng/mL or higher appeared to experience earlier progression to mCRPC, this trend did not achieve statistical significance in our analysis (log-rank p=0.093). This observation suggests that a higher baseline PSA may be linked to a greater risk of earlier disease progression; however, the lack of statistical significance indicates that our study may not have had sufficient sample size or power to confirm this association definitively. These findings highlight the need for further research with larger cohorts to clarify the prognostic value of baseline PSA in predicting mCRPC-free survival.
4.5. Clinical implications
The findings highlight the importance of:
- Prompt staging and early supportive optimization to improve eligibility for treatment intensification.
- Standardized documentation of disease volume and risk classification to guide therapy decisions.
- Expanding access to ARPIs beyond the current narrow criteria to allow more patients to benefit from evidence-based doublet therapy(33-35).
- Integrating prostate-directed radiotherapy for low-volume disease in coordination with systemic therapy(36,37).
4.6. Limitations
This study is limited by its retrospective, single-center design and small subgroup sizes, particularly for triplet therapy. Imaging modalities varied, which may have affected staging accuracy. The adverse events were not systematically recorded, and confounding by indication is possible, as fitter patients were more likely to receive intensified therapy. These analyses should be considered hypothesis-generating.
4.7. Future directions
Further research should focus on multicenter registry collaborations with standardized data collection to enable more robust comparisons of treatment strategies. Incorporating genomic profiling and evaluating care pathways that support treatment intensification after functional recovery may improve access to effective therapies. Public health initiatives to promote earlier detection could reduce the proportion of de novo metastatic presentations and improve outcomes.
5. Conclusions
This retrospective, single-center study provides a real-world perspective on the management of de novo metastatic hormone-sensitive prostate cancer in a Romanian county referral hospital. The findings highlight that a substantial proportion of patients present with advanced disease, significant comorbidities and limited functional status, which often restricts the use of treatment intensification strategies recommended by international guidelines. Despite these challenges, intensified therapy with androgen receptor pathway inhibitors and/or docetaxel was associated with improved time to castration resistance, consistent with evidence from randomized clinical trials.
Early PSA response at six months emerged as a strong prognostic marker, supporting its use in routine monitoring and risk stratification. The study also identified high Gleason grade, high-volume disease, visceral metastases and comorbidity burden as key predictors of earlier progression to castration-resistant disease. These results underscore the need for prompt diagnosis, comprehensive supportive care and standardized treatment pathways to optimize outcomes for patients with de novo mHSPC.
Expanding access to novel therapies and integrating multidisciplinary care are important steps toward improving real-world outcomes. Future research should focus on multicenter collaborations, standardized data collection and strategies to increase early detection and equitable access to evidence-based treatments.
Acknowledgements. The authors would like to thank AstraZeneca for providing support with the statistical analyses included in this manuscript.
Corresponding author: Lajos Szilard Bereczki E-mail: bereczki.szilard77@gmail.com
Conflict of interest: none declared.
Financial support: none declared.
This work is permanently accessible online free of charge and published under the CC-BY licence.
Bibliografie
-
Culp MB, Soerjomataram I, Efstathiou JA, Bray F, Jemal A. Recent global patterns in prostate cancer incidence and mortality rates. Eur Urol. 2020;77(1):38-52.
-
Sayegh N, Swami U, Agarwal N. Recent advances in the management of metastatic prostate cancer. JCO Oncol Pract. 2022;18(1):45-55.
-
George DJ, Sartor O, Miller K, Saad F, Tombal B, Kalinovský J, Jiao X, Tangirala K, Sternberg CN, Higano CS. Treatment patterns and outcomes in patients with metastatic castration-resistant prostate cancer in a real-world clinical practice setting in the United States. Clin Genitourin Cancer. 2020;18(4):284-294.
-
Hamid AA, Huang HC, Wang V, et al. Transcriptional profiling of primary prostate tumor in metastatic hormone-sensitive prostate cancer and association with clinical outcomes: correlative analysis of the E3805 CHAARTED trial. Ann Oncol. 2021;32(9):1157-1166.
-
James ND, Clarke NW, Cook A, et al. Abiraterone acetate plus prednisolone for metastatic patients starting hormone therapy: 5-year follow-up results from the STAMPEDE randomised trial (NCT00268476). Int J Cancer. 2022;151(3):422-434.
-
Baciarello G, Özgüroğlu M, Mundle S, et al. Impact of abiraterone acetate plus prednisone in patients with castration-sensitive prostate cancer and visceral metastases over four years of follow-up: A post-hoc exploratory analysis of the LATITUDE study. Eur J Cancer. 2022;162:56-64.
-
Roy S, Sun Y, Chi KN, et al. Early Prostate-Specific Antigen Response by 6 Months Is Predictive of Treatment Effect in Metastatic Hormone Sensitive Prostate Cancer: An Exploratory Analysis of the TITAN Trial. J Urol. 2024;212(5):672-681.
-
Armstrong AJ, Shore ND, Szmulewitz RZ, et al. Efficacy of Enzalutamide plus Androgen Deprivation Therapy in Metastatic Hormone-Sensitive Prostate Cancer by Pattern of Metastatic Spread: ARCHES Post Hoc Analyses. J Urol. 2021;205(5):1361-1371.
-
Sweeney CJ, Martin AJ, Stockler MR, et al. Testosterone suppression plus enzalutamide versus testosterone suppression plus standard antiandrogen therapy for metastatic hormone-sensitive prostate cancer (ENZAMET): an international, open-label, randomised, phase 3 trial. Lancet Oncol. 2023;24(4):323-334.
-
Saad F, Vjaters E, Shore N, et al. Darolutamide in Combination with Androgen-Deprivation Therapy in Patients with Metastatic Hormone-Sensitive Prostate Cancer. J Clin Oncol. 2024;42(36):4271-4281.
-
Fizazi K, Gillessen S. Updated treatment recommendations for prostate cancer from the ESMO Clinical Practice Guideline considering treatment intensification and use of novel systemic agents. Ann Oncol. 2023;34(6):557-563.
-
NCCN Guidelines. Prostate Cancer. https://www.nccn.org/guidelines/guidelines-detail?id=1459
-
Foulon S, Maldonado X, Sargos P, et al. Prostate irradiation in men with de novo, low-volume, metastatic, castration-sensitive prostate cancer. J Clin Oncol. 2023;41(17_suppl):LBA5000-LBA5000.
-
Hussain M, Tombal B, Saad F, et al. Darolutamide Plus Androgen-Deprivation Therapy and Docetaxel in Metastatic Hormone-Sensitive Prostate Cancer. J Clin Oncol. 2023;41(20):3595-3607.
-
Roberts HN, Maurice-Dror C, Chi KN. Unpacking prostate-specific antigen dynamics and patient outcomes: insights from the ARASENS trial. AME Clin Trials Rev. 2025;3-10.
-
Boevé LMS, Hulshof MCCM, Verhagen PCMS, et al. Prostate Cancer-related Events in Patients with Synchronous Metastatic Hormone-sensitive Prostate Cancer Treated with Androgen Deprivation Therapy with and Without Concurrent Radiation Therapy to the Prostate; Data from the HORRAD Trial. Eur Urol. 2025;87(3):357-363.
-
Swami U, Sinnott JA, Haaland B, et al. Treatment Pattern and Outcomes with Systemic Therapy in Men with Metastatic Prostate Cancer in the Real-World Patients in the United States. Cancers. 2021;13(19):4951.
-
Burstein HJ, Krilov L, Aragon-Ching JB, et al. Clinical Cancer Advances 2017: Annual Report on Progress Against Cancer from the American Society of Clinical Oncology. J Clin Oncol. 2017;35(12):1341-1367.
-
Wilson BE, Booth CM. Real-world data: bridging the gap between clinical trials and practice. eClinicalMedicine. 2024;78:102915.
-
Sweeney CJ, Chen YH, Carducci M, et al. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer. N Engl J Med. 2015;373(8):737-746.
-
Attard G, Murphy L, Clarke NW, et al. Abiraterone acetate and prednisolone with or without enzalutamide for high-risk non-metastatic prostate cancer: a meta-analysis of primary results from two randomised controlled phase 3 trials of the STAMPEDE platform protocol. Lancet. 2022;399(10323):447-460.
-
Mostafavi Zadeh SM, Tajik F, Gheytanchi E, Kiani J, Ghods R, Madjd Z. COVID-19 pandemic impact on screening and diagnosis of prostate cancer: a systematic review. BMJ Support Palliat Care. 2024;14(e2):e1594.
-
Mangone L, Marinelli F, Bisceglia I, et al. Impact of the COVID-19 Pandemic on Prostate Cancer Diagnosis, Staging, and Treatment: A Population-Based Study in Northern Italy. Biology. 2024;13(7):499.
-
Zhou B, Raval AD, Zhang Y, et al. Treatment landscape for older men with metastatic hormone-sensitive prostate cancer in the United States. Cancer Med. 2025;14(17):e71176.
-
Raval AD, Lunacsek O, Korn MJ, Littleton N, Constantinovici N, George DJ. Real-world evidence of combination therapy use in metastatic hormone-sensitive prostate cancer. JCO Oncol Pract. 2025;21(8):1174-1184.
-
Goebell PJ, Raina R, Chen S, Rege S, Shah R, Grossman JP, Waldeck AR. Real-world treatment of metastatic hormone-sensitive prostate cancer in the USA, Europe and Asia. Future Oncol. 2024;20(14):903-918.
-
Halabi S, Lin CY, Kelly WK, Fizazi KS, Moul JW, Kaplan EB, Morris MJ, Small EJ. Updated prognostic model for predicting overall survival in first-line chemotherapy for patients with metastatic hormone-sensitive prostate cancer. J Clin Oncol. 2014;32(7):671-677.
-
Armstrong AJ, Szmulewitz RZ, Petrylak DP, et al. ARCHES: A randomized, phase III study of androgen deprivation therapy with enzalutamide or placebo in men with metastatic hormone-sensitive prostate cancer. J Clin Oncol. 2019;37(32):2974-2986.
-
Hussain M, Tangen CM, Higano C, et al. Absolute prostate-specific antigen value after androgen deprivation is a strong independent predictor of survival in metastatic prostate cancer. J Clin Oncol. 2006;24(24):3984-3990.
-
Pezaro C, Woo HH, Davis ID. Prostate cancer: measuring PSA. Intern Med J. 2014;44(5):433-440.
-
Grimm P, Billiet I, Bostwick D, et al. Comparative analysis of prostate-specific antigen free survival outcomes for patients with low, intermediate and high risk prostate cancer treatment by radical therapy. Results from the Prostate Cancer Results Study Group. BJU Int. 2012;109:22-29.
-
Wenzel M, Cano Garcia C, Humke C, et al. Prostate-specific Antigen Nadir and Cancer-Control Outcomes in Real-world Apalutamide-treated Metastatic Hormone-Sensitive Prostate Cancer Patients: A Single-Center Analysis. EurUrol Oncol. 2025;8(2):364-371.
-
Mir N, Burke O, Yates S, et al. Androgen receptor pathway inhibitors, prostate cancer, and older adults: a global Young International Society of Geriatric Oncology drug review. Ther Adv Med Oncol. 2023;15:1-12.
-
Brown JR, Shore N, Barata PC. In the Era of Treatment Intensification for Prostate Cancer, Why Are Some Living in the Past?. JAMA Netw Open. 2024;7(12):e2448660.
-
Francini E, Agarwal N, Castro E, et al. Intensification Approaches and Treatment Sequencing in Metastatic Castration-resistant Prostate Cancer: A Systematic Review. Eur Urol. 2025;87(1):29-46.
-
Spratt DE, Malone S, Roy S, et al. Prostate Radiotherapy with Adjuvant Androgen Deprivation Therapy (ADT) Improves Metastasis-Free Survival. J Clin Oncol. 2021;39(2):136-144.
-
Imber BS, Varghese M, Goldman DA, et al. Clinical Outcomes of Combined Prostate- and Metastasis-Directed Radiation Therapy for the Treatment of De Novo Oligometastatic Prostate Cancer. Adv Radiat Oncol. 2020;5(6):1213-1224.
