Abstract
Background:
The phase 3 trial CA184–043 evaluated radiotherapy to bone metastases followed by Ipilimumab or placebo in men with metastatic castrate-resistant prostate cancer (mCRPC) who had received docetaxel previously. In a prior analysis, the trial’s primary endpoint (overall survival [OS]) was not improved significantly.
Objective:
To report the final analysis of OS.
Design, setting, and participants:
A total of 799 patients were randomized to receive a single dose of radiotherapy to one or more bone metastases followed by either Ipilimumab (n = 399) or placebo (n = 400).
Outcome measurements and statistical analysis:
OS was analyzed in the intention-to-treat population. Prespecified and exploratory subset analyses based on Kaplan-Meier/Cox methodology were performed.
Results and limitations:
During an additional follow-up of approximately 2.4 yr since the primary analysis, 721/799 patients have died. Survival analysis showed crossing of the curves at 7–8 mo, followed by persistent separation of the curves beyond that point, favoring the ipilimumab arm. Given the lack of proportional hazards, a piecewise hazard model showed that the hazard ratio (HR) changed over time: the HR was 1.49 (95% confidence interval 1.12, 1.99) for 0–5 mo, 0.66 (0.51, 0.86) for 5–12 mo, and 0.66 (0.52, 0.84) beyond 12 mo. OS rates were higher in the ipilimumab versus placebo arms at 2 yr (25.2% vs16.6%), 3 yr (15.3% vs7.9%), 4 yr (10.1% vs3.3%), and 5 yr (7.9% vs. 2.7%). Disease progression was the most frequent cause of death in both arms. In seven patients (1.8%) in the ipilimumab arm and one (0.3%) in the placebo arm, the primary cause of death was reported as study drug toxicity. No long-term safety signals were identified.
Conclusions:
In this preplanned long-term analysis, OS favored ipilimumab plus radiotherapy versus placebo plus radiotherapy for patients with postdocetaxel mCRPC. OS rates at 3, 4, and 5 yr were approximately two to three times higher in the ipilimumab arm.
Patient summary:
After longer follow-up, survival favored the group of men who received ipilimumab, with overall survival rates being two to three times higher at 3 yr and beyond.
Keywords: Prostate cancer, Ipilimumab, Immunotherapy, Radiotherapy
1. Introduction
Management of men with metastatic castration-resistant prostate cancer (mCRPC) has evolved dramatically since 2010, with taxanes (docetaxel and cabazitaxel), next-generation androgen receptor axis targeting agents (abiraterone and enzalutamide), and a bone-targeted radioisotope (radium-223) demonstrating improved overall survival (OS) in clinical trials [1–3]. In many othersolid tumors, immunotherapy based on immune checkpoint blockade has emerged as an important treatment option [4,5]. Prostate cancer was one of the first malignancies in which cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibition was tested in preclinical models [6,7] and activity was subsequently confirmed in phase I-II development [8–10]. Recent data [11,12] also suggest that anti–PD-1 has activity in some patients with mCRPC, and a randomized phase III trial adding PD-L1 blockade to second-generationandrogen receptor inhibitor treatment is currently underway (NCT03016312). Further, the autologous cell-based “vaccine" sipuleucel-T is approved by the US Food and Drug Administration for the treatment of mCRPC based on a survival benefit [13], although this treatment is not available outside of the USA.
Ipilimumab is a fully human monoclonal antibody that blocks the inhibitory signal mediated by CTLA-4, and as a consequence, enhances antitumor immunity and has immune-related side effects [14–16]. Two randomized phase III trials testing ipilimumab in men with mCRPC were designed and conducted, one in men who had not received chemotherapy previously (the “Ipi 095” trial, NCT01057810) and the other in men who had progressed after docetaxel (the “Ipi 043” trial, NCT00861614, where ipilimumab was used following radiotherapy (RT) directed to a bone lesion [17]). In both trials, progression-free survival (PFS; hazard ratio [HR]: 0.67; 95.87% confidence interval [CI], 0.55 to 0.81, and HR: 0.70, 95% CI 0.61–0.82, respectively), but not OS, was significantly improved [15,16]. In the 043 trial, which enrolled patients with more advanced disease, OS was improved nonsignificantly (HR: 0.85, 95% CI 0.72–1.00; p = 0.053), with Kaplan-Meier curves initially disfavoring the ipilimumab group and then crossing after approximately 6 mo [15]. In order to elucidate whether ipilimumab treatment is associated with improved long-term OS in men with prostate cancer, we continued to follow patients enrolled in the 043 trial; this manuscript reports mature results with an additional 2 yr of follow-up since the primary analysis.
2. Patients and methods
2.1. Patients
The randomized, double-blind, controlled phase 3 trial “CA184–043” was reported previously [15]. In brief, 043 was conducted in 191 centers from 26 countries; it enrolled male patients aged 18 yr or older with histologically or cytologically confirmed adenocarcinoma of the prostate, at least one bone metastases that could be irradiated or that warranted irradiation in the clinical judgment of the investigator, testosterone <0.50 ng/mL, an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, and progression ≤6m o after receiving docetaxel. Exclusion criteria were more than two cytotoxic chemotherapy regimens for mCRPC, brain metastases, autoimmune disease, or known HIV, hepatitis B or C infection.
Review boards at all participating institutions approved the study, which was conducted according to the provisions of the Declaration of Helsinki and Good Clinical Practice Guidelines of the International Conference on Harmonization. All patients provided written informed consent to participate in the study.
2.2. Randomization and masking
Patients (n = 799) were randomized in a 1:1 ratio to receive bone-directed RT followed by either ipilimumab intravenously at 10 mg/kg or placebo every 3 wk for up to four doses. Nonprogressing patients could receive ipilimumab at 10 mg/kg or placebo as maintenance therapy every 3 mo. Randomization was performed with an interactive voice response system, stratified by ECOG performance status (0 vs 1), alkaline phosphatase (<1.5 × upper limit of normal [ULN] vs ≥1.5 × ULN), hemoglobin (<11 vs ≥11 g/dl), and investigator site. An independent data monitoring committee had access to unblinded data.
2.3. Procedures
Patients first received a single dose of RT at 8Gy to at least one and up to five bone fields based on investigator discretion. RT was delivered within 2 d prior to the initiation of either ipilimumab or placebo. Treatment was continued until confirmed disease progression, intolerable toxicity, clinical deterioration, death, patient withdrawal of consent, or loss to follow-up.
Tumor assessments by radiographic imaging and prostate-specific antigen (PSA) levels were performed every 12 and 6 wk, respectively. Safety characterization was based on events tabulated from the first dose of study drug to 70 d after the last dose of study drug (including maintenance therapy). This interval is considered the “on-study” period. We also evaluated the incidence of immune-related adverse events (irAEs), which were determined from a predefined list of MedDRA terms representing adverse events potentially associated with inflammation and considered to be causally related to drug exposure.
2.4. Statisticalanalysis
The purpose of the analysis was to quantify preplanned updated OS and safety results through the July 13, 2015 fin.l database lock for the extension phase of the trial.
The primary end point of the trial was OS, defined as time from randomization to death from any cause. For individuals who did not die, OS was censored at the datelast known to be alive.
Yearly survival rates up to 5 yr were estimated for each treatment group using the Kaplan-Meier product limit method [18]. Two-sided 95% CIs for median survival were computed by treatment group usingthe Brookmeyer and Crowley [19] method. Descriptive statistics for safetywere tabulated for all treated patients using National Cancer Institute Common Terminology Criteria for Adverse Events version 3.0 by treatment group. Safety analyses were performed using data obtained from the start of blinded study drug dosing up to 70 d after the last dose of study therapy. Demography, baseline characteristics, and safety were summarized descriptively.
3. Results
A total of 988 patients were enrolled and screened for study participation (Supplementary Fig. 1). Of these patients, 799 (399 in the ipilimumab arm and 400 in the placebo arm) were randomly assigned from May 2009 to February 2012 to blinded study treatment at a total of 153 sites [15] (Table 1). Approximately one-half (49%) of randomized patients were from sites in Europe, 27% were from sites in North America, 20% were from sites in South America, and 3.1% were from sites in Australia.
Table 1 –
Demographic and baseline characteristics.
| Ipilimumab (N = 399) | Placebo (N = 400) | |
|---|---|---|
| Age (yr), | ||
| Median (Q1–Q3) | 69.0 (63.0–74.0) | 67.5 (62.0–72.5) |
| ≥70, no. of patients (%) | 184 (46) | 166 (42) |
| Alkaline phosphatase, no. of patients (%) | ||
| ≥1.5 × ULN | 163 (41) | 151 (38) |
| Not reported | 11 (2.8) | 17 (4.3) |
| Gleason score, no. of patients (%) | ||
| >7 | 192 (48) | 187 (47) |
| Not reported | 33 (8.3) | 23 (5.8) |
| Hemoglobin (g/dl), no. of patients (%) | ||
| <11 | 116 (29) | 111 (28) |
| Not reported | 16 (4.0) | 20 (5.0) |
| ECOG PS, no. of patients (%) | ||
| 0 | 168 (42) | 170 (43) |
| 1 | 216 (54) | 220 (55) |
| 2 | 3 (0.8) | 0 |
| Not reported | 12 (3.0) | 10 (2.5) |
| Bone metastases, no. of patients (%) | ||
| >5 | 103 (26) | 111 (28) |
| Not reported | 20 (5.0) | 36 (9.0) |
| Visceral metastases, no. of patients (%) | ||
| Yes | 113 (28) | 114 (29) |
| Not reported | 6(1.5) | 11 (2.8) |
| Lactate dehydrogenase, no. of patients (%) | ||
| >2 × ULN | 58 (15) | 53 (13) |
| Not reported | 15 (3.8) | 22 (5.5) |
| Prostate-specific antigen (ng/mL) | ||
| No. of patients (%) | 338 (85) | 334 (84) |
| Median | 138.5 | 176.5 |
| (Q1–Q3) | (44.5–452.8) | (46.6–416.0) |
| Average daily worst pain intensity, no. of patients (%) | ||
| ≥4 | 197 (49) | 186 (47) |
| Not reported | 50 (13) | 64 (16) |
ECOG PS = Eastern Cooperative Oncology Group performance status; ULN = upper limit of normal.
Overall, 387 and 392 patients received at least one dose of ipilimumab and placebo, respectively, and 198 (51%) and 267 (67%) received at least four treatment doses. After induction treatment, 95 (25%) and 62 (16%) patients entered the maintenance phase.
3.1. Overall survival
During an additional follow-up of approximately 2.4 yr since the primary analysis (final database lock: July13, 2015; primary analysis database lock: February 6, 2013), 721/799 patients had died: 49/399 remained alive in the ipilimumab arm and 29/400 were alive in the placebo arm, with a median follow-up of 50 mo (40.7, 72.0) for the survivors. Kaplan-Meier analysis of OS showed crossing of the curves at 7–8 mo, with persistent separation favoring the ipilimumab arm beyond that point with longer follow-up (Fig. 1). Given the lack of proportional hazards, a piecewise hazard model showing a changing HR over time has been reported previously [15], and an update with the final database lock shows that the HR was 1.49 (95% CI 1.12–1.99) for 0–5 mo, 0.66 (0.51–0.86) for 5–12 mo, and 0.66 (0.52–0.84) beyond 12 mo.
Fig. 1 –
Overall survival. CI = confidence interval; HR = hazard ratio; OS = overall survival.
Landmark OS rates for the ipilimumab and placebo groups are summarized in Table 2, which show that, in the ipilimumab arm, OS rates were significantly higher at 2 yr and beyond, and two to three times higher at 3, 4, and 5 yr.
Table 2 –
Overall survival.
| Ipilimumab + radiotherapy | Placebo + radiotherapy | Difference (95% CIa) | |
|---|---|---|---|
| 1-yr OS rate | 47% | 41% | 5.7% (−1.2, 13) |
| 2-yr OS rate | 25% | 17% | 8.6% (3.0, 14) |
| 3-yr OS rate | 15% | 7.9% | 7.4% (3.0, 12) |
| 4-yr OS rate | 10% | 3.3% | 6.8% (3.4, 10) |
| 5-yr OS rate | 7.9% | 2.7% | 5.2% (2.1, 8.3) |
CI = confidence interval; OS = overall survival.
95% CIs were computed for binary rate differences.
Disease progression was the most frequent cause of death in both groups (270 patients, 69% and 298 patients, 75%, respectively). At the time of the final database lock, for seven men (1.8%) in the ipilimumab group and one man (0.25%) in the placebo group, the primary cause of death was reported as study drug toxicity. The seven deaths in the ipilimumab group were due to peritonitis, acute myeloid leukemia, diverticular perforation, pulmonary embolism, pneumonia, decreased performance status, and related cholangitis with a fatal outcome. Of note, only one of these deaths (acute myeloid leukemia) was reported after primary database lock. In the placebo group, one on-study death (0.25%) was reported due to multiorgan failure associated with study drug toxicity.
3.2. Safety
The safety profile for ipilimumab was similar to that reported previously, with irAEs most commonly occurring in the gastrointestinal tract and skin, and to a lesser extent in the liver and endocrine organs (Table 3). The majority of irAEs, including severe irAEs, occurred during the initial four doses of study drug. After year 1, irAEs were reported in nine patients in the ipilimumab arm.
Table 3 –
Adverse events.
| On-study adverse events | Ipilimumab (N = 393) |
Placebo (N = 396) |
||||
|---|---|---|---|---|---|---|
| Any grade | Grade 3 or 4 | Grade 5 | Any grade | Grade 3 or 4 | Grade 5 | |
| Any event | 385 (98) | 231 (59) | 67 (17) | 365 (92) | 162 (41) | 45(11) |
| Immune-related events | 250 (64) | 102 (26) | 1 (0.3) | 86 (22) | 11 (2.8) | 1 (0.3) |
| Gastrointestinal | 160 (41) | 72 (18) | 1 (0.3) | 56 (14) | 3 (0.8) | 0 |
| Diarrhea | 154 (39) | 59 (15) | 0 | 55 (14) | 3 (0.8) | 0 |
| Colitis | 28 (7.1) | 18 (4.6) | 0 | 3 (0.8) | 0 | 0 |
| Diverticular perforation | 1 (0.3) | 0 | 1 (0.3) | 0 | 0 | 0 |
| Skin | 139 (35) | 5 (1.3) | 0 | 29 (7.3) | 0 | 0 |
| Pruritus | 80 (20) | 1 (0.3) | 0 | 15 (3.8) | 0 | 0 |
| Rash | 70 (18) | 3 (0.8) | 0 | 16 (4.0) | 0 | 0 |
| Hepatic | 34 (8.7) | 16 (4.1) | 0 | 18 (4.5) | 7 (1.8) | 0 |
| Increased aspartate aminotransferase | 22 (5.6) | 9 (2.3) | 0 | 13 (3.3) | 4 (1.0) | 0 |
| Alanine aminotransferase | 20 (5.1) | 6(1.5) | 0 | 8 (2.0) | 3 (0.8) | 0 |
| Endocrine | 19 (4.8) | 7 (1.8) | 0 | 6 (1.5) | 2 (0.5) | 0 |
| Hypothyroidism | 9 (2.3) | 2 (0.5) | 0 | 1 (0.3) | 0 | 0 |
| Neurological | 9 (2.3) | 2 (0.5) | 0 | 1 (0.3) | 0 | 0 |
Data are n (%). Patients may have had more than one event.
4. Discussion
Long-term analyses of survival are rarely reported in mCRPC studies, although this is of even greater importance for immunotherapy trials, because a delayed effect on the immune system is expected. This global study was the first phase 3 trial testing ipilimumab in men with mCRPC that included long-term follow-up, and the primary endpoint of OS was not improved at the initial analysis [15]; this intent-to-treat analysis with longer follow-up shows that beyond the crossing of the curves at 7–8 mo, there is persistent separation of the curves favoring the ipilimumab plus RT arm. This favorable impact of ipilimumab plus RT on OS was associated with an increased number of patients alive at 2 yr and beyond, some of whom had complete responses to treatment [20,21]. The primary analysis of this trial showed improved PFS over placebo (HR: 0.70, 95% CI 0.61–0.82; p < 0.0001) and a higher proportion of patients with a confirmed ≥50% PSA decline at any time (13% [95% CI 9.5–18] for ipilimumab and 5.3% [95% CI 3.0–8.4] for placebo) [15]. PFS, but not OS, was also improved significantly (HR: 0.67; 95.87% CI, 0.55–0.81) in a second trial testing ipilimumab monotherapy in men with docetaxel-naïve mCRPC [16]. Of note, the PFS improvement observed across these two CRPC ipilimumab trials is unique among all immunotherapeutic approaches in clinical development for prostate cancer [4,5]. In this long-term analysis, prespecified OS rates were significantly greater at 2, 3, 4, and 5 yr in the ipilimumab versus placebo arms, respectively.
It is likely that the failure of the primary analysis [15] of this trial to originally demonstrate significantly improved OS (HR: 0.85, 0.72–1.00; p = 0.053) with ipilimumab plus RT is due to the apparently initial worse outcome for patients in this arm during the first 7 mo. Crossover of the Kaplan-Meier survival curves was observed, after which time the ipilimumab arm appears to continuously show better OS than the placebo arm. Since this trial was first reported, this phenomenon of crossing curves has been described repetitively in phase 3 trials testing CTLA-4 or PD-1/PD-L1 inhibitors in various cancers, including lung [22] and bladder cancer [23]. Of note, hyperprogression of cancer has been described with these immunotherapies, and they may explain this phenomenon, at least in part [24]. An excess in side effects was observed in the ipilimumab arm during the initial weeks of the trial: this mayalso havefavored directly or indirectly an excess of early death events in this arm. Finally, it is plausible that men with more indolent cancers and those with a lower burden of cancer may also be more sensitive to immunotherapy, which may account forthe delayed effect on survival.
A potentially T-cell–mediated antitumor effect of ipilimumab, as quantified by a PSA decline of >50%, was observed in 13–23% of men enrolled in the two phase 3 trials [15,16]. Although an effect on PSA might not fully explain treatment effects in men with CRPC [25], these data suggest that only a minority of men with mCRPC derive a benefit from the single agent ipilimumab. This finding further emphasizes the need for identifying biomarkers predicting for ipilimumab activity. Clinically applicable biomarkers remain elusive. For example, neoepitope signature and mutational load [26,27], mismatch repair deficiency [28], lactate dehydrogenase or C-reactive protein levels [29], early rise in eosinophils and lymphocytes [30], and, recently, inactivation of CDK12 [31] have been reported to predict the efficacy of immune checkpoint inhibitors. More specifically, in men with mCRPC treated with ipilimumab, the UCSF group recently reported that low pretreatment baseline levels of PD-1(+) CD4 Teff cells correlate with longer survival [32]. They also showed that responses to ipilimumab were also associated with increased T-cell diversity [33]. Although fairly apparent in melanoma, the role of the gut microbiome in predicting checkpoint inhibitor efficacy [34] is unknown in men with prostate cancer. Unfortunately, tissue and stool samples were not collected systematically in the present trial, and therefore aiming at identifying biomarkers for efficacy and toxicity is not possible.
The safety profile of ipilimumab in this study was generally consistent with the experience in other studies using 10 mg/kg ipilimumab, and no new safety signals were identified in this long-term analysis. Common toxicities of ipilimumab (eg, inflammatory events reflecting ipilimumab's immune-based mechanism of action) affected the gastrointestinal tract and skin most frequently. There were seven deaths (1.8%) attributed to study drug toxicity in the ipilimumab group, and the frequency of gastrointestinal perforations was consistent with prior ipilimumab experience in advanced melanoma, where its incidence was approximately 1.0% [35]. Identifying biomarkers predicting for severe toxicity of immunotherapy is also a priority, and ipilimumab was previously shown to induce greater diversification in the T-cell repertoire in patients who develop irAEs than others [33]. Of note, the dose of ipilimumab (10 mg/kg) evaluated in this trial was higher than that approved for the treatment of patients with advanced melanoma (3 mg/kg), and this likely explains the observed degree of toxicity.
In terms of trial design, this study was based, to some degree, on preclinical data showing that the combination of anti-CTLA-4 and RT could induce potent off-target (abscopal) effects [36] and that such off-target effects are immunologically mediated [37]. Since bone-directed RT was administered in both study arms, its role in the efficacy outcomes is unclear. Of note, a second ipilimumab phase 3 trial in men with mCRPC (CA184–095) [16] was performed using ipilimumab monotherapy.
As in all randomized trials, there are limitations in the ascertainment of information about the effectiveness of subsequent treatments on patients in the long term. At the primary analysis cutoff, the use of subsequent cancer therapy was 41% versus 47% in the ipilimumab versus placebo arms [15]. This suggests that treatments used beyond progression in the experimental arm are unlikely to explain the observed long-term OS difference between arms. Subsequent anticancer therapies were not collected for the final database lock (July 13, 2015), so we cannot provide any update. Of note, patients were accrued in the trial from May 2009 to February 2012, a period of time when postdocetaxel life-prolonging therapies were either unavailable or scarcely available in most of the participating countries.
5. Conclusions
In summary, this long-term analysis shows that OS is improved with ipilimumab plus RT versus placebo plus RT in patients with postdocetaxel mCRPC, and treatment was associated with a fraction of patients with long-term survival. As these data were analyzed after the primary OS analysis [15], they must be considered hypothesis generating rather than definitive. As such, ipilimumab is not registered in the mCRPC indication.
Evidence was recently provided that PD-1/PD-L1 blockade is active in a subset of men with CRPC [11,12]. Based on the reported efficacy inothercancers, a combination trial of ipilimumab and nivolumab is undergoing in men with mCRPC (NCT02985957), with preliminary data supporting activity [38], as well as in a set of patients with the ARV7 mutation [39].
Supplementary Material
Acknowledgments:
Contributors—study CA184-043 investigators: Argentina: E. Batagelj (Buenos Aires), L.E. Fein (Rosario), G. Giornelli (Buenos Aires), J.I. Hernandez Moran (Buenos Aires), L.A. Kaen (La Rioja), E. Korbenfeld (Buenos Aires), C. Medina (San Miguel De Tucuman), F.S. Palazzo (San Miguel De Tucuman), N. Pilnik (Cordoba), E. Richardet (Cordoba), M. Rondinon (Buenos Aires), R. Santos (Buenos Aires), M. Tatangelo (Rosario). Australia: I. Davis (Heidelberg, Victoria), V. Ganju (Frankston, Victoria), S. Ng (Subiaco), P. Parente (Box Hill, Victoria). Austria: M. Krainer (Vienna), F. Sedlmayer (Salzburg). Belgium: T. Gil (Brussels), J-P. Machiels (Brussels), D. Schallier (Brussels), F. Van Aelst (Roeselare). Brazil: C.H. Barrios (Porto Alegre), J. Camargo (Curitiba), C. Dzik (Sao Paulo), F.A. Franke (Ljuí), M. Gifoni (Fortaleza), D. Grabarz (Mogi Das Cruzes), F. Melo Cruz (Santo Andre), A. Nogueira Rodrigues (Sao Paulo), A. Notari (Porto Alegre), M. Oliveira (Curitiba). Canada: G. Gagnon (Rimouski, QC), F. Saad (Montreal, QC), C. Sperlich (Greenfield Park, QC). Chile: A. Cordova (Santiago De Chile), P. Gonzalez Mella (Viña Del Mar), H. Harbst (Santiago—Independencia), F. Orlandi (Santiago), E. Yanez (Temuco). Colombia: M. Gonzalez (Cordoba), R. Schlesinger (Bogota), R. Varela (Bogota). Czech Republic: V. Hejzlarova (Liberec), J. Katolicka (Brno), J. Petera (Hradec Kralove). Denmark: L. Bentzen (Aarhus), G. Daugaard (Kobenhavn O), S. Hansen (Odense), M. Holmberg (Aalborg), L. Sengelov (Herlev). France: M. Baciuchka-Palmaro (Marseille Cedex 20), P. Blanchet (Pointe A Pitre), A. Bossi (Villejuif), K.Fizazi (Villejuif), N. Houede (Bordeaux), C. Linassier (Tours), H. Mahammedi (Clermont-Ferrand), A. Thiery-Vuillemin (Besancon Cedex). Germany: J. Gleissner (Wuppertal), P.J. Goebell (Erlangen) F. Koenig (Berlin), F. Lohr (Mannheim), S. Rogenhofer (Bonn). Greece: E. Efstathiou (Athens). Hungary: I. Bodrogi(Budapest), G. Pajkos (Kecskemet), B. Piko (Gyula), Z. Volgyi (Kaposvar). Ireland: J.A. McCaffrey (Dublin), R. McDermott (Dublin). Israel: R. Berger (Tel Hashomer), E. Gez (Tel Aviv), W. Mermershtain (Beer-Sheva), A. Peer (Haifa), A. Sella (Beer Jacob). Italy: P.A. Ascierto (Napoli), A. Bertolini (Sondrio), M. Maio (Siena), R. Ridolfi (Meldola), E. Villa (Milano). Mexico: G. Castelazo Rico (Mexico City), H. Duran (Acapulco), H. Gonzalez (Aguascalientes), P. Nieto (Xalapa), S. Rivera (Mexico City), P. Solano (Guadalajara), J.M. Tello (Cuernavaca). The Netherlands: A.M. Bergman (Amsterdam), W.R. Gerritsen (Nijmegen), A.J.M. van den Eertwegh (Amsterdam). Peru: C. Farfan (Lima), A. Figueroa Torrejon (Lima), J.B. Giraldez (Arequipa), M. Huaringa (Lima), C. Lozada (Lima), S. Neciosup (Lima). Poland: J. Jassem (Gdansk), S. Nawrocki (Olsztyn). Puerto Rico: H. Velez (Ponce). Romania: F. Bacanu (Bucharest), T.E. Ciuleanu (Cluj-Napoca), D.E. Ganea (Suceava). Russian Federation: O. Apolikhin (Moscow), M.Y. Biakhov (Moscow), S. Cheporov (Yaroslavl), S. Ivanov (Moscow), O. Karyakin (Obninsk), V. Matveev (Moscow), A. Nosov (St Petersburg), S. Safina (Kazan). Spain: J. Carles (Barcelona), J.M. Cervera Grau (Benidorm-Alicante), M. Climent (Valencia), E. Gallardo (Barcelona), J. Garcia Donas (Madrid), R. Lopez Lopez (Santiago De Compostela). UK: A. Choudhury (Manchester), S. Dixit (Scunthorpe), P. Leone (Chelmsford), J. Staffurth (Cardiff), S. Sundar (Nottingham). USA: N. Agarwal (Salt Lake City, Utah), S. Agarwala (Bethlehem, PA), M.V.S. Andavolu (Palm Springs, CA), B.B. Bank (Elk Grove Village, IL), A.D. Baron (San Francisco, CA), J. Beck (Fayetteville, AR), T.M. Beer (Portland, OR), W.R. Berry (Raleigh, NC), G. Chatta (Pittsburgh, PA), C.-S. Chen (Loma Linda, CA), W.R. Clark (Anchorage, AK), B.D. Curti (Portland, OR), L.H. Dang (Gainesville, FL), G.A. Daniels (San Diego, CA), T. Dorff (Los Angeles, CA), C.G. Drake (Baltimore, MD), M.R. Fesen (Hutchinson, KS), T. Flaig (Aurora, CO), N.Y. Gabrail (Canton, OH), J. George (Mobile, AL), H. Ghazal (Hazard, KY), M.G. Goldstein (Frederick, MD), A.R. Golshayan (Charleston, SC), L. Gressot (Houston, TX), T.H. Guthrie (Jacksonville, FL), C.T. Hagenstad (Lawrenceville, GA), J.J. Hajdenberg (Orlando, FL), A. Hantel (Naperville, IL), V.R. Holden (Springfield, MO), S.P. Kahanic (Sioux City, IA), E.D. Kwon (Rochester, MN), C. Logothetis (Houston, TX), J. Lutzky (Miami Beach, FL), J. Migas (Normal, IL), B.C. Mirtsching (Dallas, TX), M.R. Modiano (Tucson, AZ), R. Mudad (Hollywood, FL), L. Oleksowicz (Cincinnati, OH), R.D. Page (Fort Worth, TX), D.J. Peace (Chicago, IL), B. Poiesz (Syracuse, NY), E. Posadas (Los Angeles, CA), E. Robin (Munster, IN), F.C. Schell (Lebanon, NH), H.I. Scher (New York, NY), M. Scholz (Marina Del Rey, CA), R. Sehgal (Huntington, WV), P. Sharma (Houston, TX), S.F. Slovin (New York, NY), R. Szmulewitz (Chicago, IL), S.T. Tagawa (New York, NY), M-E. Taplin (Boston, MA), D. Vaena (Iowa City, IA), P.J. Van Veldhuizen (Kansas City, MO), R.J. Vivacqua (Upland, PA), J.L. Wade, 3rd (Decatur, IL), R. Weber (San Francisco, CA), B. Yu (Salisbury, MD).
Funding/Support and role of the sponsor: This trial was designed jointly by the steering committee members and Bristol-Myers Squibb, which served as the study sponsor and provided funding support. Data were collected by the sponsor, and were analyzed and interpreted by the sponsor in collaboration with the steering committee members. An initial draft of this article was prepared by the first author (K.F.). All authors contributed to subsequent drafts and had final responsibility for the decision to submit for publication.
Financial disclosures: Karim Fizazi certifies that all conflicts of interest, including specific financial interests and relationships and affiliations relevant to the subject matter or materials discussed in the manuscript (eg, employment/affiliation, grants or funding, consultancies, honoraria, stock ownership or options, expert testimony, royalties, or patents filed, received, or pending), are the following: Karim Fizazi reports consulting and honoraria from Astellas, Bayer, CureVac, Janssen, Orion, and Sanofi. Charles Drake reports consulting for AZ Medimmune, Bayer, BMS, Compugen, F-Star, Genocea, Janssen, Kleo, Merck, Merck-Serono, Pfizer, Pierre Fabre, Roche/Genentech, Shattuck Labs, Tizona, Urogen, and Werewolf; patents (held by Johns Hopkins University) with BMS and Janssen; options with Compugen, Harpoon, Kleo, Tizona, Urogen, and Werewolf. Tomasz Beer reports research funding from Alliance Foundation Trials, Boehringer Ingelheim, Corcept Therapeutics, Endocyte Inc., Janssen Research & Development, Medivation, Inc./Astellas, OncoGenex, Sotio, and Theraclone Sciences/OncoResponse; consulting fees from AbbVie, AstraZeneca, Astellas Pharma, Bayer, BMS, Boehringer Ingelheim, Clovis Oncology, GlaxoSmithKline, Janssen Biotech, Janssen Japan, Merck, Novartis, and Pfizer; stock ownership in Salarius Pharmaceuticals and Arvinas Inc. Fons van der Eertwegh reports study grant from Sanofi, BMS, and Roche; travel expenses from MSD Oncology, Roche, Pfizer, and Sanofi; honoraria from Bristol-Myers Squibb; participating in the advisory boards of Bristol-Myers Squibb, MSD Oncology, Amgen, Roche, Novartis, Sanofi, Pfizer, Ipsen, and Merck. Nadine Houded reports consultancies with BMS. Neeraj Agarwal reports consultancy to Astellas, Astra Zeneca, BMS, Bayer, Clovis, Eisai, Exelixis, EMD Serono, Ely Lilly, Foundation Medicine, Genentech, Janssen, Merck, Novartis, Nektar, Pfizer, and Pharmacyclics. Andre Bergman participated in the advisory boards of Janssen Pharma, Bayer, Sanofi, and Astellas; received speaker fees from Astellas, Bayer, Janssen Pharma, and Astellas; received research grants from Sanofi and Astellas, not related to this study. M. Brent McHenry and Allen Chen are employees of BMS.
Footnotes
Appendix A. Supplementary data
Supplementary material related to this article canbe found, in the online version, at doi:https://doi.org/10.1016/j.eururo.2020.07.032.
References
- [1].Gillessen S, Attard G, Beer TM, et al. Management of patients with advanced prostate cancer: the report of the Advanced Prostate Cancer consensus Conference APCCC 2017. Eur Urol 2018;73:178–211. [DOI] [PubMed] [Google Scholar]
- [2].Horwich A, Hugosson J, de Reijke T, et al. Prostate cancer: ESMO consensus conference guidelines 2012. Ann Oncol 2013;24:1141–62. [DOI] [PubMed] [Google Scholar]
- [3].Fitzpatrick JM, Bellmunt J, Fizazi K, et al. Optimal management of metastatic castration-resistant prostate cancer: highlights from a European expert consensus panel. Eur J Cancer 2014;50:1617–27. [DOI] [PubMed] [Google Scholar]
- [4].LaFleur MW, Muroyama Y, Drake CG, Sharpe AH. Inhibitors of the PD-1 pathway in tumor therapy. J Immunol 2018;200:375–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Quinn DI, Shore ND, Egawa S, Gerritsen WR, Fizazi K. Immunotherapy for castration-resistant prostate cancer: progress and new paradigms. Urol Oncol 2015;33:245–60. [DOI] [PubMed] [Google Scholar]
- [6].Kwon ED, Hurwitz AA, Foster BA, et al. Manipulation of T cell costimulatory and inhibitory signals for immunotherapy of prostate cancer. Proc Natl Acad Sci USA 1997;94:8099–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Kwon ED, Foster BA, Hurwitz AA, et al. Elimination of residual metastatic prostate cancer after surgery and adjunctive cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade immunotherapy. Proc Natl Acad Sci USA 1999;96:15074–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Madan RA, Mohebtash M, Arlen PM, et al. Ipilimumab and a poxviral vaccine targeting prostate-specific antigen in metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 2012;13:501–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].van den Eertwegh AJ, Versluis J, van den Berg HP, et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 2012;13:509–17. [DOI] [PubMed] [Google Scholar]
- [10].Slovin S, Higano CS, Hamid O, et al. Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: results from an open-label, multicenter phase I/II study. Ann Oncol 2013;24:1813–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Graff JN, Alumkal JJ, Drake CG, et al. Early evidence of anti-PD-1 activity in enzalutamide-resistant prostate cancer. Oncotarget 2016;7:52810–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Hansen A, Massard C, Ott PA, et al. Pembrolizumab for patients with advanced prostate adenocarcinoma: preliminary results from the KEYNOTE-028 study. Ann Oncol 2016;27(Suppl 6):725PD. [DOI] [PubMed] [Google Scholar]
- [13].Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411–22. [DOI] [PubMed] [Google Scholar]
- [14].Fong L, Small EJ. Anti-cytotoxic T-lymphocyte antigen-4 antibody: the first in an emerging class of immunomodulatory antibodies for cancer treatment. J Clin Oncol 2008;26:5275–83. [DOI] [PubMed] [Google Scholar]
- [15].Kwon ED, Drake CG, Scher HI, et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184–043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol 2014;15:700–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Beer TM, Kwon ED, Drake CG, et al. Randomized, double-blind, phase III trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer. J Clin Oncol 2017;35:40–7. [DOI] [PubMed] [Google Scholar]
- [17].Kaur P, Asea A. Radiation-induced effects and the immune system in cancer. Front Oncol 2012;2:191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc 1958;53:457–81. [Google Scholar]
- [19].Brookmeyer R, Crowley J. A confidence interval for the median survival time. Biometrics 1982;38:29–41. [Google Scholar]
- [20].Cabel L, Loir E, Gravis G, et al. Long-term complete remission with Ipilimumab in metastatic castrate-resistant prostate cancer: case report of two patients. J Immunother Cancer 2017;5:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Jn Graff, Puri S Bifulco Cb, Ba Fox, Tm Beer. Sustained complete response to CTLA-4 blockade in a patient with metastatic, castration-resistant prostate cancer. Cancer Immunol Res 2014;2:399–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 2015;373:1627–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Bellmunt J, de Wit R, Vaughn DJ, et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med 2017;376:1015–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Champiat S, Dercle L, Ammari S, et al. Hyperprogressive disease is a newpatternof progression in cancer patients treated byanti-PD-1/ PD-L1. Clin Cancer Res 2017;23:1920–8. [DOI] [PubMed] [Google Scholar]
- [25].Morris MJ, Molina A, Small EJ, et al. Radiographic progression-free survival as a response biomarker in metastatic castration-resistant prostate cancer: COU-AA-302 results. J Clin Oncol 2015;33:1356–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014;371:2189–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature 2013;500:415–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;372:2509–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Simeone E, Gentilcore G, Giannarelli D, et al. Immunological and biological changes during ipilimumab treatment and their potential correlation with clinical response and survival in patients with advanced melanoma. Cancer Immunol Immunother 2014;63:675–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Delyon J, Mateus C, Lefeuvre D, et al. Experience in daily practice with ipilimumab for the treatment of patients with metastatic melanoma: an early increase in lymphocyte and eosinophil counts is associated with improved survival. Ann Oncol 2013;24:1697–703. [DOI] [PubMed] [Google Scholar]
- [31].Wu YM, Cieślik M, Lonigro RJ, et al. Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate Cancer. Cell 2018;173:1770–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Kwek SS, Lewis J, Zhang L, et al. Preexisting levels of CD4 T cells expressing PD-1 are related to overall survival in prostate cancer patients treated with ipilimumab. Cancer Immunol Res 2015;3: 1008–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Oh DY, Cham J, Zhang L, et al. Immune toxicities elicited by CTLA-4 blockade in cancer patients are associated with early diversification of the T-cell repertoire. Cancer Res 2017;77:1322–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018;359:91–7. [DOI] [PubMed] [Google Scholar]
- [35].US Yervoy (ipilimumab) [package insert]. Princeton, NJ: Bristol-Myers Squibb Company; 2013. [Google Scholar]
- [36].Dewan MZ, Galloway AE, Kawashima N, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res 2009;15:5379–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Demaria S, Kawashima N, Yang AM, et al. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res 2005;11:728–34. [PubMed] [Google Scholar]
- [38].Sharma P, Pachynski R, Narayan V, et al. Initial results from a phase II study of nivolumab (NIVO) plus ipilimumab (IPI) for the treatment of metastatic castration-resistant prostate cancer (mCRPC; Checkmate 650). 2019 Genitourinary Cancer symposium. J Clin Oncol 2019;37(7_suppl):142. [Google Scholar]
- [39].Boudadi K, Suzman DL, Anagnostou V, et al. Ipilimumab plus nivolumab and DNA-repair defects in AR-V7-expressing metastatic prostate cancer. Oncotarget 2018;9:28561–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.

