scholarly journals CTNI-18. FINAL RESULTS OF A PHASE 2 STUDY OF EFFICACY, SAFETY AND INTRATUMORAL PHARMACOKINETICS (PK) OF SELINEXOR MONOTHERAPY IN RECURRENT GLIOBLASTOMA (rGBM)

2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii46-ii46
Author(s):  
Andrew Lassman ◽  
Patrick Wen ◽  
Martin van den Bent ◽  
Scott Plotkin ◽  
Annemiek Walenkamp ◽  
...  

Abstract BACKGROUND Selinexor is an FDA-approved first-in-class, oral selective nuclear export inhibitor which forces nuclear retention of many tumor suppressor proteins. METHODS We conducted a phase 2 trial of selinexor monotherapy for adults with recurrent GBM including a surgical arm to explore intratumoral PK and 3 medical arms to optimize dosing. Prior treatment with radiotherapy and temozolomide was required; prior bevacizumab was exclusionary. The primary endpoint was 6-month progression-free survival (6mPFS) rate. RESULTS Selinexor administered ~2 hours pre-operatively yieleded average intratumoral concentration (136 nM, n=6) comparable to the in vitro IC50 (130 nM) from 7 primary human GBM cell lines. Among all 68 patients accrued to 3 medical arms (~85 mg BIW, n=24; 60 mg BIW, n=14; 80 mg QW, n=30), median age was 56 years (21–78). Median number of prior lines of therapies was 2 (1–7). At 80 mg QW, 28% patients were progression-free at the end of cycle 6; the 6mPFS was 17%; disese control rate by RANO was 37% (1 CR, 2 PRs, 7 SD) among 27 evaluable patients; responses were durable (median 11.1 months), and treatment lasted for 442, 547 and 1282 days in 3 responders, as of data lock, with one responder remaining on treatment off study; median overall survival was 10.2 months with 95% CI (7.0, 15.4). The ~85 mg BIW-schedule was abandoned due to poor tolerability. The related adverse events (all grades) in patients on ~85 mg BIW/60 mg BIW/80 mg QW were nausea (41.7%/64.3%/66.7%), fatigue (70.8%/71.4%/50.0%), neutropenia (29.2%/14.3%/33.3%), decreased appetite (45.8%/71.4%/26.7%), thrombocytopenia (66.7%/28.6%/23.3%) and weight loss (16.7%,/42.9%/6.7%). CONCLUSION Selinexor monotherapy demonstrated encouraging intratumoral penetration and efficacy, with durable disease control in rGBM. Monotherapy dose at 80 mg QW is recommended for further development in rGBM. A phase 1/2 study of combination therapy for newly diagnosed or rGBM has been initiated (NCT04421378).

2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 2005-2005 ◽  
Author(s):  
Andrew B. Lassman ◽  
Patrick Y. Wen ◽  
Martin J. Van Den Bent ◽  
Scott Randall Plotkin ◽  
Anna Maria Elisabeth Walenkamp ◽  
...  

2005 Background: New treatment modalities are needed for recurrent glioblastoma (rGBM). Selinexor (SEL) is a novel, oral selective inhibitor of nuclear export which forces nuclear retention of tumor suppressor proteins including p53 and p27, leading to apoptosis. We previously reported interim results showing tolerability, preliminary efficacy, and blood-brain barrier penetration in a surgical cohort (N = 8). We now report updated results following completion of accrual to non-surgical cohorts (N = 68). Methods: This is an open-label, multicenter, phase 2 study of SEL monotherapy. Patients (pts) not undergoing surgery for measurable rGBM (per RANO) were enrolled in one of 3 arms encompassing different dosing schedules. Treatment was continuous, although cycles were defined as 28 days and response was assessed every other cycle by MRI. Prior treatment with radiotherapy and temozolomide was required and prior bevacizumab was exclusionary. The primary endpoint was 6-month progression free survival (6mPFS) rate, calculated by the Kaplan Meier method. Results: A total of 76 pts were enrolled. Median age was 56 years (range 21-78). Median number of prior treatments was 2 (range 1-7). At the end of the 6 cycles, 30.2% patients on 80 mg QW were free from progression. The 6mPFS rate on 80 mg QW was 15.1%. Best RANO-defined responses (assessed locally) among 26 evaluable pts on 80 mg QW included 1 complete response, 2 partial responses, 7 stable disease, and 16 with progressive disease. Median duration of response was 10.8 months. The most common related adverse events in pts on ~85 mg BIW/60 mg BIW/80 mg QW were nausea (42%/64%/60%), leukopenia (38%/7%/43%), fatigue (71%/71%/43%), neutropenia (29%/14%/33%), decreased appetite (46%/71%/27%), and thrombocytopenia (67%/29%/23%). Conclusions: SEL demonstrated efficacy, with durable responses and disease stabilization in rGBM. Based on the favorable efficacy and safety profile, SEL at a dose of 80 mg QW is recommended for further development in rGBM. Clinical trial information: NCT01986348. [Table: see text]


Cancer ◽  
2019 ◽  
Vol 125 (21) ◽  
pp. 3790-3800 ◽  
Author(s):  
Evanthia Galanis ◽  
S. Keith Anderson ◽  
Erin L. Twohy ◽  
Xiomara W. Carrero ◽  
Jesse G. Dixon ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 541-541 ◽  
Author(s):  
Issa F. Khouri ◽  
Wei Wei ◽  
Rosamar Valverde ◽  
Martin Korbling ◽  
Francesco Turturro ◽  
...  

Abstract Background We previously reported the use of BFR, with phase 1 escalating daily doses of 70, 90, 110, or 130 mg/m2 of bendamustine daily x 3 days prior to transplantation without dose-limiting toxicity (Khouri et al. ASH 2011, Abstract # 894). In this study, we report results of an expanded phase 2 trial. Methods Bendamustine 130 mg/m2 IV daily on days -5 to - 3 prior to transplantation , together with 30 mg/m2 IV of fludarabine given on the same days. Rituximab was given at a dose of 375 mg/m2 IV on day –13 and 1000 mg/m2 on days -6, +1, and +8, as previously described. Rituximab was omitted in patients with T-cell diseases. Tacrolimus and mini-methotrexate were used for GVHD prophylaxis. In addition, thymoglobulin 1 mg/kg IV was given on days -2, and -1 in patients receiving an unrelated donor transplant. Results The analysis included all 56 patients who received BFR (48 from the expanded phase 2 trial, and 8 from the phase 1 part). Histologies included: mantle cell (n=16), CLL (n=15) (3 with 17p- and 2 Richter’s), follicular (n=13), Diffuse large cell (n=9), peripheral T-cell lymphoma (n=3). Median age was 56 (range, 59-70) years. Median prior treatments was 3 (range, 1-7); 7 (12%) had failed a prior autotransplant. At study entry,27 (48%) patients were in CR/CRu, 23 (41%) in PR, and 6 (11%) had refractory disease. Thirty (54%) received their transplants from HLA-compatible siblings and 26 (46%) from unrelated donors. Median number of CD34-positive cells infused was 5.58 x 106/kg. Neutrophil counts recovered to > 0.5 x 109/L a median of 6 days after transplantation (range, 0-16 days). The median number of days on G-CSF was 1.5 days (range, 0-8 days). Thirteen patients (23%) did not require G-CSF as they never experienced an ANC < 0.5 x 109/L. Forty-nine patients (87.5%) did not require platelet transfusions. Platelet counts recovered to > 20 x 109/L in the remaining 7 patients after a median of 11 days (range, 10-19 days). All patients engrafted donor cells. By day 30, median donor myeloid and T-cells were 85% and 97%, respectively. Both increased to 100% by day 90. Recovery of donor cells was similar in both sibling and unrelated donor transplantation. Acute grade 2-4 GvHD and chronic extensive GvHD were observed in 7 (12.5%) and 8 (14%) of patients respectively. Treatment-related mortality at 1-year was 9%. Six patients died: 2 of progression, 2 of infection and 2 of GVHD complications. With a median follow-up time of 12 months (range, 2-37 months), the OS and PFS rates were 89% and 80%, respectively. Conclusions Our results show that the BFR regimen with bendamustine at a dose level of 130 mg/m2 daily x 3 days, constitutes a well-tolerated nonmyeloablative allogeneic conditioning for lymphoid malignancies. It allows engraftment of both sibling and unrelated donor cells with minimal myelosuppression. This regimen can be used as a platform for allogeneic transplantation in the outpatient setting. Disclosures: Khouri: Cephalon: Research Funding. Off Label Use: Bendamustine in transplantation.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4773-4773 ◽  
Author(s):  
Christine I. Chen ◽  
Martin Gutierrez ◽  
David S. Siegel ◽  
Joshua R. Richter ◽  
Nina Wagner-Johnston ◽  
...  

Abstract Introduction: The nuclear export protein exportin 1, (XPO1) is overexpressed in a wide variety of cancers including MM and often correlate with poor prognosis. Selinexor (KPT-330) is an oral Selective Inhibitor of Nuclear Export (SINE) XPO1 antagonist in Phase 1 and 2 clinical studies. Selinexor forces nuclear retention and reactivation of tumor suppressor proteins (TSPs) and reduction of many proto-oncogenes, including MDM2, MYC and Cyclin D. In addition, selinexor potently deactivates NF-κB, through forced nuclear retention of IκBα. Together these effects induce selective apoptosis in MM cells and inhibition of NF-κB dependent osteoclast activation. XPO1 is also responsible for nuclear export of the glucocorticoid receptor (GR). We hypothesized that selinexor will enhance the activity of dexamethasone (DEX)-bound GR, resulting in synergistic tumor cell killing. Methods: In vitro tumor cell viability measurements were based on MTT (CellTiter 96¨/Promega) and combination indices were calculated using CalcuSyn software. For xenograft studies, utilized NOD-SCID mice with subcutaneous inoculation of MM.1s cells. GR nuclear localization was measured with immunofluorescent anti-GR (phosphor-S211) antibody and quantitative imaging. To assess GR transcriptional activation, GR binding to a GCR consensus sequence was measured in nuclear extracts using an ELISA method (GR ELISA kit/Affymetrix). Patients (pts) with heavily pretreated refractory MM were dosed with oral selinexor at doses of up to 60 mg/m2 (8-10 doses/4 wk cycle) as part of a Phase 1 program in advanced hematological malignancies. Response we defined based on the IMWG criteria. The effect of combining DEX with selinexor was analyzed in all pts who received selinexor at moderate to high doses (30-60 mg/m2). Safety and efficacy were analyzed separately in three groups: no DEX, <20 mg DEX and 20 mgs DEX. Results: In MM.1s cells Sel-Dex showed synergy for nuclear retention of the DEX activated GR (Ser211-phosphorylated) and concomitant GR transcriptional activation. Sel-Dex showed highly synergistic cytotoxicity in MM.1s cells in vitro and in vivo, with a corresponding increase in apoptosis. Selinexor alone was potently cytotoxic in the DEX resistant MM cell lines MM.1R and ANBL6, but addition of DEX provided no additional effect. Twenty-eight pts with heavily pretreated refractory MM (16 M, 12 F; median age 62; ECOG PS 0/1: 7/21; median prior regimens: 6) received selinexor at 30 – 60 mg/m2 with either 0, <20, or 20 mgs DEX. All pts have received a proteasome inhibitor and an Imid and the majority of the pts have received pomalidomide (68%) and/or carfilzomib (36%). The most common Grade 1/2 AEs for these three groups were: nausea (82%/86%/70%), fatigue (55%/86%/40%), anorexia (36%/71%/60%), and vomiting (36%/57%/10%). Of the 28 pts treated; 10 heavily pretreated refractory MM pts treated with a combination of selinexor (45 mg/m2 twice weekly) and DEX (20 mg with each selinexor dose) were found to have dramatically improved disease response (n=10, ORR 60%), with one stringent complete response (sCR, 10%), 5 partial responses (PR, 50%) and clinical benefit rate (CBR) rate of 80% (Figure 1). Treatment with ³30mg/m2 selinexor and <20 mg DEX (n=7), resulted in ORR of 14% and CBR of 86%, while treatment with selinexor (30-60 mg/m2) without DEX (n=12) showed best response of stable disease (50%). Sel-Dex was also associated with an increase in time on study relative to selinexor alone, with 7 of out 10 pts in the 20 mg DEX combo group still on study (11-25 weeks). Five additional pts were treated with selinexor at a dose of 60 mg/m2 in combination with 20 mg DEX. Response evaluation is pending. Conclusions: Sel-Dex combination is markedly synergistic in preclinical models, which is supported by the preliminary clinical data presented. One potential mechanism underlying this synergy is the amplification of GR activity due the combined effects of selinexor-induced nuclear retention of activated GR coupled with DEX-mediated GR agonism. These results provide a promising basis for the continuing study of Sel-Dex for treatment of pts with refractory MM. Phase 2 studies of Sel-Dex in pts with MM refractory to both pomalidomide and carfilzomib are planned for early 2015. Disclosures Chen: Celgene: Honoraria; Janssen: Honoraria. Off Label Use: Lenalidomide maintenance therapy after ASCT. Gutierrez:Senesco: PI Other. Siegel:Celgene, Millennium, Onyx: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Baz:Celgene: Research Funding; Millennium: Research Funding; Bristol Myers Squibb: Research Funding; Karyopharm: Research Funding; Sanofi: Research Funding. Kukreti:Celgene: Honoraria. Azmi:Karyopharm Therpeutics: Research Funding. Kashyap:Karyopharm Therapeutics: Employment. Landesman:Karyopharm Therapeutics: Employment. Marshall:Karyopharm Therpeutics: Employment. McCartney:Karyopharm Therpeutics: Employment. Saint-Martin:Karyopharm Therpeutics: Employment. Norori:Karyopharm Therpeutics: Consultancy. Savona:Karyopharm Therpeutics: Membership on an entity's Board of Directors or advisory committees. Rashal:Karyopharm Therapeutics: Employment. Carlson:Karyopharm Therapeutics: Employment. Mirza:Karyopharm Therpeutics: Consultancy, Membership on an entity's Board of Directors or advisory committees. Shacham:Karyopharm Therapeutics Inc.: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties. Kauffman:Karyopharm Therapeutics: Employment, Equity Ownership. Reece:Millennium: Honoraria, Research Funding; Millennium: Honoraria, Research Funding; Janssen: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria, Research Funding; Otsuka: Honoraria, Research Funding; Otsuka: Honoraria, Research Funding; Merck: Research Funding; Merck: Research Funding; BMS: Research Funding; BMS: Research Funding; Novartis: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Amgen : Honoraria; Amgen : Honoraria.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 8530-8530 ◽  
Author(s):  
Cristina Gasparetto ◽  
Brea Lipe ◽  
Sascha Tuchman ◽  
Natalie Scott Callander ◽  
Suzanne Lentzsch ◽  
...  

8530 Background: Selinexor is a novel, first-in-class selective inhibitor of nuclear export (SINE), which blocks exportin 1 (XPO1), forcing the nuclear retention and activation of tumor suppressor proteins. Selinexor in combination with low dose dexamethasone (Sel-dex) was approved by the FDA, based on data from the STORM study wherein Sel-dex induced an overall response rate (ORR) of 26.2% in patients (pts) with refractory MM. We hypothesize that once weekly (QW) SKd may be an active well tolerated regimen and evaluated this combination in a dose escalation/expansion study. Methods: STOMP is a phase 1b/2 study evaluating various doses and enrolled pts with carfilzomib naive relapsed MM. Oral selinexor was dosed QW at 80 or 100 mg. Carfilzomib was dosed QW (on days 1, 8 and 15 of 28-day cycle) at 56 mg/m2 or 70 mg/m2. Dexamethasone was dosed at 40 mg QW. The primary objectives of the study are to assess the maximum tolerated dose (MTD), recommended phase 2 dose (RP2D), as well as explore the efficacy and safety of SKd. Results: As of January 2020, 18 pts were enrolled. Median age was 71 years (range: 50-76). Median number of prior regimens was 4 (range: 1-8). All pts (n = 18) were previously treated with bortezomib and lenalidomide, and 50% and 56% pts were refractory to bortezomib and lenalidomide respectively. Nine (50%) pts received prior pomalidomide treatment and 8 (44%) pts were refractory. Eleven (61%) pts received prior daratumumab treatment and 9 (50%) were refractory. The MTD was selinexor 80 mg QW, carfilzomib 56 mg/m2 QW and dexamethasone 40 mg QW. The ORR and CBR were 72% and 79% respectively with 4 complete responses, 7 very good partial responses, 2 partial responses, and 1 minimal response. Stable disease was observed in 3 pts. With a median follow-up period of 4.7 (1.8-16.3) months, median progression-free survival has not been reached. Common treatment-related adverse events (total, Grade ≥3) were thrombocytopenia (83.3%, 66.7%), nausea (66.7%, 0%), anemia (55.6%, 11.1%), fatigue (50%, 11.1%), anorexia (44%, 5.6%), weight loss (44%, 0%), and neutropenia (33.3%, 11.1%). Conclusions: Once weekly SKd demonstrated an encouraging ORR of 72% in pts with a median of 4 lines of prior therapy. The majority of responses are deep and predominantly CR and VGPR. The combination is well tolerated with no new safety signal, no Grade ≥3 nausea, vomiting, diarrhea, weight loss or anorexia. The side effects are a function of the dose and schedule and can be managed with dose modification and supportive care. Enrolment is ongoing and supports a phase 3 study of SKd. Clinical trial information: NCT02343042 .


2019 ◽  
Vol 21 (Supplement_3) ◽  
pp. iii3-iii4 ◽  
Author(s):  
A B Lassman ◽  
P Y Wen ◽  
M van den Bent ◽  
S R Plotkin ◽  
A Walenkamp ◽  
...  

Abstract BACKGROUND New treatment modalities are needed for recurrent glioblastoma (rGBM). Selinexor is a novel, oral selective inhibitor of nuclear export which forces nuclear retention of tumor suppressor proteins including p53 and p27, leading to apoptosis. We previously reported interim results showing tolerability, preliminary efficacy, and blood-brain barrier penetration in a surgical cohort (N=8). We now report updated results following completion of accrual to non-surgical cohorts (N=68). MATERIALS AND METHODS This is an open-label, multicenter, phase 2 study of selinexor monotherapy. Patients (pts) not undergoing surgery for measurable rGBM per response assessment neuro-oncology criteria (RANO) were enrolled in one of 3 arms encompassing different dosing schedules of selinexor (50 mg/m2 [~ 85 mg] BIW, 60 mg BIW, and 80 mg QW). Treatment was continuous, although cycles were defined as 28 days and response was assessed every other cycle by MRI. Prior treatment with radiotherapy and temozolomide was required and prior bevacizumab was exclusionary. The primary endpoint was 6-month progression free survival (6mPFS) rate, calculated by the Kaplan-Meier method. RESULTS 76 pts were enrolled; 24, 14 and 30 pts on doses of ~85 mg BIW, 60 mg BIW, and 80 mg QW, respectively. Median age was 56 years (range 21–78). Median number of prior treatments was 2 (range 1–7) At the end of the 6 cycles, 30.2% pts on 80 mg QW were free from progression. The 6mPFS rate on 80 mg QW was 18.9%. Best RANO-defined responses (assessed locally) among 26 evaluable pts on 80 mg QW included 1 complete response, 2 partial responses, 7 stable disease, and 16 with progressive disease. Complete and partial responses were durable: the complete and a partial responder remain on selinexor for 393 and 1093 days respectively, as of the cut-off date. Median duration of response was 10.8 months. The most common related adverse events (all grades) in pts on ~85 mg BIW/60 mg BIW/80 mg QW were nausea (42%/64%/63%), leukopenia (38%/7%/43%), fatigue (71%/71%/47%), neutropenia (29%/14%/33%), decreased appetite (46%/71%/27%), and thrombocytopenia (67%/29%/23%). CONCLUSION Selinexor demonstrated efficacy, with durable responses and disease stabilization in rGBM. Based on the favorable efficacy and safety profile, selinexor at a dose of 80 mg QW is recommended for further development in rGBM.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii195-ii195
Author(s):  
Patrick Wen ◽  
Yazmin Odia ◽  
Minesh Mehta ◽  
Samuel Goldlust ◽  
Sharon Tamir ◽  
...  

Abstract BACKGROUND GBM is the most common and most aggressive primary brain tumor with poor prognosis and median overall survival (mOS) of patients with ndGBM and rGBM being 15 vs 5-7 months, respectively. Selinexor is a first-in class, oral, selective nuclear export inhibitor which forces nuclear retention and reactivation of many tumor suppressor proteins. Selinexor with low dose dexamethasone was recently approved for patients with triple-class refractory multiple myeloma. Additionally, selinexor monotherapy has demonstrated broad activity in other hematologic and solid malignancies. In a Phase 2 study in rGBM (NCT01986348), selinexor demonstrated encouraging intratumoral penetration and single-agent efficacy at 80 mg once weekly with durable response and disease stabilization in heavily pretreated patients. Preclinical GBM studies showed synergy when combining selinexor with radiation, temozolomide and lomustine. METHOD This is a phase 1 (PH-1) dose finding study followed by a 1:1 randomized phase 2 (PH-2; n= 350) efficacy exploration trial to independently evaluate 3 different combination regimens: Arm A: radiation +/- selinexor in uMGMT ndGBM; Arm B: radiation and temozolomide +/- selinexor in MGMT ndGBM; Arm C:omustine +/- selinexor in first relapse rGBM following frontline radiation and temozolomide. The PH-1 primary endpoint is MTD/RP2D, with secondary endpoints of ORR per modified RANO, duration of response (DOR), PFS, and OS. The PH-2 primary endpoint for Arms A and B in ndGBM is PFS, with key secondary endpoints being OS, PFS6, ORR, DOR. For Arm C, the PH-2 primary endpoint is OS while key secondary endpoints are PFS, PFS6, ORR, DOR. The study has 70% power to detect a hazard ratio of 0.67 between selinexor and control for primary efficacy for arms A & B, and 80% power to detect a hazard ratio of 0.70 for arm C. We are currently enrolling patients nationwide. Clinical trial identifier: NCT04421378


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Andrew J. Brenner ◽  
John Floyd ◽  
Lisa Fichtel ◽  
Joel Michalek ◽  
Kunal P. Kanakia ◽  
...  

AbstractEvofosfamide (Evo or TH302) is a hypoxia-activated prodrug which is reduced leading to the release of alkylating agent bromo-isophosphoramide mustard, which has shown safety and signals of efficacy in a prior phase 1 study in recurrent glioblastoma. We performed a dual center single-arm Phase II study to expand on the safety and efficacy of Evo plus bevacizumab in bevacizumab refractory glioblastoma. 33 patients with bevacizumab refractory GBM received Evo 670 mg/m2 in combination with Bevacizumab 10 mg/kg IV every 2 weeks. Assessments included adverse events, response, and survival. Median age of patients was 47 (range 19–76) and 24 (69%) were male. At the time of study entry, 9 (26%) had ongoing corticosteroid use. ECOG performance status was 0 or 1 in 83% of patients. Patients were mostly heavily pretreated with 77% have three or more prior regimens. A total of 12 patients (36%) suffered grade 3–4 drug associated adverse event (AE); no grade 5 AE were reported. Of the 33 evaluable patients, best response was PR in 3 (9%), SD in 14 (43%), and PD in 16 (48%) with responses confirmed by a second reviewer. Median time to progression of disease was 53 days (95% CI 42–113) and Median time to death was 129 days (95% CI 86–199 days). Progression free survival at 4 months (PFS-4) on Evo-Bev was 31%, which was a statistically significant improvement over the historical rate of 3%. The median overall survival of patients receiving Evo-Bevacizumab was 4.6 months (95% CI 2.9–6.6). The progression free survival of patients on Evo-Bevacizumab met the primary endpoint of progression free survival at 4 months of 31%, although the clinical significance of this may be limited. Given the patient population and Phase II design, these clinical outcomes will need further validation.


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 15005-15005
Author(s):  
E. G. Chiorean ◽  
T. Dragovich ◽  
J. Hamm ◽  
C. H. Barrios ◽  
C. F. Gorini ◽  
...  

15005 Background: Glufosfamide is glucose linked to isophosphoramide mustard, the active metabolite of ifosfamide. Cancer cells use glucose at a higher rate than normal cells, which may lead to preferential metabolic targeting by GLU. The Phase 1 study established a GLU dose of 4500 mg/m2 for the GLU + GEM regimen. The objectives of the Phase 2 part of this study are to evaluate the safety and efficacy of GLU+GEM in pts with pancreatic adenocarcinoma. Methods: Eligible pts had metastatic and/or locally advanced pancreatic adenocarcinoma previously untreated with chemotherapy, Karnofsky Performance Status =70, creatinine clearance (CrCL) =60 mL/min and acceptable hematologic and liver function. Pts received GLU 4500 mg/m2 iv over 4 hours on Day 1 and GEM 1000 mg/m2 iv over 30 minutes on Days 1, 8 and 15 of every 28-day cycle. CT scans were obtained every 8 weeks. Primary endpoint was response rate. Results: Twenty-nine pts were enrolled. One patient with ineligible histology was excluded from efficacy analyses. The 14 male/15 female pts had a median age of 59 years. Median cycles on treatment was 4 (range 1–10+) and 9 pts completed all 6 cycles. Six of 28 (21%; 95% CI: 8- 41%) pts had a partial response (duration 1.0+ to 5.8+ months), one unconfirmed. Ten of 28 (36%) pts had stable disease (median duration 5.3 months). Median progression-free survival was 3.7 months. Six-month survival was 54% (95% CI: 38–78%). Grade 3/4 neutropenia and thrombocytopenia occurred in 21/29 (72%) and 8/29 (28%) pts. Five pts (18%) had a GLU-related serious adverse event (SAE). Two pts died due to SAE unrelated to GLU. Three pts developed renal failure; two were GLU-related with evidence of renal tubular acidosis (RTA). One pt developed GLU-related SAE of RTA without renal failure. The CrCL fell below 60 mL/min in 7 of 27 (26%) pts with CrCL =60 at baseline. Conclusions: Preliminary data indicate that GLU + GEM may benefit pts with chemotherapy naïve pancreatic adenocarcinoma. [Table: see text]


2014 ◽  
Vol 32 (4_suppl) ◽  
pp. 495-495
Author(s):  
Richard C. Lauer ◽  
Ian Rabinowitz ◽  
Terry Novak ◽  
Martin J. Edelman

495 Background: B is a small molecule proteasome inhibitor that affects multiple signaling pathways. S is an orally active multikinase inhibitor with known activity in renal cell cancer. In vitro data has shown that S and B interact synergistically in a number of neoplastic cell lines to cause apoptosis. A phase 1 trial of S and B demonstrated no unexpected toxicities. We now report the results of this phase 2 efficacy trial. Methods: Eligibility included cytologically confirmed clear cell cancer with no prior chemotherapy, PS 0-1, Cr < 1.5 mg/dl, normal LFTs. Regimen: S 200 mg PO BID and B 1mg/m2 IV days 1,4,8, & 11 every 21 days. Patients were treated until disease progression or unacceptable toxicity. The primary objective of the study was to achieve a PFS of 70 weeks. Results: Seventeen patients were enrolled between April of 2011 and January of 2013. Median age was 62y (range 44-75). Four of 17 patients had known brain metastasis on entry to the trial. Median number of cycles = 4 (range 1-45+). Response: CR/PR/SD/PD =0/1/12/4 (RR: 6%, 95% CI = 0%, 29%) Median progression free survival was 13.7 weeks; median overall survival was 110 weeks Toxicity: See table. Only 1 pts. treatment was stopped due to an AE of pancreatitis. There were no toxic deaths. The study was halted for futility. Conclusions: 1.The combination of S and B was well tolerated. 2. The RR of 6% and PFS of 13.7 weeks is not superior to S as a single agent. 3. The combination of SB is not recommended for further development. Clinical trial information: NCT 01100242.


Sign in / Sign up

Export Citation Format

Share Document