This information is intended for US healthcare professionals to access current scientific information about J&J Innovative Medicine products. It is prepared by Medical Information and is not intended for promotional purposes, nor to provide medical advice.

ERLEADA - Metastasis-Free Survival (MFS)

Last Updated: 07/31/2024

SUMMARY

  • MFS has been shown to be a strong surrogate endpoint for overall survival (OS) in localized prostate cancer as well as castration-resistant prostate cancer (CRPC).1,2
  • In SPARTAN, the phase 3 study that evaluated the efficacy and safety of ERLEADA plus androgen deprivation therapy (ADT) compared to placebo plus ADT in patients with high-risk non-metastatic castration-resistant prostate cancer (nmCRPC; N=1207), a significant improvement in the primary endpoint MFS was observed.3
    • At the final analysis (following 378 events) for MFS, significant improvement in median MFS was observed: 40.5 months in the ERLEADA group vs 16.2 months in the placebo group (HR, 0.28; 95% CI, 0.23-0.35; P<0.001). At the first interim analysis for overall survival (OS; secondary endpoint), ERLEADA was associated with longer median OS compared to placebo (HR, 0.70; 95% CI, 0.471.04; P=0.07), however this data was considered immature (104 events; 24% of required number of events).3,4
    • On the basis of the efficacy and safety data from the primary analysis, the independent data and safety monitoring committee unanimously recommended unblinding the study and offering patients assigned to the placebo group the option to receive ERLEADA.3
    • At the final analysis (following 428 events) for OS, after a median follow-up of 52 months, a statistically significant improvement in median OS was observed: 73.9 months in the ERLEADA group vs 59.9 months in the placebo group (HR, 0.78; 95% CI, 0.64-0.96, P=0.016).5
    • Adverse events (AEs) that occurred in ≥15% of patients in the ERLEADA group included: fatigue, hypertension, diarrhea, fall, arthralgia, nausea, weight decreased, back pain, and hot flush.5
  • Additional analyses, including the second interim analysis for OS, a subgroup analysis, post-hoc analyses, and multivariate analyses have been conducted.6-17
  • PROTEUS (NCT03767244) is an ongoing phase 3, randomized, double-blind, placebo controlled, multicenter study evaluating the efficacy and safety of ERLEADA plus ADT compared to placebo plus ADT before and after radical prostatectomy (RP) in patients with localized or locally advanced high-risk prostate cancer. The dual primary endpoints are pathologic complete response (pCR) rate and MFS. The study is expected to enroll approximately 2000 patients from >203 sites in 18 countries. Safety and efficacy results from the PROTEUS study have not been published.18,19

BACKGROUND

Surrogate Endpoints in Prostate Cancer Clinical Trials

Due to prolonged survival, surrogate endpoints for OS are needed in CRPC to assess clinical efficacy within a feasible time frame.2,20 A number of endpoints have been proposed for clinical trials and are being validated in clinical studies.21,22 Proposed surrogate endpoints for OS include MFS, biochemical failure, prostate-specific antigen (PSA) doubling time (PSADT), PSA nadir, end of treatment PSA, disease-free survival, and radiographic progression-free survival (PFS).1,20,22

MFS assesses the time interval from PSA recurrence (or start of trial/randomization) to first radiographic metastasis or death.1,2,23 MFS has been shown to correlate with OS in men with localized and biochemically recurrent prostate cancer in several studies, including a recent study by the International Clinical Endpoints for Cancer of the Prostate (ICECaP) Working Group.1,2,22 The aim of the ICECaP Working Group study (to accelerate adjuvant prostate cancer study readouts) resulted in rigorous methodology that evaluates potential intermediate clinical endpoints for OS surrogacy, using individual patient data from international randomized controlled trials.22

In this systematic review of 19 randomized controlled trials evaluating 21 study units in 12,712 patients with localized prostate cancer, the correlation between OS and MFS was 0.91 (95% CI, 0.91-0.91) at the patient level, as measured by Kendall’s τ. At the trial level, R2 was 0.83 (95% CI, 0.71-0.88) from weighted linear regression of 8-year OS rates vs 5year MFS rates. The treatment effect (measured by log HR) for MFS and OS was well correlated (R2, 0.92 [95% CI, 0.81-0.95]).1 Similarly, in an analysis of patients with nmCRPC at a single cancer center, MFS was found to be strongly correlated with OS in a real world population.24

In a retrospective study using the Optum electronic health record database in patients with nmCRPC, MFS independently predicted mortality risk. Patients developing metastasis within 1 year had a 4.4-fold greater risk for mortality (95% CI, 2.2-8.8) than those who remained metastasis-free at year 3.25

Delaying metastases, or extending MFS, may also delay symptomatic progression, morbidity, mortality, costs, and healthcare resource utilization.26,27

MFS - Regulatory History and Selection as Primary Endpoint in the SPARTAN Study

MFS was the primary endpoint of the pivotal study, SPARTAN, used to support regulatory agency approval of ERLEADA.3,28

The appropriate primary efficacy endpoint to use in trials for study patients with nmCRPC was addressed at two meetings of the Food and Drug Administration’s (FDA) Oncology Drugs Advisory Committee. The first meeting was non-product specific and discussed general issues related to the development of drugs for the treatment of nmCRPC, while the second meeting was focused on the review of a specific drug for potential approval in the nmCRPC setting. There was recognition that the transition to a state where the patient developed measurable metastatic disease represented a significant event for patients with nmCRPC. A consensus emerged that an MFS endpoint would likely be clinically meaningful if it was of sufficient magnitude and was accompanied by data from supportive secondary endpoints such as OS. Approval of ERLEADA based on MFS, the primary efficacy endpoint of SPARTAN, established a new regulatory precedent.28

Additionally, in November 2018, the FDA issued a draft guidance which provides recommendations to sponsors regarding the use of MFS as an endpoint in clinical trials for nmCRPC development programs for drug or biological products regulated by the Center for Drug Evaluation and Research (CDER) and the Center for Biologics Evaluation and Research (CBER). The FDA has recognized that a prolonged delay in development of metastatic disease is an objective and clinically relevant measure. Furthermore, FDA has stated that future agents may be approved on the basis of MFS only if substantial effects on MFS are demonstrated and the safety profile is acceptable for a medication taken long-term.29,30 This endpoint also has broad support within the oncologic community as an intermediate endpoint that is well correlated with OS.31

Results from the SPARTAN study have been summarized in the CLINICAL DATA section.

CLINICAL DATA

SPARTAN Study

The SPARTAN study evaluated the efficacy and safety of ERLEADA compared to placebo in patients with high-risk (defined as PSADT ≤10 months) nmCRPC (N=1207).3

Study Design/Methods

SPARTAN (NCT01946204) was a phase 3, randomized, double-blind, placebo-controlled, multicenter study (Figure: SPARTAN Study Design). A total of 332 sites in 26 countries in North America, Europe, and Asia-Pacific were included.

SPARTAN Study Design3,32

Abbreviations: ADT, androgen deprivation therapy; CT, computed tomography; GnRH, gonadotropin-releasing hormone; MFS, metastasis-free survival; mCRPC; metastatic castration-resistant prostate cancer; nmCRPC, non-metastatic castration-resistant prostate cancer; PSADT, prostate-specific antigen doubling time.
aContinuous ADT with a GnRH analog or surgical castration was required for patients in both groups in order to maintain castrate levels of testosterone (<50 ng/dL); choice of medical castration was at the discretion of the investigator.32
bAfter the first detection of distant metastasis, patients were eligible to receive sponsor-provided abiraterone acetate plus prednisone as a treatment option. All post study treatment for mCRPC was at the treating physician’s discretion.3

  • Disease assessments using technetium-99m bone scans and computed tomography (CT) scans were performed every 16 weeks for the pelvis, abdomen, and chest and additionally if distant metastases were suspected.3
  • Radiographic evidence of distant metastasis was determined based on Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 and all imaging was assessed by blinded independent central review (BICR). When new bone lesions were detected by bone scan, a second imaging modality (CT or magnetic resonance imaging) was required to confirm metastasis.
  • Select exclusion criteria: presence of detectable distant metastases; prior treatment with second-generation antiandrogens, cytochrome P450 (CYP) 17 inhibitors, radiopharmaceutical agents, immunotherapy, or any other investigational agent(s) for nmCRPC;32 prior chemotherapy for prostate cancer, except if administered in the adjuvant/neoadjuvant setting;32 history of seizure or condition that may predispose to seizure32
  • Primary endpoint: MFS, defined as the time from randomization to the time of first evidence of BICR-confirmed distant metastasis, defined as new bone or soft tissue lesions or enlarged lymph nodes above the iliac bifurcation, or death due to any cause, whichever occurred first32
  • Select secondary endpoints:
    • Time to metastasis, defined as time from randomization to first detection of distant metastasis involving bone or soft tissue on imaging, as assessed by BICR
    • PFS, defined as time from randomization to first detection of local or distant metastatic disease on imaging, assessed by BICR, or death from any cause, whichever occurred first
    • Time to symptomatic progression, defined as time from randomization to skeletalrelated event, pain progression, or worsening of disease-related symptoms leading to initiation of a new systemic anticancer therapy, or the time to development of clinically significant symptoms due to local or regional tumor progression requiring surgery or radiation therapy
    • OS
    • Safety and tolerability32
  • Other exploratory endpoints were additionally assessed.
  • After the first detection of distant metastasis, patients were eligible to receive sponsorprovided abiraterone acetate plus prednisone as a treatment option. All poststudy treatment for mCRPC was at the treating physician’s discretion.

Results

Patient Characteristics
  • Patient demographics and disease characteristics were well balanced between the 2 groups, and there were no significant differences.
  • Six patients (3 per group) were randomized, but never received study treatment.
  • At the clinical cutoff date for the primary analysis, driven by metastasis events or death, the median follow-up was 20.3 months, with 60.9% of patients still on treatment in the ERLEADA group vs 29.9% in the placebo group.
  • On the basis of the efficacy and safety data from the primary analysis, the independent data and safety monitoring committee unanimously recommended unblinding the study and offering patients assigned to the placebo group the option to receive ERLEADA.
    • After unblinding of the study, 76 patients (19%) in the placebo group received therapy with ERLEADA plus ADT (crossover group).5
  • At the clinical cutoff date for the final OS analysis, 30% (237/803) and 61% (46/76) of the patients were still on treatment in the ERLEADA and crossover groups, respectively.5
Efficacy
  • At the final analysis for MFS performed after 378 events of distant metastasis or death (184 [22.8%] patients in the ERLEADA group and 194 [48.4%] patients in the placebo group), significant improvement in median MFS was observed: 40.5 months vs 16.2 months in the ERLEADA group vs the placebo group, respectively (HR, 0.28; 95% CI, 0.23-0.35; P<0.001).
  • The treatment effect of ERLEADA on MFS was consistently favorable across prespecified subgroups, including patients with PSADT ≤6 months vs >6 months, use of bonesparing agents, and local-regional disease.
  • At the primary analysis, ERLEADA was associated with improvements in all secondary endpoints, with significant improvement observed in time to metastasis, PFS, time to symptomatic progression (all P<0.001).3 At the final analysis, ERLEADA was associated with significant improvement in the secondary endpoint of OS (P=0.016).5
    • At the final analysis (following 428 events) for OS, after a median follow-up of 52 months, a statistically significant improvement in median OS was observed: 73.9 months in the ERLEADA group vs 59.9 months in the placebo group (HR, 0.78; 95% CI, 0.64-0.96, P=0.016).5
  • A total of 48% of ERLEADA-treated patients and 71% of placebo-treated patients received first subsequent systemic therapy for prostate cancer, with abiraterone acetate plus prednisone as the most common first subsequent therapy (73% in the ERLEADA group and 72% in the placebo group). Of the 401 patients randomized to the placebo group, 338 (84%) patients received either life-prolonging active therapy as the first subsequent therapy upon disease progression or ERLEADA as a crossover treatment option without progression after study unblinding.5
Safety
  • The safety population included all patients who received at least 1 dose of study drug.
  • At the final analysis for OS, the median treatment duration was 32.9 months in the ERLEADA group, 11.5 months in the placebo group, and 26.1 months in the crossover group.5
  • AEs that occurred at ≥15% in the ERLEADA group (all grades) included: fatigue (30.4%), hypertension (24.8%), rash (23.8%), diarrhea (20.3%), nausea (18.1%), weight loss (16.1%), arthralgia (15.9%), and falls (15.6%). Other AEs of interest included fracture (11.7%), dizziness (9.3%), hypothyroidism (8.1%), mental impairment disorder (5.1%), and seizure (0.2%).3
    • In the final OS analysis, AEs that occurred in ≥15% of patients in the ERLEADA group (all grades) included: fatigue (33%), hypertension (28%), diarrhea (23%), fall (22%), arthralgia (20%), nausea (20%), weight decreased (20%), back pain (18%), and hot flush (15%).5
  • Treatment discontinuation due to progressive disease was reported in 155 (19.3%) patients in the ERLEADA group and in 210 (52.8%) patients in the placebo group.3
    • In the final OS analysis, treatment discontinuation due to progressive disease was reported in 343 (43%) patients in the ERLEADA group, 238 (74%) patients in the placebo group, and 11 (14%) patients in the crossover group.33
  • Treatment discontinuation due to AEs was reported in 85 (10.6%) and 28 (7.0%) patients who received ERLEADA or placebo, respectively.3 Rash was the most common AE leading to treatment discontinuation (2.4% vs 0%), dose reduction (2.7% vs 0.3%), and dose interruption (6.8% vs 1.3%) in the ERLEADA group vs the placebo group, respectively.34
    • In the final OS analysis, treatment discontinuation due to AEs was reported in 122 (15%), 27 (8.4%), and 8 (11%) patients in the ERLEADA, placebo, and crossover groups, respectively.33
  • AEs were associated with death in 10 patients in the ERLEADA group: prostate cancer (n=2), sepsis (n=2), acute myocardial infarction (n=1), cardiorespiratory arrest (n=1), cerebral hemorrhage (n=1), myocardial infarction (n=1), multiple organ dysfunction (n=1), and pneumonia (n=1).3
Landmark and Correlation Analysis of MFS and OS
  • A landmark and correlation analysis was conducted to determine the relationship between MFS and OS in patients with nmCRPC included in the SPARTAN study.35
  • MFS was analyzed at 6, 9, and 12 months after treatment initiation.
    • Patients were analyzed according to their metastatic status at each time point.
    • The Kaplan-Meier method was used to estimate OS by metastatic status at each landmark time point.
  • A multivariate proportional hazards model evaluated the significance of metastasis to predict OS at 6, 9, and 12 months.
  • A parametric statistical model developed by Fleischer and colleagues was implemented to account for dependence of MFS on OS.
    • The model explains the correlation between endpoints in terms of the proportion of variability of OS attributable to the variability of MFS, and accounts for censoring.
  • Results of the landmark analysis showed that patients who developed metastases at 6, 9, and 12 months had a significantly shorter median OS compared with those patients without metastasis. In the ERLEADA group, for patients who developed metastases by 6, 9, and 12 months compared with patients who did, the adjusted for prognostic factors HR were 6.55 (95%, 2.94-14.49), 7.63 (95% CI, 4.12-14.08), and 9.17 (95% CI, 5.0816.67), respectively.
    • Overall, 5, 14, and 26 patients died before 6, 9, and 12 months, respectively, and were excluded from the analyses.
  • Results from the parametric Fleischer’s statistical model confirmed the positive association between MFS and OS, with an estimated correlation coefficient of 0.69 (bootstrap standard error, 0.002; 95% CI, 0.69-0.70).
Additional Information

Additional information regarding the SPARTAN study, including the clinical study report, protocol, and statistical analysis plan, can be found at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/Erleada_210951_toc.cfm (scroll to the “Sponsor Clinical Study Reports ARN-509-003 SPARTAN NCT # 01946204” section at the bottom of the web page).

Literature Search

A literature search of MEDLINE®, Embase®, BIOSIS Previews®, and Derwent Drug File (and/or other resources, including internal/external databases) was conducted on 31 July 2024. Summarized in this response are relevant data from the phase 3 SPARTAN study and a landmark and correlation analysis.

 

References

1 Xie W, Regan M, Buyse M, et al. Metastasis-free survival is a strong surrogate of overall survival in localized prostate cancer. J Clin Oncol. 2017;35(27):3097-3104.  
2 Schweizer MT, Zhou XC, Wang H, et al. Metastasis-free survival is associated with overall survival in men with PSA-recurrent prostate cancer treated with deferred androgen deprivation therapy. Ann Oncol. 2013;24(11):2881-2886.  
3 Smith MR, Saad F, Chowdhury S, et al. Apalutamide treatment and metastasis-free survival in prostate cancer. N Engl J Med. 2018;378(15):1408-1418.  
4 Small E, Saad F, Chowdhury S, et al. SPARTAN, a phase 3 double-blind, randomized study of apalutamide vs placebo in patients with nonmetastatic castration-resistant prostate cancer. Oral presentation presented at: the American Society of Clinical Oncology Genitourinary (ASCO-GU) Cancers Symposium; February 8-10, 2018; San Francisco, CA.  
5 Smith MR, Saad F, Chowdhury S, et al. Apalutamide and overall survival in prostate cancer. Eur Urol. 2021;79(1):150-158.  
6 Smith M, Saad F, Rathkopf D, et al. Relationship of time to metastasis and site of metastases in patients with nonmetastatic castration-resistant prostate cancer: results from the phase 3 SPARTAN trial. Poster presented at: The American Society of Clinical Oncology Annual Meeting; June 1-5, 2018; Chicago, IL.  
7 Small E, Saad F, Rathkopf D, et al. Predicting disease progression in patients with nonmetastatic castration-resistant prostate cancer: an analysis from the phase 3 SPARTAN trial. Poster presented at: The American Society of Clinical Oncology Annual Meeting; June 1-5, 2018; Chicago, IL.  
8 Small EJ, Saad F, Chowdhury S, et al. Apalutamide and overall survival in non-metastatic castration-resistant prostate cancer. Ann Oncol. 2019;30(11):1813-1820.  
9 Graff J, Saad F, Hadaschik B, et al. Metastasis-free survival in nonmetastatic castration-resistant prostate cancer patients with prostate-specific antigen decline to < 0.2 ng/mL following apalutamide treatment: post hoc results from the phase 3 SPARTAN study. Poster presented at: American Urological Association (AUA) Annual Meeting; May 3-6, 2019; Chicago, IL.  
10 Hadaschik B, F Saad, JN Graff, et al. Efficacy and safety of apalutamide plus ongoing androgen deprivation therapy in nonmetastatic castration-resistant prostate cancer patients with or without prior radical prostatectomy and/or external radiotherapy: post hoc analysis of SPARTAN. presented at: Poster presented at: American Urological Association Annual Meeting; May 3-6, 2019; Chicago, IL.  
11 Graff J, Smith MR, Saad F, et al. Age-related efficacy and safety of apalutamide (APA) plus ongoing androgen deprivation therapy (ADT) in subgroups of patients (pts) with nonmetastatic castration-resistant prostate cancer (nmCRPC): post hoc analysis of SPARTAN. Poster presented at: American Society of Clinical Oncology (ASCO) Annual Meeting; May 31-June 4, 2019; Chicago, IL.  
12 Small E, Saad F, Chowdhury S, et al. Efficacy of apalutamide plus ongoing androgen deprivation therapy in patients with nonmetastatic castration-resistant prostate cancer and baseline comorbidities. Poster presented at: American Society of Clinical Oncology (ASCO) Annual Meeting; May 31 - June 4, 2019; Chicago, IL.  
13 Chi K, Saad F, Chowdhury S, et al. Prostate-specific antigen kinetics in patients with advanced prostate cancer treated with apalutamide: results from the TITAN and SPARTAN studies. Poster presented at: American Society of Clinical Oncology (ASCO) Scientific Program; May 29-31, 2020; Virtual.  
14 Feng F, Thomas S, Gormley M, et al. Identifying molecular determinants of response to apalutamide in patients with nonmetastatic castration-resistant prostate cancer in the SPARTAN study. Poster presented at: 2019 Genitourinary Cancers Symposium; February 14-16, 2019; San Francisco, CA.  
15 Aguilar C, Gormley M, Thomas S, et al. Novel molecular subtypes identified in prostate cancer: results from the SPARTAN study. Poster presented at: American Association for Cancer Research (AACR) 2020 Virtual Meeting II; June 22-24, 2020.  
16 Uemura H, Satoh T, Tsumura H, et al. Efficacy and safety of apalutamide in Japanese patients with nonmetastatic castration-resistant prostate cancer: a subgroup analysis of a randomized, double-blind, placebo-controlled, phase-3 study. Prostate Int. 2020;8(4):190-197.  
17 Feng FY, Thomas S, Saad F, et al. Association of molecular subtypes with differential outcome to apalutamide treatment in nonmetastatic castration-resistant prostate cancer. Jama Oncol. 2021;7(7):1005-1014.  
18 Kibel AS, Gleave M, Brookman-May SD, et al. PROTEUS: a randomized, double-blind, placebo-controlled, phase 3 trial of apalutamide plus androgen deprivation therapy (ADT) vs placebo plus ADT prior to and after radical prostatectomy in patients with localized or locally advanced high-risk prostate cancer. Poster presented at: American Society of Clinical Oncology (ASCO) Genitourinary Cancers Symposium; February 17-19, 2022; San Francisco, CA.  
19 Janssen Research & Development, LLC. A randomized, double-blind, placebo-controlled, phase 3 study of apalutamide in subjects with high-risk, localized or locally advanced prostate cancer who are candidates for radical prostatectomy. In: ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2000- [cited 2024 May 21]. Available from: https://clinicaltrials.gov/ct2/show/NCT03767244 NLM Identifier: NCT03767244.  
20 Morris M, Molina A, Small E, et al. Radiographic progression-free survival as a response biomarker in metastatic castration-resistant prostate cancer: COU-AA-302 results. Medicina. 2015;33(12):1356-1363.  
21 Scher HI, Halabi S, Tannock I, et al. Design and end points of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the Prostate Cancer Clinical Trials Working Group. J Clin Oncol. 2008;26(7):1148-1159.  
22 ICECaP Working Group, Sweeney C, Nakabayashi M, et al. The development of intermediate clinical endpoints in cancer of the prostate (ICECaP). Jnci J National Cancer Inst. 2015;107(12):djv261.  
23 Antonarakis E, Chen Y, Elsamanoudi S, et al. Long-term overall survival and metastasis-free survival for men with prostate-specific antigen-recurrent prostate cancer after prostatectomy: analysis of the Center for Prostate Disease Research National Database. BJU Int. 2011;108(3):378-385.  
24 Gagnon R, Alimohamed NS, Batuyong E, et al. Metastasis-free survival as a predictor of overall survival in non-metastatic castration-resistant prostate cancer. J Clin Oncol. 2019;37(7_suppl):201-201.  
25 Li S, Ding Z, Lin J, et al. Association of prostate-specific antigen (PSA) trajectories with risk for metastasis and mortality in non-metastatic castration-resistant prostate cancer (nmCRPC). Abstract presented at: 2018 Genitorurinary Cancers Symposium; February 8-10, 2018; San Francisco, CA.  
26 Luo J, Beer T, Graff J. Treatment of nonmetastatic castration-resistant prostate cancer. Oncology. 2016;30(4):336-344.  
27 Li TT, Shore ND, Mehra M, et al. Impact of subsequent metastases on costs and medical resource use for prostate cancer patients initially diagnosed with localized disease. Cancer. 2017;123(18):3591-3601.  
28 Center for Drug Evaluation and Research. NDA/BLA Multi-Disciplinary Review and Evaluation (Summary Review, Office Director, Cross Discipline Team Leader Review, Clinical Review, Non-Clinical Review, Statistical Review and Clinical Pharmacology Review) NDA 210951 - ERLEADA (apalutamide) - Reference ID: 4221387. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/210951Orig1s000MultidisciplineR.pdf. Published March 19, 2018. Accessed May 20, 2024.  
29 Center for Drug Evaluation and Research (CDER) & Center for Biologics Evaluation and Research (CBER). Nonmetastatic, castration-resistant prostate cancer: considerations for metastasis-free survival endpoint in clinical trials guidance for industry DRAFT GUIDANCE. 2018-[cited 2024 May 14]. Available from: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/nonmetastatic-castration-resistant-prostate-cancer-considerations-metastasis-free-survival-endpoint.  
30 Beaver JA, Kluetz PG, Pazdur R. Metastasis-free survival - a new end point in prostate cancer trials. New Engl J Med. 2018;378(26):2458-2460.  
31 Parikh RB, Prasad V. Metastasis-free survival in prostate cancer: faster drug approvals, better drugs? J Clin Oncol. 2019;37(4):266-268.  
32 Smith MR, Saad F, Chowdhury S, et al. Protocol for: Apalutamide treatment and metastasis-free survival in prostate cancer. N Engl J Med. 2018;378(15):1408-1418.  
33 Smith MR, Saad F, Chowdhury S, et al. Supplement for: Apalutamide and overall survival in prostate cancer. Eur Urol. 2021;79(1):150-158.  
34 Smith MR, Saad F, Chowdhury S, et al. Supplement for: Apalutamide treatment and metastasis-free survival in prostate cancer. N Engl J Med. 2018;378(15):1408-1418.  
35 Smith MR, Mehra M, Nair S, et al. Relationship between metastasis-free survival and overall survival in patients with nonmetastatic castration-resistant prostate cancer. Clin Genitourin Canc. 2020;18(2):e180-e189.