Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Rajesh Bandekar is active.

Publication


Featured researches published by Rajesh Bandekar.


Lancet Oncology | 2014

Siltuximab for multicentric Castleman's disease: a randomised, double-blind, placebo-controlled trial

Frits van Rhee; Raymond Siu Ming Wong; Nikhil C. Munshi; Jean François Rossi; Xiao Yan Ke; Alexander Fosså; David Simpson; Marcelo Capra; Ting Liu; Ruey Kuen Hsieh; Yeow Tee Goh; Jun Zhu; Seok-Goo Cho; Hanyun Ren; James Cavet; Rajesh Bandekar; Margaret Rothman; Thomas A. Puchalski; Manjula Reddy; Helgi van de Velde; Jessica Vermeulen; Corey Casper

BACKGROUND Multicentric Castlemans disease is a rare lymphoproliferative disorder driven by dysregulated production of interleukin 6. No randomised trials have been done to establish the best treatment for the disease. We assessed the safety and efficacy of siltuximab-a chimeric monoclonal antibody against interleukin 6-in HIV-negative patients with multicentric Castlemans disease. METHODS We did this randomised, double-blind, placebo-controlled study at 38 hospitals in 19 countries worldwide. We enrolled HIV-negative and human herpesvirus-8-seronegative patients with symptomatic multicentric Castlemans disease. Treatment allocation was randomised with a computer-generated list, with block size six, and stratification by baseline corticosteroid use. Patients and investigators were masked to treatment allocation. Patients were randomly assigned (2:1) to siltuximab (11 mg/kg intravenous infusion every 3 weeks) or placebo; all patients also received best supportive care. Patients continued treatment until treatment failure. The primary endpoint was durable tumour and symptomatic response for at least 18 weeks for the intention-to-treat population. Enrolment has been completed. The study is registered with ClinicalTrials.gov, number NCT01024036. FINDINGS We screened 140 patients, 79 of whom were randomly assigned to siltuximab (n=53) or placebo (n=26). Durable tumour and symptomatic responses occurred in 18 (34%) of 53 patients in the siltuximab group and none of 26 in the placebo group (difference 34·0%, 95% CI 11·1-54·8, p=0·0012). The incidence of grade 3 or more adverse events (25 [47%] vs 14 [54%]) and serious adverse events (12 [23%] vs five [19%]) was similar in each group despite longer median treatment duration with siltuximab than with placebo (375 days [range 1-1031] vs 152 days [23-666]). The most common grade 3 or higher were fatigue (five vs one), night sweats (four vs one), and anaemia (one vs three). Three (6%) of 53 patients had serious adverse events judged reasonably related to siltuximab (lower respiratory tract infection, anaphylactic reaction, sepsis). INTERPRETATION Siltuximab plus best supportive care was superior to best supportive care alone for patients with symptomatic multicentric Castlemans disease and well tolerated with prolonged exposure. Siltuximab is an important new treatment option for this disease. FUNDING Janssen Research & Development.


Clinical Cancer Research | 2014

A Phase I/II, Multiple-Dose, Dose-Escalation Study of Siltuximab, an Anti-Interleukin-6 Monoclonal Antibody, in Patients with Advanced Solid Tumors

Eric Angevin; Josep Tabernero; Elena Elez; Steven J. Cohen; Rastilav Bahleda; Jean-Luc Van Laethem; Christian C. Ottensmeier; José A. López-Martín; Sally Clive; Florence Joly; Isabelle Ray-Coquard; Luc Dirix; Jean-Pascal Machiels; Neil N. Steven; Manjula Reddy; Brett Hall; Thomas A. Puchalski; Rajesh Bandekar; Helgi van de Velde; Brenda B. Tromp; Jessica Vermeulen; Razelle Kurzrock

Purpose: This phase I/II study evaluated safety, efficacy, and pharmacokinetics of escalating, multiple doses of siltuximab, a chimeric anti-interleukin (IL)-6 monoclonal antibody derived from a new Chinese hamster ovary (CHO) cell line in patients with advanced/refractory solid tumors. Experimental Design: In the phase I dose-escalation cohorts, 20 patients with advanced/refractory solid tumors received siltuximab 2.8 or 5.5 mg/kg every 2 weeks or 11 or 15 mg/kg every 3 weeks intravenously (i.v.). In the phase I expansion (n = 24) and phase II cohorts (n = 40), patients with Kirsten rat sarcoma-2 (KRAS)-mutant tumors, ovarian, pancreatic, or anti-EGF receptor (EGFR) refractory/resistant non–small cell lung cancer (NSCLC), colorectal, or H&N cancer received 15 mg/kg every 3 weeks. The phase II primary efficacy endpoint was complete response, partial response, or stable disease >6 weeks. Results: Eighty-four patients (35 colorectal, 29 ovarian, 9 pancreatic, and 11 other) received a median of three (range, 1–45) cycles. One dose-limiting toxicity occurred at 5.5 mg/kg. Common grade ≥3 adverse events were hepatic function abnormalities (15%), physical health deterioration (12%), and fatigue (11%). Ten percent of patients had siltuximab-related grade ≥3 adverse events. Neutropenia (4%) was the only possibly related adverse event grade ≥3 reported in >1 patient. Serious adverse events were reported in 42%; most were related to underlying disease. The pharmacokinetic profile of CHO-derived siltuximab appears similar to the previous cell line. No objective responses occurred; 5 of 84 patients had stable disease >6 weeks. Hemoglobin increased ≥1.5 g/dL in 33 of 47 patients. At 11 and 15 mg/kg, completely sustained C-reactive protein suppression was observed. Conclusions: Siltuximab monotherapy appears to be well tolerated but without clinical activity in solid tumors, including ovarian and KRAS-mutant cancers. The recommended phase II doses were 11 and 15 mg/kg every 3 weeks. Clin Cancer Res; 20(8); 2192–204. ©2014 AACR.


American Journal of Hematology | 2015

A phase 2, randomized, double-blind, placebo-controlled study of siltuximab (anti-IL-6 mAb) and bortezomib versus bortezomib alone in patients with relapsed or refractory multiple myeloma.

Robert Z. Orlowski; Liana Gercheva; Cathy Williams; Heather J. Sutherland; Tadeusz Robak; Tamas Masszi; Vesselina Goranova-Marinova; Meletios A. Dimopoulos; J Cavenagh; Ivan Spicka; Angelo Maiolino; Alexander Suvorov; Joan Bladé; Olga Samoylova; Thomas A. Puchalski; Manjula Reddy; Rajesh Bandekar; Helgi van de Velde; Hong Xie; Jean-Franςois Rossi

We compared the safety and efficacy of siltuximab (S), an anti‐interleukin‐6 chimeric monoclonal antibody, plus bortezomib (B) with placebo (plc) + B in patients with relapsed/refractory multiple myeloma in a randomized phase 2 study. Siltuximab was given by 6 mg/kg IV every 2 weeks. On progression, B was discontinued and high‐dose dexamethasone could be added to S/plc. Response and progression‐free survival (PFS) were analyzed pre‐dexamethasone by European Group for Blood and Marrow Transplantation (EBMT) criteria. For the 281 randomized patients, median PFS for S + B and plc + B was 8.0 and 7.6 months (HR 0.869, P = 0.345), overall response rate was 55 versus 47% (P = 0.213), complete response rate was 11 versus 7%, and median overall survival (OS) was 30.8 versus 36.8 months (HR 1.353, P = 0.103). Sustained suppression of C‐reactive protein, a marker reflective of inhibition of interleukin‐6 activity, was seen with S + B. Siltuximab did not affect B pharmacokinetics. Siltuximab/placebo discontinuation (75 versus 66%), grade ≥3 neutropenia (49 versus 29%), thrombocytopenia (48 versus 34%), and all‐grade infections (62 versus 49%) occurred more frequently with S + B. The addition of siltuximab to bortezomib did not appear to improve PFS or OS despite a numerical increase in response rate in patients with relapsed or refractory multiple myeloma.


European Urology | 2016

Phase 2 Study of the Safety and Antitumor Activity of Apalutamide (ARN-509), a Potent Androgen Receptor Antagonist, in the High-risk Nonmetastatic Castration-resistant Prostate Cancer Cohort

Matthew R. Smith; Emmanuel S. Antonarakis; Charles J. Ryan; William R. Berry; Neal D. Shore; Glenn Liu; Joshi J. Alumkal; Celestia S. Higano; Edna Chow Maneval; Rajesh Bandekar; Carla J. de Boer; Margaret K. Yu; Dana E. Rathkopf

Background Apalutamide is a potent androgen receptor (AR) antagonist that targets the AR ligand-binding domain and prevents AR nuclear translocation, DNA binding, and transcription of AR gene targets. Objective: To evaluate the activity and safety of apalutamide in patients with high-risk nonmetastatic castration-resistant prostate cancer (nmCRPC). Design, setting, and participants We conducted a multicenter phase 2 study of nmCRPC patients with a high risk for progression (prostate-specific antigen [PSA] ≥8 ng/ml or PSA doubling time [PSA DT] ≤10 mo). Intervention Patients received 240 mg/d apalutamide while continuing on androgen-deprivation therapy. Outcome measurements and statistical analysis Primary end point was 12-wk PSA response (Prostate Cancer Working Group 2 criteria). Secondary end points included safety, time to PSA progression (TTPP), and metastasis-free survival (MFS). Results and limitations A total of 51 patients were enrolled; four patients with metastatic disease were excluded from the efficacy analysis. Patient characteristics included median age, 71 yr; Eastern Cooperative Oncology Group performance status 0 (76%); Gleason score ≤ 7 (57%); median PSA 10.7 ng/ml; and PSA DT ≤10 mo (45%). At median follow-up of 28.0 mo, 18 patients (35%) remained in the study. Overall, 89% of patients had ≥50% PSA decline at 12 wk. Median TTPP was 24.0 mo (95% confidence interval [CI], 16.3 mo–not reached [NR]); median MFS was NR (95% CI, 33.4 mo–NR). Most of the patients discontinued study treatment (n = 33) due to disease progression (n = 11 [22%]) or adverse events (AEs) (n = 9 [18%]). The most common AE was fatigue (any grade, n = 31 [61%]) although grade ≥3 fatigue was uncommon (n = 2 [4%]). These represent the first apalutamide nmCRPC patient clinical data. Conclusions In high-risk nmCRPC patients, apalutamide was safe with robust activity based on durable PSA responses and disease control. Patient summary Antitumor activity and the safety of apalutamide in patients with nonmetastatic castration-resistant prostate cancer support continued development in this setting. Trial registration ClinicalTrials.gov identifier NCT01171898


Clinical Cancer Research | 2015

Analysis of Inflammatory and Anemia-Related Biomarkers in a Randomized, Double-Blind, Placebo-Controlled Study of Siltuximab (Anti-IL6 Monoclonal Antibody) in Patients With Multicentric Castleman Disease

Corey Casper; Shalini Chaturvedi; Nikhil C. Munshi; Raymond Sm Wong; Ming Qi; Michael Schaffer; Rajesh Bandekar; Brett Hall; Helgi van de Velde; Jessica Vermeulen; Manjula Reddy; Frits van Rhee

Purpose: Siltuximab (IL6 antibody) is approved for the treatment of multicentric Castleman disease (MCD). Effects of IL6 inhibition on the inflammatory milieu accompanying MCD have not been characterized. Experimental Design: Trends in inflammatory- and anemia-associated markers, measured over the course of a placebo-controlled study of siltuximab (11 mg/kg q3w) in patients with MCD (n = 79), were characterized. Results: Baseline IL6 and C-reactive protein (CRP) levels were significantly correlated (r = 0.708; P < 0.0001). CRP levels decreased (median, 92%) by cycle 1 day 8 (C1D8), remaining suppressed during siltuximab treatment while remaining stable in the placebo group. There were no associations between baseline CRP or IL6 and MCD symptom burden, histologic subtype, ethnicity, maximum CRP decrease, and response parameters. A hemoglobin response (change ≥ 15 g/L at week 13) was observed with siltuximab (61%; P = 0.0002). Median hepcidin decrease from baseline at C1D8 with siltuximab was 47% versus median 11% increase with placebo. Maximum post-baseline changes in hepcidin levels among siltuximab recipients were correlated with maximum changes for hemoglobin (r = −0.395; P = 0.00607), total iron-binding capacity (TIBC; r = −0.354; P = 0.01694), and ferritin (r = 0.599; P = 0.0001). Greater median changes from baseline in ferritin, hemoglobin, and TIBC were observed in anemic siltuximab-treated patients. Conclusions: IL6 neutralization with siltuximab resulted in sustained CRP suppression and improvement of anemia, in part, by hepcidin pathway inhibition. Clin Cancer Res; 21(19); 4294–304. ©2015 AACR.


Clinical Cancer Research | 2017

Safety and antitumor activity of apalutamide (ARN-509) in metastatic castration-resistant prostate cancer with and without prior abiraterone acetate and prednisone

Dana E. Rathkopf; Emmanuel S. Antonarakis; Neal D. Shore; Ronald Tutrone; Joshi J. Alumkal; Charles J. Ryan; Mansoor N. Saleh; Ralph J. Hauke; Rajesh Bandekar; Edna Chow Maneval; Carla J. de Boer; Margaret K. Yu; Howard I. Scher

Purpose: To evaluate the efficacy of apalutamide before or after treatment with abiraterone acetate and prednisone (AAP) in patients with progressive metastatic castration-resistant prostate cancer (mCRPC). Experimental Design: Two cohorts were studied: AAP-naïve and post-AAP patients who had received ≥6 months of AAP. Patients had progressive mCRPC per rising prostate-specific antigen (PSA) and/or imaging, without prior chemotherapy exposure. All received apalutamide 240 mg/day. Primary endpoint was ≥50% decline in 12-week PSA according to Prostate Cancer Working Group 2 criteria. Secondary endpoints included time to PSA progression and time on treatment. Results: Forty-six patients enrolled in the AAP-naïve (n = 25) and post-AAP (n = 21) cohorts. The 12-week PSA response rate was 88% (22/25) and 22% (4/18), median time to PSA progression was 18.2 months [95% confidence interval (CI), 8.3 months–not reached) and 3.7 months (95% CI, 2.8–5.6 months), and median time on treatment 21 months (range, 2.6–37.5) and 4.9 months (range, 1.3–23.2), for the AAP-naïve and post-AAP cohorts, respectively. Eighty percent (95% CI, 59–93) and 64% (95% CI, 43–82) of AAP-naïve and 43% (95% CI, 22–66) and 10% (95% CI, 1–30) of post-AAP patients remained on treatment for 6+ and 12+ months, respectively. Common treatment-emergent adverse events in both cohorts were grade 1 or 2 fatigue, diarrhea, nausea, and abdominal pain. Conclusions: Apalutamide was safe, well tolerated, and demonstrated clinical activity in mCRPC, with 80% of AAP-naïve and 43% of post-AAP patients, remaining on treatment for 6 months or longer. Clin Cancer Res; 23(14); 3544–51. ©2017 AACR.


Clinical pharmacology in drug development | 2014

A phase 1, randomized study to assess the pharmacokinetic comparability of siltuximab derived from two different cell lines in healthy subjects

Chao Xu; Chao Han; Joseph C. Marini; Joyce Ford; Stanley J. Marciniak; Manny Lopez; Bart Frederick; Dick E. de Vries; Rajesh Bandekar; Hugh M. Davis; Honghui Zhou; Thomas A. Puchalski

Siltuximab, a monoclonal antibody (mAb) against interleukin (IL‐6), is under development by Janssen Research & Development, LLC. During early clinical development, siltuximab was produced in a murine Sp2/0 myeloma cell line. The production cell line was switched to stably transfected Chinese hamster ovary (CHO) cell line for subsequent clinical development. A two‐part, parallel‐group, phase 1 study was designed to evaluate the safety and pharmacokinetics (PK) of a single IV administration of Sp2/0‐ and CHO‐derived siltuximab in healthy subjects. The results from this study demonstrated PK comparability of siltuximab produced from Sp2/0 and CHO cell lines. The 90% confidence interval of the ratios of geometric means of Cmax and AUC0–84day following 1.4 mg/kg doses was (99.4%, 111.3%) and (98.1%, 109.6%), respectively, both within the pre‐specified comparability range of 80–125%. Siltuximab derived from either the Sp2/0 or CHO cell lines was in general well tolerated and was not found to be immunogenic in this study.


Cancer Research | 2014

Abstract CT239: ARN-509 in patients with metastatic castration-resistant prostate cancer (mCRPC) previously treated with abiraterone acetate (AA)

Dana E. Rathkopf; Emmanuel S. Antonarakis; Neal D. Shore; Ronald Tutrone; Joshi J. Alumkal; Charles J. Ryan; Mansoor N. Saleh; Ralph J. Hauke; Rajesh Bandekar; Edna Chow Maneval; Carla J. de Boer; Mary Beth Todd; Margaret K. Yu; Howard I. Scher

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Background: ARN-509 is a novel second-generation antiandrogen that binds directly to the ligand-binding domain of the androgen receptor (AR), impairing AR nuclear translocation and DNA binding to the androgen response element. Phase 2 of a multicenter phase 1/2 study evaluates ARN-509 activity in 3 distinct patient populations of men with CRPC: 1) nonmetastatic chemotherapy-naive CRPC; 2) chemotherapy-naive mCRPC; 3) mCRPC post-AA treatment. Study ARN-509-001 is the first to prospectively examine response to novel second-generation antiandrogens post-AA treatment. We present the results of the post-AA treated cohort, as of July 2013. Methods: All patients had mCRPC with progressive disease based on rising prostate-specific antigen (PSA) and/or imaging. No prior chemotherapy for CRPC was allowed. Patients in the AA-pretreated cohort had to have been treated with AA for at least 6 months. All patients received ARN-509 at the recommended phase 2 dose of 240 mg/d (Rathkopf et al. J Clin Oncol. 2013). The primary end point was PSA response at 12 weeks according to the Prostate Cancer Working Group 2 criteria. Secondary end points included safety, time to PSA progression, and objective response rates. PSA assessments were collected every 4 weeks and tumor imaging was performed every 12 weeks. Results: By July 2013, 21 patients were enrolled and treated in the post-AA cohort. The median age was 67 years (range 48-83). At baseline, 62% of patients had an Eastern Cooperative Oncology Group performance status 0, and 29% had a Gleason score ≥ 8; median PSA was 58.4 ng/mL. Median duration on ARN-509 treatment post AA was 5.6 months (range 1.9-16.7). At 12 weeks, 24% (5/21) of patients had ≥ 50% decline in PSA from baseline. Median time to PSA progression was 16 weeks (95% confidence interval, 12-31 weeks). The best objective response was stable disease in 4 (36%) patients. Patients discontinued the study due to disease progression (n = 13), adverse events (n = 2), consent withdrawn (n = 1), and other reasons (n = 4). The most common treatment-related adverse events were fatigue (n = 11), nausea (n = 5), and diarrhea (n = 3). Conclusions: In men with mCRPC, post-AA treatment, ARN-509 is safe and well tolerated, with modest activity in a subset of patients who develop resistance to AA. Clinical trial information: [NCT01171898][1]. Citation Format: Dana E. Rathkopf, Emmanuel S. Antonarakis, Neal D. Shore, Ronald Tutrone, Joshi J. Alumkal, Charles J. Ryan, Mansoor N. Saleh, Ralph J. Hauke, Rajesh Bandekar, Edna Chow Maneval, Carla de Boer, Mary Todd, Margaret K. Yu, Howard I. Scher. ARN-509 in patients with metastatic castration-resistant prostate cancer (mCRPC) previously treated with abiraterone acetate (AA). [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr CT239. doi:10.1158/1538-7445.AM2014-CT239 [1]: /lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT01171898&atom=%2Fcanres%2F74%2F19_Supplement%2FCT239.atom


Cancer Research | 2015

Abstract CT134: Androgen receptor (AR) mutations in patients (pts) with castration-resistant prostate cancer (CRPC) with and without prior abiraterone acetate (AA) treatment

Dana E. Rathkopf; Matthew R. Smith; Emmanuel S. Antonarakis; Charles J. Ryan; William R. Berry; Neal D. Shore; Glenn Liu; Celestia S. Higano; Joshi J. Alumkal; Ralph J. Hauke; Ronald Tutrone; Mansoor N. Saleh; Edna Chow Maneval; Shibu Thomas; Deborah Ricci; Margaret K. Yu; Carla J. de Boer; Angela Trinh; Thian Kheoh; Rajesh Bandekar; Howard I. Scher

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Background: ARN-509 is a second-generation antiandrogen with antitumor activity in CRPC. Activating mutations in the AR ligand-binding domain (LBD) have been associated with resistance to first- (T877A) and second- (F876L) generation antiandrogens. We evaluated the type and frequency of relevant AR LBD mutations in ARN-509-treated CRPC pts at baseline (BL) and disease progression (PD). Methods: ARN-509-001 was a phase I/II study that evaluated ARN-509 activity in nonmetastatic (M0), chemotherapy-naive, and post-AA CRPC. Of the 97 pts enrolled in phase II at a dose of 240 mg/d, 92 were evaluable for the mutation analysis at BL and 82 at PD. Relevant mutations in circulating tumor DNA were detected using a digital PCR method called BEAMing (Beads, Emulsification, Amplification, and Magnetics) (Richardson AL. Clin Cancer Res. 2012). Results: Median duration of therapy was ∼16 months. One pt in the M0 cohort and one in the chemotherapy-naive cohort had the F876L mutation at BL. Two pts in the chemotherapy-naive cohort and one in the post-AA cohort acquired the AR F876L mutation during treatment. Pts with M0 CRPC did not acquire a mutation (Table 1). Three pts in the post-AA cohort had the T877A mutation at BL; the T877A mutation was not detected in any other cohort at BL. In the post-AA cohort, one pt acquired the T877A mutation during treatment while another lost the mutation (Table). The two pts with detectable F876L at BL developed prostate-specific antigen (PSA) progression at 4 and 6 months, respectively, compared with a median time to PSA progression of 16.4 months in the remainder of pts. Conclusions: Pts with metastatic CRPC who were treated with ARN-509 had a low rate of acquisition of the AR F876L (3/82 = 4%) and AR T877A (1/82 = 1%) mutations. These results suggest that ARN-509 may be continued in the setting of a rising PSA. Larger studies are needed to confirm the prevalence of F876L, T877A, and the conversion rate. | | | AR F876L | AR T877A | |:------------------ | --------- | -------- | -------- | -------- | | | Evaluable | BL | PD | Acquired | BL | PD | Acquired | | | patients | n/N | n/N | | n/N | n/N | | | Total | 92 | 2/92 | 5/82 | 3/82 | 3/92 | 3/82 | 1/82 | | M0 | 49 | 1/49 | 1/47 | 0/47 | 0/50 | 0/47 | 0/47 | | Chemotherapy-naive | 24 | 1/24 | 3/20 | 2/20 | 0/20 | 0/20 | 0/20 | | Post-AA | 19 | 0/19 | 1/15 | 1/15 | 3/19 | 3/15 | 1/15 | Citation Format: Dana E. Rathkopf, Matthew R. Smith, Emmanuel S. Antonarakis, Charles J. Ryan, William R. Berry, Neal D. Shore, Glenn Liu, Celestia Higano, Joshi J. Alumkal, Ralph Hauke, Ronald Tutrone, Mansoor Saleh, Edna Chow Maneval, Shibu Thomas, Deborah Ricci, Margaret K. Yu, Carla J. de Boer, Angela Trinh, Thian Kheoh, Rajesh Bandekar, Howard I. Scher. Androgen receptor (AR) mutations in patients (pts) with castration-resistant prostate cancer (CRPC) with and without prior abiraterone acetate (AA) treatment. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr CT134. doi:10.1158/1538-7445.AM2015-CT134


Annals of Oncology | 2014

761PDNOVEL ANTIANDROGEN ARN-509 IN HIGH-RISK NON-METASTATIC (M0) CASTRATION-RESISTANT PROSTATE CANCER (CRPC)

Matthew R. Smith; Emmanuel S. Antonarakis; Charles J. Ryan; William R. Berry; Neal D. Shore; Glenn Liu; Joshi J. Alumkal; Celestia S. Higano; E. Chow-Maneval; Rajesh Bandekar; C. J. de Boer; Mary Beth Todd; Margaret K. Yu; Dana E. Rathkopf

ABSTRACT Aim: ARN-509 selectively binds the androgen receptor and impairs its nuclear translocation and DNA binding to androgen response elements. Study ARN-509-001 evaluates ARN-509 activity in pts with high-risk M0 CRPC, chemotherapy-naive metastatic CRPC (mCRPC), and mCRPC post abiraterone acetate. We report the updated results for the M0 CRPC cohort as of February 2014. Methods: All pts had CRPC with no radiographic evidence of metastases (pelvic lymph nodes Results: M0 CRPC cohort enrolled 51 pts from Nov 2011 to Jun 2012; 4 pts had metastatic disease and were excluded from the efficacy analysis. Median age was 71 years (range 51–88). Baseline characteristics were: ECOG performance status, 0 (76%); Gleason score, ≤ 7 (57%); median PSA, 10.7 ng/mL (range 0.5-201.7); and PSADT, ≤ 10 mos (45%). All pts received prior treatment with a luteinizing hormone–releasing hormone analog with or without a first-generation antiandrogen. At median follow-up of 20.2 mos, 29 (57%) pts remain on study. Pts discontinued due to disease progression (n = 7), adverse events (AEs) (n = 7), or other reasons (n = 8). Most common treatment-emergent AEs: fatigue (57%), diarrhea (41%), nausea (29%), dysgeusia (20%), arthralgia (18%), and weight decreased (16%). No seizures were reported. Efficacy M0 CRPC N 47 PSA responsea,b, n (%) 12 wks 42/46 (91) 24 wks 40/46 (87) 36 wks 22/46 (48) Median time to PSA progression, wks (95% CI) NR (67, NR) Median MFS, wks (95% CI) NR (NR, NR) NR, not reached. a≥ 50% decline in PSA from baseline per PCWG2. b1 pt did not have postbaseline PSA results. Conclusions: ARN-509 is well tolerated in pts with high-risk M0 CRPC with robust activity based on durable PSA responses and disease control. Disclosure: M.R. Smith: has served as a consultant/advisor to and received research funding from Janssen; E.S. Antonarakis: has received honoraria and research funding from Janssen; C.J. Ryan: has received honoraria from Janssen;W. Berry: has served on a speaker bureau for Janssen;N.D. Shore: has served as a consultant/advisor to Astellas, Algeta, Bayer, Ferring, Dendreon, Janssen, Millennium, Sanofi, and BNIT;G. Liu: has received research funding from Aragon and Janssen;C. Higano: has served as a consultant/advisor to Astellas/Medivation, Bayer, Dendreon, and Johnson & Johnson, and has received research funding from Algeta, Aragon, Astellas/Medivation, Dendreon, and Sanofi;R. Bandekar, C. De Boer, M. Todd and M. Yu: is an employee of Janssen and holds stock in Johnson & Johnson;D.E. Rathkopf: has served as a consultant/advisor for Johnson & Johnson and has received research funding (uncompensated) from Celgene, Janssen/J&J, Medivation/Astellas, Millennium, and Novartis. All other authors have declared no conflicts of interest.

Collaboration


Dive into the Rajesh Bandekar's collaboration.

Top Co-Authors

Avatar

Ming Qi

Janssen Pharmaceutica

View shared research outputs
Top Co-Authors

Avatar

Helgi van de Velde

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Dana E. Rathkopf

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Emmanuel S. Antonarakis

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Neal D. Shore

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge