scholarly journals Selective Inhibition of Nuclear Export With Oral Selinexor for Treatment of Relapsed or Refractory Multiple Myeloma

2018 ◽  
Vol 36 (9) ◽  
pp. 859-866 ◽  
Author(s):  
Dan T. Vogl ◽  
David Dingli ◽  
Robert Frank Cornell ◽  
Carol Ann Huff ◽  
Sundar Jagannath ◽  
...  

Purpose Selinexor, a first-in-class, oral, selective exportin 1 (XPO1) inhibitor, induces apoptosis in cancer cells through nuclear retention of tumor suppressor proteins and the glucocorticoid receptor, along with inhibition of translation of oncoprotein mRNAs. We studied selinexor in combination with low-dose dexamethasone in patients with multiple myeloma refractory to the most active available agents. Patients and Methods This phase II trial evaluated selinexor 80 mg and dexamethasone 20 mg, both orally and twice weekly, in patients with myeloma refractory to bortezomib, carfilzomib, lenalidomide, and pomalidomide (quad-refractory disease), with a subset also refractory to an anti-CD38 antibody (penta-refractory disease). The primary end point was overall response rate (ORR). Results Of 79 patients, 48 had quad-refractory and 31 had penta-refractory myeloma. Patients had received a median of seven prior regimens. The ORR was 21% and was similar for patients with quad-refractory (21%) and penta-refractory (20%) disease. Among patients with high-risk cytogenetics, including t(4;14), t(14;16), and del(17p), the ORR was 35% (six of 17 patients). The median duration of response was 5 months, and 65% of responding patients were alive at 12 months. The most common grade ≥ 3 adverse events were thrombocytopenia (59%), anemia (28%), neutropenia (23%), hyponatremia (22%), leukopenia (15%), and fatigue (15%). Dose interruptions for adverse events occurred in 41 patients (52%), dose reductions occurred in 29 patients (37%), and treatment discontinuation occurred in 14 patients (18%). Conclusion The combination of selinexor and dexamethasone has an ORR of 21% in patients with heavily pretreated, refractory myeloma with limited therapeutic options.

2020 ◽  
pp. JCO.20.02259
Author(s):  
Paul G. Richardson ◽  
Albert Oriol ◽  
Alessandra Larocca ◽  
Joan Bladé ◽  
Michele Cavo ◽  
...  

PURPOSE Melphalan flufenamide (melflufen) is a first-in-class peptide-drug conjugate that targets aminopeptidases and rapidly and selectively releases alkylating agents into tumor cells. The phase II HORIZON trial evaluated the efficacy of melflufen plus dexamethasone in relapsed and refractory multiple myeloma (RRMM), a population with an important unmet medical need. PATIENTS AND METHODS Patients with RRMM refractory to pomalidomide and/or an anti-CD38 monoclonal antibody received melflufen 40 mg intravenously on day 1 of each 28-day cycle plus once weekly oral dexamethasone at a dose of 40 mg (20 mg in patients older than 75 years). The primary end point was overall response rate (partial response or better) assessed by the investigator and confirmed by independent review. Secondary end points included duration of response, progression-free survival, overall survival, and safety. The primary analysis is complete with long-term follow-up ongoing. RESULTS Of 157 patients (median age 65 years; median five prior lines of therapy) enrolled and treated, 119 patients (76%) had triple-class–refractory disease, 55 (35%) had extramedullary disease, and 92 (59%) were refractory to previous alkylator therapy. The overall response rate was 29% in the all-treated population, with 26% in the triple-class–refractory population. In the all-treated population, median duration of response was 5.5 months, median progression-free survival was 4.2 months, and median overall survival was 11.6 months at a median follow-up of 14 months. Grade ≥ 3 treatment-emergent adverse events occurred in 96% of patients, most commonly neutropenia (79%), thrombocytopenia (76%), and anemia (43%). Pneumonia (10%) was the most common grade 3/4 nonhematologic event. Thrombocytopenia and bleeding (both grade 3/4 but fully reversible) occurred concomitantly in four patients. GI events, reported in 97 patients (62%), were predominantly grade 1/2 (93%); none were grade 4. CONCLUSION Melflufen plus dexamethasone showed clinically meaningful efficacy and a manageable safety profile in patients with heavily pretreated RRMM, including those with triple-class–refractory and extramedullary disease.


2008 ◽  
Vol 26 (30) ◽  
pp. 4952-4957 ◽  
Author(s):  
Peter H. Wiernik ◽  
Izidore S. Lossos ◽  
Joseph M. Tuscano ◽  
Glen Justice ◽  
Julie M. Vose ◽  
...  

PurposeThe major cause of death in aggressive lymphoma is relapse or nonresponse to initial therapy. Lenalidomide has activity in a variety of hematologic malignancies, including non-Hodgkin's lymphoma (NHL). We report the results of a phase II, single-arm, multicenter trial evaluating the safety and efficacy of lenalidomide oral monotherapy in patients with relapsed or refractory aggressive NHL.Patients and MethodsPatients were treated with oral lenalidomide 25 mg once daily on days 1 to 21, every 28 days, for 52 weeks, until disease progression or intolerance. The primary end point was response; secondary end points included duration of response, progression-free survival (PFS), and safety.ResultsForty-nine patients with a median age of 65 years received lenalidomide in this study. The most common histology was diffuse large B-cell lymphoma (53%), and patients had received a median of four prior treatment regimens for NHL. An objective response rate of 35% was observed in 49 treated patients, including a 12% rate of complete response/unconfirmed complete response. Responses were observed in each aggressive histologic subtype tested (diffuse large B-cell, follicular center grade 3, mantle cell, and transformed lymphomas). Of patients with stable disease or partial response at first assessment, 25% improved with continued treatment. Estimated median duration of response was 6.2 months, and median PFS was 4.0 months. The most common grade 4 adverse events were neutropenia (8.2%) and thrombocytopenia (8.2%); the most common grade 3 adverse events were neutropenia (24.5%), leukopenia (14.3%), and thrombocytopenia (12.2%).ConclusionOral lenalidomide monotherapy is active in relapsed or refractory aggressive NHL, with manageable side effects.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2452-2452
Author(s):  
Huiqiang Huang ◽  
Yan Gao ◽  
Huilai Zhang ◽  
Keshu Zhou ◽  
Jianqiu Wu ◽  
...  

Abstract Background The prognosis of R/R PTCL and ENKTL remains poor. Current therapeutic options are limited, highlighting the need for novel approaches. ATG-010 (selinexor) is a novel, oral selective inhibitor of nuclear export, which blocks exportin 1. It has demonstrated clinical activity in different hematological malignancies. Preclinical synergistic anti-tumor effects are confirmed when combined with gemcitabine, cisplatin, or etoposide. Method The aim of this study was to evaluate safety and efficacy of ATG-010 plus GemOx or ICE regimen followed by ATG-010 maintenance in patients (pts) with R/R PTCL or ENKTL in China. The study planned to enroll 30 pts with PTCL-NOS, AITL, ALCL, PTCL-TFH, FTL and ENKTL. Pts must have had prior exposure to an anthracycline-based regimen for PTCL or an asparaginase-based regimen for ENKTL, and were relapsed or refractory from the last therapy. ATG-010 was administered orally (60 mg day 1, 8) plus standard-dose of either ICE or GemOx regimen every 3 weeks as per investigator's choice. If response was achieved after initial 2-6 cycles, pts continued to receive ATG-010 maintenance at the dose of 60 mg weekly until disease progression, intolerability or withdrawal of consent. Primary endpoints included safety according to the NCI CTCAE 5.0 and overall response rate (ORR) evaluated by investigators according to the Lugano criteria (2014). Secondary endpoints included duration of response (DOR), progression-free survival (PFS) and overall survival (OS). This trial was registered at ClinicalTrials.gov (NCT04425070). Results From Aug 18, 2020 to June 25, 2021, 24 pts (20 in GemOx cohort, 4 in ICE cohort) were enrolled and received at least one cycle of treatment. In this abstract, we report results of the GemOx cohort with 9 (45%) PTCL-NOS, 6 (30%) ENKTL, 4 (20%) AITL, and 1 (5%) ALCL ALK-. At study entry, median age was 55 years (range 35-69); 17 (85%) had stage III/IV disease. Five (36%) pts with PTCL had IPI score ≥3, and 5 (83%) ENKTL pts with had PINK-E score ≥2. Median number of prior regimens was 3.5 (range 1-7) with 5.5 for ENKTL and 2 for PTCL. All pts were refractory from last therapy; 11 (55%) pts had received gemcitabine-based regimens. Median time from last-line therapy to this trial was 1.5 months (range 1.0-16.7). The most common treatment-emergent adverse events (TEAEs) were hematological toxicities. All grade hematological TEAEs were neutropenia (90%), thrombocytopenia (90%) and anemia (85%). Grade≥3 hematological TEAEs included neutropenia (85%), thrombocytopenia (80%) and anemia (40.0%). The most common (>30%) non-hematological TEAEs were nausea (70%), diarrhea (65%), decreased appetite (65%), asthenia/fatigue (60%), vomiting (50%), pyrexia (40%), and hyponatremia (35%). Grade≥3 non-hematological TEAEs (≥10%) were diarrhea (15%) and pyrexia (10%). Serious TEAEs occurred in 7 (35%) pts with the most common being thrombocytopenia (20%). Three (15%) pts discontinued treatment due to TEAEs. TEAEs with a fatal outcome occurred in 1(5%) patient, who experienced rapid disease progression before death. The majority of adverse events were manageable by dose modification and supportive care. Of 17 efficacy evaluable pts, ORR was 52.9% (9/17), and CR rate was 35.3% (6/17). ORR of PTCL-NOS, ENKTL, AITL, ALCL were 62.5% (5/8), 60% (3/5), 0 (0/3), 100% (1/1); CR rate were 37.5%, 40%, 0, 100%, respectively. At a median follow-up of 6.1months, median PFS, DOR and OS of the whole cohort was 2.9, 3.1 months, and not reached (6-month OS rate 68.9%), respectively. Patients with ENKTL enjoyed a relatively longer median PFS (4.7 months). Conclusions ATG-010 plus GemOx regimen showed a manageable safety profile, and favorable activity with impressive CR rate in refractory pts, potentially offering a new therapeutic option for heavily pretreated pts with refractory PTCL or ENKTL. Disclosures Lou: Antengene Therapeutics Ltd.: Current Employment. Fei: Antengene Therapeutics Ltd.: Current Employment. Wang: Antengene Therapeutics Ltd.: Current Employment. Lynch: Antengene Therapeutics Ltd.: Current Employment.


Blood ◽  
2018 ◽  
Vol 132 (24) ◽  
pp. 2546-2554 ◽  
Author(s):  
Nizar J. Bahlis ◽  
Heather Sutherland ◽  
Darrell White ◽  
Michael Sebag ◽  
Suzanne Lentzsch ◽  
...  

Abstract Selinexor is an oral inhibitor of the nuclear export protein exportin 1. Preclinical studies demonstrated synergistic antimyeloma activity between selinexor and proteasome inhibitors (PI) through suppression of NF-κB signaling and nuclear retention of tumor suppressor proteins. We tested selinexor in combination with low-dose bortezomib and dexamethasone (SVd) for the treatment of relapsed or refractory multiple myeloma (MM). The primary objectives of this study were to determine the safety profile, overall response rate (ORR), and a recommended phase 2 dose (RP2D) of SVd. We enrolled 42 patients to receive selinexor (60, 80, or 100 mg orally) plus bortezomib (1.3 mg/m2 subcutaneously) and dexamethasone (20 mg orally) once or twice weekly in 21- or 35-day cycles. Patients had a median of 3 (range 1-11) prior lines of therapy, and 50% were refractory to a PI. Treatment-related grade 3 or 4 adverse events reported in ≥10% of patients were thrombocytopenia (45%), neutropenia (24%), fatigue (14%), and anemia (12%). Incidence (4 patients, 10%) and grade (≤2) of peripheral neuropathy were low. The ORR for the entire population was 63%: 84% ORR for PI nonrefractory and 43% for PI-refractory patients. The median progression-free survival for all patients was 9.0 months; 17.8 months for PI nonrefractory, and 6.1 months for PI refractory. SVd treatment produced high response rates in patients with relapsed or refractory MM, including borezomib-refractory MM, with no unexpected side effects. The RP2D is selinexor (100 mg once weekly), bortezomib (1.3 mg/m2 once weekly for 4 weeks), and dexamethasone (40 mg once weekly) per 35-day cycle. This trial was registered at www.clinicaltrials.gov as #NCT02343042.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 8038-8038
Author(s):  
Cristina Gasparetto ◽  
Gary J. Schiller ◽  
Sascha Tuchman ◽  
Natalie Scott Callander ◽  
Muhamed Baljevic ◽  
...  

8038 Background: Exportin 1 (XPO1) mediates the nuclear export and functional inactivation of tumor suppressor proteins (TSPs), is associated with poor prognosis in MM, and contributes to proteasome inhibitor (PI) and immunomodulatory drug (IMiD) resistance. Selinexor (SEL) is a novel, oral, first-in-class selective inhibitor of nuclear export (SINE) compound that blocks XPO1, forcing the nuclear retention and activation of TSPs. SEL in combination with low dose dexamethasone (dex) ± bortezomib (BOR) is FDA approved for previously treated MM. The synergy of SEL with the PI BOR has been confirmed in the phase 3 BOSTON study in MM patients (pts) with 1-3 prior therapies; once weekly (QW) SEL, QW BOR, and dex (XVd) significantly increased progression-free survival (PFS), time to next therapy, and overall response rate (ORR) as compared to standard twice weekly BOR/dex (Vd), despite XVd using 40% less BOR and 25% less dex than standard Vd. We hypothesized that the addition of QW SEL to the PI carfilzomib (CAR)-dex (XKd) would be an active and tolerable regimen in pts with heavily pretreated MM. Methods: In the XKd arm of the multi-arm Phase 1b/2 STOMP study, SEL at 80 or 100 mg QW was evaluated in combination with CAR at 56 or 70 mg/m2 QW plus dex at 40 mg QW in pts with heavily pretreated MM not refractory to CAR. Study objectives were to determine the maximum tolerated dose and recommended phase 2 dose (RP2D) and assess the safety and activity of the XKd regimen. Results: As of 4Jan2021, 27 pts were enrolled: 18 (67%) were male, median age 71 years (range 50-76), and median of 4 (range 1-8) prior lines of therapy. All 27 pts were previously treated with BOR, 26 (96%) lenalidomide (LEN), 19 (70%) pomalidomide (POM), 18 (67%) daratumumab (dara). The majority (67%) of pts were triple-class pretreated (PI, IMiD, and anti-CD38 mAb), and 44% had triple-class refractory MM. Nine pts (33%) had MM quad-refractory to BOR, LEN, POM, and dara. Common hematologic treatment-related adverse events (TRAEs) (total, grade ≥3) included thrombocytopenia (74%, 56%), anemia (59%, 19%), and neutropenia (30%, 7%). Non-hematologic TRAEs included nausea (67%, 4%), fatigue (52%, 7%), and anorexia (52%, 4%). RP2D was identified as SEL 80 mg QW, CAR 56 mg/m2 QW, and dex 40 mg QW. As of 3Feb2021, ORR was 78% (21/27) with 5 pts reaching CR (19%), 8 VGPR (30%), and 8 PR (30%). Median PFS was 23.7 months. Among 18 pts pretreated with dara, ORR was 67% and median PFS 23.7 months. In 9 pts whose MM was refractory to BOR, LEN, POM, and dara, ORR was 67% with 4 VGPR (44%). Conclusions: In pts with heavily pretreated MM, weekly XKd is highly active with an ORR of 78% and deep responses (≥VGPR 48%) with an overall PFS of 23 months. All AEs including grade 3/4 thrombocytopenia can be managed with appropriate supportive care and dose modifications. These data support further investigation of XKd in pts with previously treated MM including those previously treated with dara.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5151-5151
Author(s):  
Maria Teresa Petrucci ◽  
Vincenza Martini ◽  
Cristiano Gallucci ◽  
Erica Finolezzi ◽  
Anna Levi ◽  
...  

Abstract Velcade (PS-341, Bortezemib), a proteasome inhibitor, has recently been approved for the treatment of relapsed and refractory multiple myeloma (MM). We hereby report the results obtained using this new drug at our center. Between April 2003 and December 2004 we treated 23 patients with MM. The patient population consists of 13 males and 10 females, with a median age of 53 years (range 32–72); 16 were IgG MM, 6 IgA, 1 light chain. All patients were in stage III of their disease, with a median time of observation (from diagnosis to Velcade therapy) of 59 months (range 11–120), were pretreated with at least two lines of therapies and were refractory or in relapse after the last treatment. Velcade 1.3 mg/m2 was scheduled on days 1, 4, 8 and 11 of a 21-day treatment cycle for 8 cycles according to the tolerability and response. All cycles were delivered in day hospital. A median of 6 cycles (range 3–8) were administered to the overall population. Eleven patients concluded their program, while 12 discontinued the treatment: 1 because she received an allogeneic stem cell transplantation, 7 for adverse events and 4 due to disease progression. In this heavily pretreated population, our primary end point was to obtain a decline in the monoclonal component (MC) of at least 25%. All patients were considered evaluable for response because treated with at least 3 cycles of therapy. Thirteen patients responded to treatment: 1 (4%) obtained a disappearance of the MC, 4 (17%) achieved a reduction of the MC level >75% and 8 (34%) >50%. Six further patients (26%) obtained a decline in the MC <25%. The median number of cycles required to achieve a response was 3 (range 1–8) and the median duration of response was 6 months, with 8 patients presently still responsive. Among the responding patients, 5 relapsed and 4 of them have died due to disease progression. Among the 4 patients (17%) who did not respond to Velcade, 1 died. The majority of adverse events, which resolved with the discontinuation of treatment, were nausea, vomiting, diarrhea, fatigue, thrombocytopenia and peripheral neuropathy. Taking into consideration the very poor prognostic likelihood of our patients, this study adds further evidence con the efficacy of this new drug in MM. Velcade can be considered an effective anti-myeloma drug even though its toxicity must be taken into account in designing new clinical trials.


2016 ◽  
Vol 34 (34) ◽  
pp. 4094-4101 ◽  
Author(s):  
Thomas B. Alexander ◽  
Norman J. Lacayo ◽  
John K. Choi ◽  
Raul C. Ribeiro ◽  
Ching-Hon Pui ◽  
...  

Purpose To characterize the toxicity, pharmacokinetics, and pharmacodynamics of selinexor, a selective inhibitor of nuclear export, when combined with fludarabine and cytarabine, in children with relapsed or refractory leukemia. Patients and Methods Eighteen patients with relapsed or refractory acute leukemia were enrolled in the SELHEM (Selinexor With Fludarabine and Cytarabine for Treatment of Refractory or Relapsed Leukemia or Myelodysplastic Syndrome) clinical trial (NCT02212561). Selinexor, initially at 30 mg/m2 per dose, was given orally on days 1, 3, 8, 10, 22, and 24 and was escalated according to a rolling-six design. Fludarabine 30 mg/m2 and cytarabine 2 g/m2 were administered on days 15 to 19. Pharmacokinetic and pharmacodynamic studies were performed on days 1 and 22. Response evaluations were performed on day 15 and at the completion of course 1. Results Among the 17 patients who were evaluable for toxicity, three were treated at 30 mg/m2, three at 40 mg/m2, six at 55 mg/m2, and five at 70 mg/m2. The most common grade 3 nonhematologic toxicity was asymptomatic hyponatremia. Two patients who were treated at 70 mg/m2 experienced reversible cerebellar toxicity, thereby defining the dose-limiting toxicity. Pharmacokinetic parameters demonstrated that plasma exposure was dose proportional. Fifteen of 16 patients demonstrated at least a twofold increase of XPO1 mRNA, indicating inhibition of the XPO1 protein. In this group of heavily pretreated, relapsed, and refractory patients, seven of 15 evaluable patients (47%) achieved complete response or complete response with incomplete count recovery. Conclusion Selinexor, in combination with fludarabine and cytarabine, is tolerable at doses up to 55 mg/m2 in pediatric patients with relapsed or refractory leukemia. All patients who received selinexor at ≥ 40 mg/m2 demonstrated XPO1 target inhibition. Response rates are promising and will be further explored in a phase II trial.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 258-258 ◽  
Author(s):  
Christine Chen ◽  
Ramiro Garzon ◽  
Martin Gutierrez ◽  
Meagan A. Jacoby ◽  
Peter Brown ◽  
...  

Abstract Background: The nuclear export protein XPO1 is overexpressed in all types of hematological malignancies. The SINE selinexor (KPT-330) is a novel, first-in-class, slowly reversible XPO1 antagonist that forces the nuclear retention and activation of over tumor suppressor proteins (TSPs) such as p53, IkB, FOXO and p21. Forced nuclear retention of TSPs leads to their reactivation, which can counteract a multitude of oncogenic pathways that perpetuate the neoplastic phenotype. In addition, XPO1 inhibition prevents the eIF4e-mediated export of messenger mRNA for a variety of oncoproteins (FLT3, c-KIT, cyclin D1, c-MYC, Bcl2), leading to decreased expressed to further provide anti-cancer activity. An ongoing Phase 1 (NCT01607892) open label, dose escalation, multi-center study in hematological malignancies was designed to evaluate the safety and tolerability of selinexor as well as response rate as the secondary objective. A maximum tolerated dose (MTD) was not identified yet in this study, but based on the ongoing Phase1 study of selinexor in pts with solid tumors, a MTD of 65 mg/m2 twice weekly was determined. The goal of the analyses reported here was to identify the recommended Phase 2 dose (RP2D) based on both tolerability and efficacy in patients with heavily pretreated hematological malignancies. Methods: A subset (N=157) of the Phase 1 patient population received oral selinexor twice weekly (8 doses/28-d cycle). General clinical observations suggested that doses of selinexor >35 mg/m2 (> ~60 mg flat dose) are associated with suboptimal tolerability. Therefore, based on the actual dose administered, patients were divided into groups receiving 45-65 mg (median 60 mg; N=59) and >65 mg (70-160 mg; median 90 mg, N=98) for comparison of safety and efficacy endpoints. The majority of the patients have heavily pretreated myeloma, NHL, and AML. Results: 157 pts received 45-160 mg selinexor twice weekly (89 M/68 F, median age 66 yr; median 4 prior regimens). The most common adverse events (AEs) were fatigue (66%), nausea (64%), anorexia (55%), vomiting (38%), which were mostly gr 1/2, and thrombocytopenia (44%), which was mostly grade 3/4. Incidence of certain selinexor-related high grade (3/4) adverse events was greater in pts receiving >65 mg selinexor vs those receiving 45-65 mg (Table 1). Grade 3 nausea (4%), anorexia (3%), vomiting (3%) and hypokalemia (3%) were observed in the >65 mg group but were not seen in the 45-65 mg group. Grade 3 and 4 anemia were 19% vs 14% and 4% vs 2% for the >65 mg vs 45-65 mg groups, respectively. Grade 3 and 4 thrombocytopenia was similar in both groups, but slightly higher in the >65 mg group, with 8% and 24 % in the 45-65 mg group vs 12% and 27% in the >65 mg group. Neutropenia was also very similar in Grade 3 and 4 toxicity for both groups with 10% and 12% in the 45-65 mg group vs 11% and 10% in the >65 mg group. In contrast, high grade cataract was only seen in the 45-65 mg group (8%; 3 gr 3, 1 gr 4). Selinexor-induced weight loss, as compared to baseline, was maximal by the end of cycle 2 in both dose groups, without further loss through at least cycle 4, but the >65 mg group lost on average >5-fold more weight (average of 3.8 ± 1.1 kg vs 0.7 ± 0.1 kg in the 65 mg group from 56 d- 126 d; p<0.001). Also, pts in the 45-65 mg group remained on study longer (average of 101 d vs 69 d in the >65 mg group; p=0. 05). In contrast, overall efficacy in the two dose groups was comparable, with 5 complete responses (CRs, 10%) and an overall response rate (ORR) of 23% in the 45-65 mg group and 4 CRs (5%) and ORR of 24% in the >65 mg group. A listing of all responses for both groups can be seen in Table 2. Conclusions: While efficacy is comparable, doses of selinexor from 45-65 mg (median 60 mg) are better tolerated than doses >65 mg, based upon decreased weight loss, incidence of high grade AEs, and greater numbers of days on study. Based on this superior risk-benefit, a flat dose of 60 mg selinexor, twice weekly, is the RP2D for patients with hematological cancers. Similar results have been observed for solid tumors. Disclosures Chen: Celgene: Consultancy, Honoraria, Research Funding. Jacoby:Novo Nordisk: Consultancy; Sunesis: Research Funding. Stone:AROG: Consultancy; Celator: Consultancy; Novartis: Research Funding; Pfizer: Consultancy; Juno: Consultancy; Celgene: Consultancy; Karyopharm: Consultancy; Amgen: Consultancy; Agios: Consultancy; Roche/Genetech: Consultancy; Abbvie: Consultancy; Sunesis: Consultancy, Other: DSMB for clinical trial; Merck: Consultancy. Baz:Sanofi: Research Funding; Celgene Corporation: Research Funding; Karyopharm: Research Funding; Millennium: Research Funding. Gabrail:Sanofi: Honoraria, Speakers Bureau; Janssen: Speakers Bureau; BI: Honoraria, Speakers Bureau; Onyx: Honoraria, Speakers Bureau. Wang:Celgene: Research Funding. Martin:Janssen: Consultancy, Honoraria; Acerta: Consultancy; Gilead: Consultancy; Celgene: Consultancy; Novartis: Consultancy; Bayer: Consultancy. Siegel:Celgene Corporation: Consultancy, Speakers Bureau; Amgen: Speakers Bureau; Takeda: Speakers Bureau; Novartis: Speakers Bureau; Merck: Speakers Bureau. Marshall:Karyopharm: Employment. Saint-Martin:Karyopharm: Employment. Carlson:Karyopharm: Employment. Shacham:Karyopharm: Employment, Equity Ownership. Kauffman:Karyopharm: Employment, Equity Ownership. Kuruvilla:Gilead: Consultancy; Hoffmann LaRoche: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria; Seattle Genetics: Consultancy, Honoraria; Merck: Honoraria; Bristol-Myers Squibb: Honoraria; Lundbeck: Honoraria; Karyopharm: Honoraria.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5374-5374
Author(s):  
Shotaro Hagiwara ◽  
Shinichiro Okamoto ◽  
Kosei Matsue ◽  
Shinsuke Iida ◽  
Kazutaka Sunami ◽  
...  

Abstract Introduction: Pomalidomide plus low-dose dexamethasone (POM + LoDEX) is approved for the treatment of patients with relapsed and refractory multiple myeloma (RRMM). In RRMM, POM + LoDEX significantly prolonged progression-free survival (PFS) compared with POM alone (MM-002; Richardson et al Blood, 2014) and significantly improved PFS and overall survival (OS) compared with high-dose DEX alone (MM-003; San Miguel et al Lancet Oncol, 2013), while demonstrating an acceptable safety profile. In Japanese patients with RRMM, a phase 1 study identified POM 4 mg/day as the tolerated dose (MM-004; Iida ASH 2014), consistent with observations in Caucasian patients. The study presented here, MM-011, was conducted to evaluate the efficacy and safety of POM + LoDEX in Japanese patients with RRMM. Methods: MM-011 was a multicenter, single-arm, open-label phase 2 study in patients with RRMM. Eligible patients had received ≥ 2 prior therapies, including ≥ 2 cycles of lenalidomide and ≥ 2 cycles of bortezomib (either separately or in combination) and had developed progressive disease on or within 60 days of the last prior therapy. Patients received POM 4 mg orally on days 1-21, and LoDEX orally on days 1, 8, 15, and 22 of each 28-day cycle. LoDEX was given at 40 or 20 mg in patients aged ≤ 75 or > 75 years, respectively. Treatment was continued until disease progression, unacceptable toxicity, or withdrawal of consent. The primary endpoint was overall response rate (ORR; partial response rate [PR] or better) of POM + LoDEX according to the International Myeloma Working Group criteria. The required sample size was calculated using the expected response rate of 25%, the threshold response rate of 10% on 1-sided alpha of 0.05, and the statistical power of 80% based on the test for 1 sample proportion. Thirty-three pts were needed as a result of this calculation. Results: A total of 36 patients were enrolled. Median age was 64.5 years (range, 43-78 years), and 11% were aged > 75 years. Patients had a high tumor burden (81% had Durie-Salmon stage II or III disease) and were heavily pretreated (median of 6.5 prior anti-myeloma regimens; range, 2-15 regimens). All but 1 patient (97%) were refractory to lenalidomide and 58% were refractory to both lenalidomide and bortezomib. At the data cutoff (February 3, 2015), 16 patients (44%) remained on treatment. Disease progression was the most common reason for discontinuation (n = 14; 39%). All 36 patients enrolled were evaluable for efficacy. The ORR was 42% (n = 15 [95% CI, 26%-58%]), including complete response in 3% (n = 1) and PR in 39% (n = 14). Median PFS was 10.2 months (median follow-up, 4.6 months), and median OS was not reached (median follow-up, 7.7 months). In the 15 responders, median time to response was 1.9 months and median duration of response was not reached. The most common grade 3/4 hematologic adverse events (AEs) were neutropenia (n = 23; 64%), anemia (n = 15; 42%), and thrombocytopenia (n = 11; 31%), and the most common grade 3/4 non-hematologic AEs were pneumonia (n = 3; 8%) and decreased appetite (n = 3; 8%). Peripheral neuropathy (any grade) occurred in 8% (n = 3). No deep vein thrombosis or pulmonary embolism was reported. Overall, AEs were similar to those in other POM studies, and no new significant AEs were reported in Japanese RRMM patients. Three patients (8%) had an AE that led to discontinuation. Three patients died on study (or within 28 days of the last dose of study drug). Two of these patients died due to multiple myeloma and 1 due to AE (asthma and pneumonia, suspected to be related to study drug). Conclusions: POM + LoDEX is an effective regimen in heavily pretreated Japanese patients with RRMM with acceptable safety profiles that are comparable with those of POM studies in RRMM in other regions. Disclosures Hagiwara: Celgene Corporation: Membership on an entity's Board of Directors or advisory committees. Matsue:Celgene Corporation: Honoraria. Iida:Celgene Corporation: Honoraria, Research Funding; Janssen Pharmaceuticals Inc: Honoraria; Kyowa Hakko Kirin Inc: Research Funding; Ono Pharmaceutical Inc: Research Funding; Chugai Pharmaceutical Co: Research Funding; Eli Lilly Inc: Research Funding. Sunami:Ono Pharmaceutical Company, Limited: Research Funding; Takeda Pharmaceutical Company Limited: Research Funding. Ando:Kyowa Hakko Kirin Co., Ltd: Research Funding. Tobinai:Gilead Sciences: Research Funding. Chou:Celgene Corporation: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Ono Pharmaceutical CO., LTD.: Consultancy, Honoraria. Kaneko:Celgene Corporation: Research Funding. Uemura:Celgene Corporation: Employment. Tamakoshi:Celgene Corporation: Employment. Zaki:Celgene Corporation: Employment, Equity Ownership. Doerr:Celgene Corporation: Employment.


2020 ◽  
Vol 36 (S1) ◽  
pp. 36-37
Author(s):  
Pei Wang ◽  
Jing Li ◽  
Yang Yang ◽  
Peng Liu

IntroductionThe treatment of relapsed/refractory multiple myeloma (RRMM), a common hematological malignancy, remains a great challenge in China, partially due to the limited accessibility to novel agents and inadequate public health insurance coverage. Ixazomib, a novel oral proteasome inhibitor (PI), was approved by the China Food and Drug Administration (CFDA) for RRMM in 2018. While bortezomib, a traditional PI, is the recommended agent in the clinical guideline for MM. Here, we compared their costs and effectiveness.MethodsRRMM patients who has received an ixazomib-based regimen (at least 2 cycles) were analyzed. Using a propensity score matching method, we generated a control group of RRMM patients who received the bortezomib-based regimen. The criteria included the number of treatment lines, age, and the revised international staging system stage (R-ISS) which representing the disease stage for myeloma, and paired at a ratio of 1:2 (allowing one control to match multiples). The difference in hospitalization stay, grade 3/4 adverse events rates, overall response rate (ORR), mortality during treatment, and treatment costs was then compared.ResultsNineteen patients received ixazomib and twenty-seven that received bortezomib were included. The ixazomib-group demonstrated a shorter hospital stay (9 days versus 27 days, p < 0.001), lower grade 3–4 adverse events rates (42.1% versus 55.6%, p < 0.001), higher ORR (63.2% versus 48.1%, p = 0.228), and lower mortality rate during treatment (0% versus 7.4%, p = 0.169) than that of bortezomib-group. The ixazomib group had lower total costs (127,620CNY versus 156,424CNY [18,033USD versus 22,103USD], p > 0.05), lower drug costs (98,376CNY versus 103,307CNY [13,901USD versus 14,598USD], p > 0.05), and the lower costs of supportive treatment (5,507CNY versus 14,701 CNY [778USD versus 2,077USD], p < 0.001). Only in terms of self-funded costs, the bortezomib-based regimen was significantly lower (37,127CNY versus 11,521CNY [5,246USD versus 1,628USD], p < 0.001).ConclusionsCompared with the bortezomib-based regimen, the ixazomib-based regimen has better therapeutic effects on MM patients while saving costs. Hence, it may be preferable for use in the treatment of RRMM in China.


Sign in / Sign up

Export Citation Format

Share Document