Elotuzumab in Combination With Lenalidomide and Low-Dose Dexamethasone in Relapsed or Refractory Multiple Myeloma

2012 ◽  
Vol 30 (16) ◽  
pp. 1953-1959 ◽  
Author(s):  
Sagar Lonial ◽  
Ravi Vij ◽  
Jean-Luc Harousseau ◽  
Thierry Facon ◽  
Philippe Moreau ◽  
...  

Purpose This phase I study evaluated elotuzumab, lenalidomide, and dexamethasone in patients with relapsed or refractory multiple myeloma (MM). Patients and Methods Three cohorts were enrolled and treated with elotuzumab (5.0, 10, or 20 mg/kg intravenously) on days 1, 8, 15, and 22 of a 28-day cycle in the first two cycles, and days 1 and 15 of each subsequent cycle; lenalidomide 25 mg orally [PO] on days 1 to 21; and dexamethasone 40 mg PO weekly. Dose-limiting toxicities (DLTs) were assessed during cycle 1 of each cohort, and clinical responses were evaluated during each cycle. The first five patients received up to six cycles of therapy; subsequent patients were treated until disease progression. Results Twenty-nine patients with advanced MM and a median of three prior MM therapies were enrolled; 28 patients were treated, three each in the 5.0-mg/kg and 10-mg/kg cohorts and 22 in the 20-mg/kg cohort. No DLTs were observed up to the maximum proposed dose of 20 mg/kg. The most frequent grade 3 to 4 toxicities were neutropenia (36%) and thrombocytopenia (21%). Two patients experienced a serious infusion reaction (one grade 4 anaphylactic reaction and one grade 3 stridor) during the first treatment cycle. Objective responses were obtained in 82% (23 of 28) of treated patients. After a median of 16.4 months follow-up, the median time to progression was not reached for patients in the 20-mg/kg cohort who were treated until disease progression. Conclusion The combination of elotuzumab, lenalidomide, and low-dose dexamethasone was generally well tolerated and showed encouraging response rates in patients with relapsed or refractory MM.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2782-2782
Author(s):  
Christian Gerecke ◽  
Stefan Knop ◽  
Max S. Topp ◽  
Peter Liebisch ◽  
Christina Vollmuth ◽  
...  

Abstract Background: Lenalidomide (Revlimid®) is an analogue of thalidomide, with immunomodulatory properties and is effective and safe in the treatment of multiple myeloma. We report follow-up data of effects of a combination regimen of lenalidomide, adriamycin and dexamethasone (RAD) on toxicity, prognostic factors, time to progression, and overall survival in patients with multiple myeloma. Patients and Methods: From January 2005 through June 2007 we enrolled 69 patients, 66 patients between 46 and 77 (median age 65) were eligible for evaluation. To be included, patients had to have multiple myeloma with Durie-Salmon stage II or III and considered to have disease progression after 1 to 3 previous anti-myeloma regimens. The feasibility of administering lenalidomide in combination with Doxorubicin and Dexamethasone and the MTD of the combination were determined in the phase I part of the study (Part A). RAD was administered for six 28-day cycle. The MTD was not reached at the highest dose level G5 (R 25 mg d1–21, Adriamycin 9 mg/m2 d1–4 as a 24h infusion, Dex 40 mg d1–4 and 17–20) This dose was used for the phase II part. Results: Forty-eight of 66 patients (73%) achieved an objective response to therapy. Respectively, 10 (15%) of 66 patients achieved a immunofixation-negativ CR, 30 patients (45%) a VGPR and additional 8 patients (12%) a PR. The median follow up was 14,6 (range 7–35) months. Median time to disease progression (TTP) was 45 weeks (95% confidence interval [39,3 to 50,7 weeks] and the one-year survival probability was 88%. 63/66 patients (95%) were evaluable for β2-Microglobulin serum level. Forty-four patients had a level lower than 3,5 mg/l and 19 patients had a higher level than 3,5 mg/l. Subgroup analyses showed a statistical benefit for the group with β2-Microglobulin level < 3,5 mg/l in terms of response (p = 0,002). 37 patients were evaluable for cytogenetic abnormalities with FISH analyses. We found in 15 patients (41%) a Del 13q, four patients (11%) had a t(4;14) and 5 patients (14%) had a Del 17p. There was no significant correlation between response to treatment and the Del 13q (p = 0,40) and the t(4;14) translocation (p = 0,176). Although absolute numbers of patients were low, presence of Del 17p was identified as adverse prognostic factor: 87% without versus 20% with Del 17p responded (p = 0,001). The most common side effects was haematological toxicity with grade 3/4 neutropenia (48%), thrombocytopenia (38%) and anemia (16,6%). Under thrombosis prophylaxis with aspirin 100 mg or enoxaparin 40 mg per day we observed thrombembolic complications in 3 patients (4,5%). Other non haematological side effects were pain (grade 3/4-1 patient), infection (grade 3/4-7 patients), diarrhoea (grade 3/4-1 patient). Neither neurotoxicity nor constitutional symptoms of grade 3/4 was found. Conclusion: In our study, lenalidomide in combination with doxorubicin and dexamethason has shown encouraging activity in heavily pretreated patients with relapsed or refractory multiple myeloma. The combination was well tolerated with a mild toxicity profile. Lower level of β2-Microglobulin and the absence of the deletion 17p had a statistical benefit in terms of response. An update of these data will be presented at the meeting.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1874-1874 ◽  
Author(s):  
Donna E. Reece ◽  
Esther Masih-Khan ◽  
Arooj Khan ◽  
Saima Dean ◽  
Peter Anglin ◽  
...  

Abstract Abstract 1874 Poster Board I-899 Lenalidomide (Revlimid®) and dexamethasone is an effective regimen in relapsed/refractory multiple myeloma (MM) patients (pts), with an overall response rate of 60.6% and median time to progression (TTP) of 13.4 months (Dimopoulos MA, et al, Leukemia 2009 Jul 23 [Epub ahead of print]). Oral cyclophosphamide and prednisone is an older regimen with excellent patient tolerance, and we sought to enhance the efficacy of lenalidomide by adding oral cyclophosphamide and prednisone in this phase I-II trial. The CPR regimen consisted of cyclophosphamide on days 1, 8 and 15; lenalidomide on days 1–21; and prednisone 100 mg every other day in a 28 day cycle. ASA 81 mg/day was given to all pts as DVT prophylaxis. Three dose levels were evaluated using a 3 by 3 dose escalation design. Between 11/2007–07/2009, 31 pts with relapsed/refractory MM who had not previously received lenalidomide were entered onto study. Median age was 61 (40–78) years and 61% were male. Immunoglobulin subtype was IgG in 19 pts (61%), IgA in 8 pts (26%) and light chain only in 4 pts (13%). Median number of prior regimens was 2 (1–5) and 28 pts had undergone previous ASCT, including double transplants in 6 pts. Prior therapy included thalidomide in 9 (29%) and bortezomib in 15 (48%). FISH cytogenetics were available in 13 pts; one had 13q deletion but none had t(4;14) or p53 deletion. At the time of protocol entry, median β 2-microglobulin level was 246 (92–767) nm/L, albumin 39 (34–48) g/L, creatinine 83 (50–126) μmol/L, platelet count 230 (75–337) × 109/L and ANC 2.5 (1.1–6.1) x 109/L. Protocol treatment is summarized in Table 1. Dose limiting toxicity was not observed during cycle 1 at any dose level. Grade 3–4 toxicities included thrombocytopenia in 5 pts (16%) and neutropenia in 9 pts (29%). These were managed with dose reduction and/or growth factor support. Four episodes of febrile neutropenia occurred. Other grade 3–4 non-hematologic toxicities included abdominal pain/bacteremia in 1 pt in cohort 1; hypokalemia in 1 pt in cohort 2; and DVT in 2 pts, dizziness in 2 pts and fatigue in 1 pt in cohort 3. Using the International uniform response criteria (Durie BG, et al, Leukemia 2006; 20:1467–1473), the best response was documented at a median of 6 (1–5) cycles and included the following: dose level 1 (1 CR, 2 PR); dose level 2 (1 VGPR, 2 PR); dose level 3 (5 CR, 9 VGPR, 9 PR, 1 MR and 1 stable disease). At a median follow-up (F/U) of 12 (8–21) months, 20 pts remain on study, 2 have withdrawn and 9 pts have progressed at a median of 9 (4–13) months; only 1 one has died (due to MM). We conclude: 1) the combination of full doses of the agents in CPR can be given in a 28 day cycle with minimal toxicity; 2) the overall response rate (CR + VGPR + PR) in 31 pts to date is 93%; 3) at a median F/U of 1 year, only 9 pts (29%) have progressed; 4) longer follow-up is required to assess the TTP and survival of the CPR regimen. Disclosures: Reece: Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau. Off Label Use: Lenalidomide in combination with drugs other than dexamethasone. Anglin:Celgene: Honoraria. Chen:Celgene: Honoraria, Research Funding. Kukreti:Celgene: Honoraria. Mikhael:Celgene: Honoraria. Trudel:Celgene: Honoraria.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4831-4831
Author(s):  
HyeonSeok Eom ◽  
Yeo-Kyeoung Kim ◽  
Jooseop Chung ◽  
Kihyun Kim ◽  
HyoJeong Kim ◽  
...  

Abstract VTD and MPT chemotherapies have been known to be active regimens in pts with MM. The objective of this study is to examine response and toxicities and to estimate survival of pts with VTD followed by MPT, who are non-transplant candidates with previously untreated MM. Total of 29 pts were enrolled from March, 2006 through August, 2007 and this study is still ongoing. 13 pts were men and 16 pts were women. The median age was 67 years (range, 61–75 years) and median follow up was 5 months (range, 1–16 months). Here 23 pts who completed at least first two cycles of VTD were analyzed. Pts received bortezomib (Velcade®) 1.3 mg/m2 on days 1, 4, 8, 11, thalidomide 100 mg daily, dexamethasone 40 mg on days 1–4 every 3 weeks for a maximum of 6 cycles of treatment, and thereafter melphalan 4 mg/m2 on days 1–7, prednisone 40 mg/m2 days 1–7, thalidomide 100 mg daily every 4 weeks for a maximum of 12 cycles. In these 23 pts, 92% of them showed responses (18% CR, 4% nCR, and 70% PR). 17 pts completed 4 cycles of VTD and all of them showed 100% response rates (CR 35%, nCR 18%, PR 47%). 13 out of 14 who completed 6 cycles of VTD showed responses (CR 50%, nCR 14%, PR 29%, PD 7%). It is too early to see whether improved response rate translates into improved overall survival (OS) and progression free survival (PFS). The median OS and PFS have not been reached yet. 11 pts (37%) stopped protocol therapy because of consent withdrawal (2 pts), death (4 pts), disease progression (2 pts) and severe adverse reaction (3 pts). The causes of death were infection-related in 2 pts who had been in remission. Other 2 pts were related to disease progression. Although peripheral neuropathy affected all of pts, only 20% of the pts were grade 3. The most common side effects of the chemotherapies greater than grade 3 were pneumonia (24%), asthenia (12%), diarrhea (16%), nausea (4%), thrombocytopenia (16%), neutropenia (12%) and anemia (12%). Although VTD followed by MPT chemotherapy in pts with previously untreated MM, who are non-transplant candidates showed high response rates with manageable toxicities, they showed high withdrawal rates from the study which attributed partly to the characteristics of the pts at baseline who were non-transplant candidates because of old age and morbidities, and a few major neuropathies. Follow up data will be presented.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5216-5216
Author(s):  
Jie Jin ◽  
Jiejing Qian ◽  
Haitao Meng ◽  
Wenbin Qian ◽  
Hongyan Tong ◽  
...  

Abstract Purpose: This study assessed the safety, tolerability, and response rate of bortezomib in combination with dexamethasone and/or doxorubibcin in patients with relapsed or refractory multiple myeloma. Methods: Botezomib was administered 1.3mg/m2 on days 1, 4, 8, 11 of a 28-day cycle. Intravenous dexamethasone was administered 20–40mg/d on day 1–2, 4–5, 8–9,11–12, and 5 patients were also given 20mg/d doxorubibcin on day 1–4. Results: Thirty relapsed or refractory myeloma patients (table 1) were enrolled; two patients could not be evaluated because they were lost to follow-up. Of the remaining patients 19 are male, 9 are female. Median age of the patients were 61(49–78). Median number of prior therapies was 4 (1–11). 22 of 28 patients who had been evaluated were assessable for response according to the EBMT criteria (European Group for Blood and Marrow Transplantation criteria). One CR(3.6%), twelve immunofixation-positive CRs[nCR] (42.9%), six PRs(21.4%), and three MR (10.7%) were observed. All patients who had an objective response were alive as of this analysis. Nine patients died during follow-up. Mean response time (time to best response)for the 28 patients was 25 days. Most common Grade ≥3 toxicities (table 2) were peripheral neuropathy(3/28), thrombocytopenia (3/28), rash(one in 28 patients). Six patients(21.4%) suffered herpes after one or two cycles. Table 1. Patents and Disease Characteristics (N=28) No. % Abbreviations: Ig, immunoglobulin; Parameter 61 49–78 Age, years Median Range 19 Sex Male Female 9 Paraprotein type IgG IgA Light-chain only 18 β2..Cmicroglubulin, 3 64.3 10.7 Range 7 25.0 Prior treatments 1262–8691 Median 4 Range 1–11 Table 2. Treatment-emergent adverse events Total No. of patients with event(%) Adverse event 0 Hematologic Neutropenia No. with 3(10.7) grade ≥3 event Thrombocytopenia No. 0 with grade ≥3 event Anemia No. with 6(21.4) grade ≥3 event 5(17.8) Nonhematologic herpes Diarrhea 10(35.7) Fatigue Rash No. with 1(3.6) grade ≥3 event Tachycardia Peripheral 1(3.6) neuropathy Total No. No. 16(57.1) with grade ≥ 3 event 3(10.7) Conclusion: Bortezomib plus dexamethasone given on a 28-day schedule showed encouraging activity with manageable toxicity and represents a promising treatment for multiple myeloma patients.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 620-620 ◽  
Author(s):  
Antonio Palumbo ◽  
Sara Bringhen ◽  
Maide Cavalli ◽  
Roberto Ria ◽  
Massimo Offidani ◽  
...  

Abstract Abstract 620 Background. The combination of bortezomib-melphalan-prednisone (VMP) is a new standard of care for elderly newly diagnosed multiple myeloma. This phase 3 study compared the 4 drug combination bortezomib-melphalan-prednisone-thalidomide followed by maintenance bortezomib-thalidomide (VMPT-VT) with VMP alone. Methods. Patients (N=511) older than 65 years were randomized to receive nine 6-week cycles of VMPT-VT (N=254; induction: bortezomib 1.3 mg/m2, d 1, 4, 8, 11, 22, 25, 29, 32, cycles 1–4, d 1, 8, 22, 29, cycles 5–9; melphalan 9 mg/m2 d 1–4, prednisone 60 mg/m2, d 1–4, thalidomide 50 mg d 1–42; maintenance: bortezomib 1.3 mg/m2 every 14 days and thalidomide 50 mg/day) or VMP (N=257) alone. In March 2007, the protocol was amended: both VMPT-VT and VMP induction schedules were changed to nine 5-week cycles and bortezomib schedule was modified to weekly administration (1.3 mg/m2 d 1,8,15,22, all cycles). The primary end point was progression-free survival (PFS). Results. All patients have been evaluated in intention-to-treat. Patient characteristics were similar in both groups, median age was 71 years. The response rates were superior in the VMPT-VT group with a complete remission (CR) rate of 38% vs 24% (p=0.0008). After a median follow-up of 26.1 months, the 3-year PFS were 55% in patients receiving VMPT-VT and 38% in those receiving VMP (HR 0.65, 95% CI 0.49–0.85, P=0.002, Table). The 3-year time-to-next-therapy were 69% with VMPT-VT and 55% with VMP (HR 0.60, 95% CI 0.42–0.85, P=0.004). The 3-year overall survival was 86% with VMPT-VT and 84% with VMP (HR 0.88, 95% CI 0.53–1.45, P=0.62). The achievement of CR was a strong predictive factor of longer PFS in both groups (P=0.0001): in VMPT-VT arm, 3-year PFS was 66% in patients who obtained CR and 47% in those achieving PR; in VMP arm, it was 70% and 30%, respectively. The PFS benefit of VMPT-VT was seen consistently across different subgroups defined by creatinine clearance, LDH and bortezomib schedule; by contrast, in patients older than 75 years (HR 0.87; 95% CI 0.54–1.43, P=0.59) and in those at increased risk of disease progression, defined as presence of cytogenetic abnormalities [t(4;14) or t(14;16) or del17p] and ISS 3 (HR 1.35; 95% CI 0.45–4.06, P=0.60), VMPT-VT seemed not to add any significant PFS advantage to VMP. Grade 3–4 neutropenia (38% vs. 28%, p=0.02), cardiological events (10% vs. 5%, p=0.04) and thromboembolic events (5% vs. 2%, p=0.08) were more frequent among patients assigned to the VMPT-VT group; treatment-related deaths were 4% with VMPT-VT and 3% with VMP. In both groups, the once-weekly infusion of bortezomib significantly reduced the incidence of severe sensory peripheral neuropathy from 16% to 3% (p<0.0001). One hundred and forty-nine VMPT-VT patients were assessable for maintenance treatment. After a median duration of maintenance of 14.4 months, the PR rate was 90%, including 45% CR. The 1-year landmark analysis of PFS in patients completing the 9 induction cycles, showed a 2-year PFS of 63% in the VMPT-VT group and 40% in the VMP group, demonstrating that maintenance with VT reduced the risk of disease progression of 51% (HR 0.49, 95% CI 0.33–0.72, p=0.0003, Figure). This advantage was less evident in patients older than 75 years (HR 0.97, 95% CI 0.52–1.78, P=0.91) and in those with high-risk of disease progression, defined as presence of cytogenetic abnormalities and ISS 3 (HR 1.31, 95% CI 0.22–7.85, p=0.77). Continuous therapy with VT had favourable safety profile: 3% of patients experienced grade 3–4 hematological toxicity, 5% grade 3–4 peripheral neuropathy and 7% discontinued due to adverse events. Conclusion. In summary the current results indicate that: 1. VMPT-VT prolonged PFS with an unprecedented 3-year PFS of 55% in elderly patients; 2. once-weekly infusion of bortezomib improved safety without affecting outcome; 3. higher dose-intensity regimens seemed to be less effective in frail patients (≥ 75 years) and 4. maintenance therapy with VT further improved PFS with a good safety profile. Disclosures: Palumbo: Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen Cilag: Honoraria, Membership on an entity's Board of Directors or advisory committees. Bringhen:Celgene: Honoraria; Janssen-Cilag: Honoraria. Patriarca:Celgene: Honoraria; Janssen Cilag: Honoraria; Roche: Honoraria; Merck: Membership on an entity's Board of Directors or advisory committees. Guglielmelli:Celgene: Honoraria; Janssen Cilag: Honoraria. Petrucci:Celgene: Honoraria; Janssen Cilag: Honoraria. Musto:Celgene: Honoraria; Janssen Cilag: Honoraria. Boccadoro:Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen Cilag: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding.


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.


Leukemia ◽  
2020 ◽  
Vol 34 (12) ◽  
pp. 3286-3297 ◽  
Author(s):  
David S. Siegel ◽  
Gary J. Schiller ◽  
Christy Samaras ◽  
Michael Sebag ◽  
Jesus Berdeja ◽  
...  

AbstractPatients with multiple myeloma who have relapsed after or become refractory to lenalidomide in early treatment lines represent a clinically important population in need of effective therapies. The safety and efficacy of pomalidomide, low-dose dexamethasone, and daratumumab was evaluated in lenalidomide-pretreated patients with relapsed or refractory multiple myeloma (RRMM) after one to two prior treatment lines in the phase 2 MM-014 study. Patients received pomalidomide 4 mg daily from days 1–21 and dexamethasone 40 mg weekly (28-day cycles). Daratumumab 16 mg/kg was administered per label. Primary endpoint was overall response rate (ORR); secondary endpoints included progression-free survival (PFS) and safety. Per protocol, all patients (N = 112) had received lenalidomide in their most recent prior regimen (75.0% lenalidomide refractory). ORR was 77.7% (76.2% in lenalidomide-refractory patients); median follow-up was 17.2 months. Median PFS was not reached (1-year PFS rate 75.1%). The most common hematologic grade 3/4 treatment-emergent adverse event was neutropenia (62.5%). Grade 3/4 infections were reported in 31.3% of patients, including 13.4% with grade 3/4 pneumonia. These results demonstrate the safety and efficacy of pomalidomide-based therapy as early as second line in patients with RRMM, even immediately after lenalidomide failure, indicating that switching from the immunomodulatory agent class is not necessary.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5737-5737
Author(s):  
Xin Du ◽  
Jie Jin ◽  
Zhen Cai ◽  
Fangping Chen ◽  
Dao-bin Zhou ◽  
...  

Abstract Introduction: In China, the Rd regimen is approved for use in multiple myeloma (MM) pts who have received ≥ 1 prior treatment (Tx). This approval was based on the efficacy and safety of Rd demonstrated in the MM-021 study, a phase 2 multicenter registration trial in which 199 pts were enrolled (Hou, J Hematol Oncol 2013). MM-024 is both an EAP and an extension study that was initiated in order to provide continued Rd regimen for pts enrolled in the MM-021 study. Updated safety and efficacy information from the EAP is presented here. Methods: Eligible pts had participated in MM-021, were still on Tx for ≥ 1 year (yr) and were progression-free. Pts received the same regimen of Rd as in MM-021 (lenalidomide 25 mg/day on days 1–21, and low-dose dexamethasone 40 mg/day on days 1, 8, 15, and 22 of each 28-day cycle). The starting doses of Rd were the same as the last doses received in MM-021 unless dose adjustments were required prior to rollover, as per protocol. Tx was continued until progressive disease (PD) or withdrawal due to toxicities, and pts were followed-up for a maximum of 5 yrs (which included 1 yr spent in the MM-021 study). The primary endpoint was safety, including the incidence of second primary malignancy (SPM). Secondary endpoints included progression-free survival (PFS), time to progression (TTP), and overall survival (OS). Results: By the cutoff date (May 5th, 2014), of the 41 pts who entered the Tx phase of the EAP, 21 are still receiving Tx with a median number of 22 cycles. A total of 20 pts discontinued Tx: 18 discontinued due to PD; 1 due to death (related to lung infection); and 1 was lost to follow-up. Median follow-up was 37.5 months (mos) from initial enrolment in the MM-021 study. Median age was 59 yrs (range 47–74) and 34% of pts were aged > 65 yrs. Most pts (78%) had Durie-Salmon stage III disease at baseline. All pts had received prior Tx for MM, including thalidomide and bortezomib; 7.3% of pts had undergone surgery and 2.4% of pts had received radiation therapy. The median number of prior Tx was 3 (range 1–11); 76% of pts had received prior bortezomib, 68% had received prior thalidomide, and 46% had received both. Median duration of Rd Tx was 21.6 mos (range 13.1–37.5) and 22% of pts have received > 25 mos of the regimen. All efficacy endpoints were measured from the enrolment date of the MM-021 study. Median PFS and median TTP were both 36.0 mos. Median OS has not been reached due to the low number of deaths. Overall, 48.8% of pts had grade 3–4 treatment-emergent adverse events (TEAEs); the most common TEAEs were anemia (51.2%), decreased neutrophil count (48.8%), upper respiratory tract infection (41.5%), neutropenia (31.7%), cough (24.4%), diarrhea (22.0%), and pyrexia (22.0%). Grade ≥ 3 TEAEs included neutropenia (14.6%), anemia (4.9%), thrombocytopenia (2.4%), and pneumonia (7.3%). No TEAEs led to Tx discontinuation; however, TEAEs led to lenalidomide dose reduction (14.6%), interruption (39%), or both (14.6%). Updated efficacy, survival, and SPM analysis will be presented at the meeting. Conclusions: After 37.5 mos of follow-up, median PFS was 36 mos, compared with the median PFS of 7.5 mos in the MM-021 final analyses (cutoff date January 4th, 2013). This indicates that long-term use of Rd is well tolerated and continues to be an effective Tx in Chinese pts with RRMM who have received multiple prior Tx. Disclosures DeMarco: Celgene Corporation: Employment. Zhang:Celgene Corporation: Employment. Mei:Celgene Corporation: Employment. Hou:Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene Corporation: Honoraria, Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1857-1857 ◽  
Author(s):  
Cristina Gasparetto ◽  
Michael Green ◽  
Anandgopal Srinivasan ◽  
Yubin Kang ◽  
David A. Rizzieri ◽  
...  

Abstract Background Bendamustine, a bifunctional mechlorethamine derivative with alkylating properties and pomalidomide, an IMiD® immunomodulatory agent, have both demonstrated efficacy as single agents or in combination with dexamethasone in relapsed/refractory multiple myeloma(RRMM). Bendamustine in combination with lenalidomide, thalidomide, and bortezomib have had high response rates and good tolerability. We combined bendamustine and pomalidomide with dexamethasone (Ben-Pom-d) and hypothesized that this regimen would be highly effective in patients with RRMM. Dose-escalation started with 120mg/m2 bendamustine/3mg pomalidomide [or 4mg in the cohort 2]/40 mg dexamethasone using a standard 3+3 schema based on dose-limiting toxicities (DLTs) occurring in cycle 1. The MTD was 120mg/m2 bendamustine/3mg pomalidomide/40 mg dexamethasone. Here, we report our findings to date from the phase I/II trial of Ben-Pom-d in patients with RRMM (NCT01754402). Methods The primary objective of the phase I portion was to determine the MTD. Data for overall response, progression free survival, and overall survival, includes all patients treated on the phase I and II portions of the study. All patients had to be refractory to prior lenalidomide, and must have relapsed or were refractory to their most recent therapy. Patients had to be pomalidomide naïve. Treatment consisted of oral pomalidomide once daily on days 1-21, intravenous (IV) bendamustine given over 30 minutes on day 1 and dexamethasone 40 mg on days 1, 8, 15, and 22 of a 28-day cycle. Adverse events (AEs) were graded by NCI-CTCAE v4. Response was assessed by the modified International Uniform Response Criteria. Results A total of 9 patients were enrolled in the phase I portion. The MTD was the starting dose level (bendamustine 120 mg/m2, pomalidomide 3mg, dexamethasone 40 mg). In Phase II we enrolled an additional 16 patients resulting in a total study population of 25 patients evaluable for toxicity and 22 for efficacy, with 6 still receiving treatment. The median age was 65 years (range 43-81), 46% were male. The median number of prior regimens was 3 (range 2-6), median time from diagnosis is 3.9 years (range 1.1-9.10 years), 88% of patients had a prior stem cell transplant, 100% had prior bortezomib, 20% had prior carfilzomib and all were lenalidomide-refractory. Fifteen patients had high risk cytogenetic, including 8 patients with del17. Patients received a median of 6 cycles of therapy (range 1-18 cycles). Best response assessments in 22 evaluable patients for efficacy, showed 5 (23%) VGPR, 12 (55%) PR, 3 (14%) MR, and 2 (9%) SD, for an ORR of 77% and a ≥MR rate of 91%. The median follow-up of survivors is 10 months (range: 2-19+ months). Median PFS and OS were 4.5 months (range 1-15+ months) and 9.5 months (range 2-19+ months), respectively, for the entire cohort with 13 of 22 still alive in follow-up. The Median PFS for patient with del 17 is 5.5 months (range 2-15 months) with >MR rate of 88%. During the first cycle, 3 patients of all 25 evaluable enrolled experienced a DLT at the different doses, including 1 nausea/vomiting [cohort 1], and 2 with rash and fever in cohort 2. The therapy was tolerated well, but toxicities reported at any point while on therapy included 32% grade 4 neutropenia, 16% grade 4 thrombocytopenia, and half the patients requiring delay of subsequent cycles due to cytopenias and 17 of 22 (77%) had a dose reduction of pomalidomide per protocol guidelines at some point in the continuation cycles. The major non-hematologic Grade ≥3 drug-related AEs that occurred included febrile neutropenia in 12%, grade 3 mucositis in 8%, grade 3 pneumonia 16% and grade 4%, and grade 4 sepsis 4%. Conclusions The Ben-Pom-d regimen is a well-tolerated regimen and achieves a high response rate (ORR of 77%; ≥MR rate of 91%) in a heavily pre-treated Lenalidomide-refractory population with prior bortezomib exposure. Therapy is ongoing for many and longer follow-up is needed to better assess the true durability of this approach. Disclosures Gasparetto: Onyx: Honoraria, Other: Advisory Board; Millennium/takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; teva: Other: spouse-ad board and speaker bureau. Off Label Use: Bendamustine-pomalidomide-dexa for treatment of relapsed myeloma. Rizzieri:Teva: Other: ad board, Speakers Bureau; Celgene: Other: ad board, Speakers Bureau. Rao:novartis: Other: ad board; amgen: Other: ad board; Boehringer-Ingelheim: Other: Advisory Board. Tuchman:celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; Millennium/takeda: Honoraria, Research Funding, Speakers Bureau.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3470-3470 ◽  
Author(s):  
Martin Kropff ◽  
Martin Vogel ◽  
Andrea Kreter ◽  
Rudolf Schlag ◽  
Lothar Müller ◽  
...  

Abstract Background Multiple myeloma (MM) typically follows a relapsing course and many patients require multiple lines of therapy. The combination of bortezomib and dexamethasone (VD), alone or with cyclophosphamide (VCD) has shown good efficacy and tolerability in patients with relapsed or refractory (rel/ref) MM in non-randomized trials (Mikhael Br J Haematol 2009, Kropff Br J Haematol 2007). This phase III, randomized, controlled study (NCT00813150) aimed to evaluate VCD vs VD in patients with rel/ref MM. Methods Adult patients with rel/ref MM (1st–3rd relapse) with Karnofsky performance status ≥60% were eligible for inclusion; patients with grade ≥2 peripheral neuropathy (PN) were excluded. Patients were randomized 1:1 to receive VD (up to 8 x 3-week cycles of intravenous bortezomib 1.3 mg/m2 on days 1, 4, 8, and 11 plus oral dexamethasone 20 mg on the day of bortezomib administration and the following day) or VCD (as VD with the addition of oral cyclophosphamide 50 mg daily). The study was designed with 80% power to detect a difference of 2.8 months in the primary endpoint of time to disease progression (TTP) at the 5% significance level (two-sided); a sample size of 270 patients was required. Secondary endpoints included overall survival (OS), response rate (RR), and safety. Disease progression was assessed per IMWG guidelines and adverse events (AEs) were assessed using NCI-CTCAE v3.0. TTP and OS were calculated using Kaplan-Meier methodology and compared using two-sided log-rank test. Results In total, 96 patients were randomized; 93 began study medication and were included in the safety population, and 90 received at least one dose of study drugs and one efficacy assessment and were included in the intention to treat (ITT) population. The planned sample size was not reached due to lack of recruitment and the study was stopped prematurely as a result of a joint decision of sponsor and coordinating investigator. The VD and VCD arms were well balanced in terms of baseline characteristics. Median age was 71 years (range 30–85) with most patients (97%) suffering from relapse of myeloma disease (3% refractory disease). One, 2, and 3 prior lines of therapy were reported by 57%, 30%, and 9% of patients, respectively; 14% had received prior bortezomib. In total, 19%, 24%, and 29% of patients were ISS disease stage I, II, and III, respectively. Patients in both VD and VCD arms received a median of 5 cycles of therapy. Reasons for premature termination of treatment included AEs (35% and 32% of patients in the VD and VCD arms, respectively) and progressive disease (2% in the VD arm only). After a median follow-up of 24 months, median TTP was 12.6 months in the VD arm vs 9.9 months in the VCD arm (p=0.192); the corresponding hazard ratio (HR) was in favor of the VD arm (HR 0.71, 95% CI 0.43–1.19), although this was not statistically significant (p=0.196). Median OS could not be determined in the VD arm vs 42 months in the VCD arm. The resulting Kaplan-Meier plots were superimposable with a HR of 0.85 (95% CI, 0.41–1.73; p=0.645). In total, 32 (74%) and 33 (70%) patients in the VD and VCD arms achieved ≥PR; 23 (54%) and 22 (47%) patients achieved ≥VGPR. The most common treatment-emergent non-hematologic AEs included PN (28% vs 47% in the VD vs VCD arms), fatigue (28% vs 40%), diarrhea (24% vs 36%), and constipation (22% vs 28%). Grade ≥3 PN occurred in 7 (15%) patients in the VCD arm vs 2 (4%) in the VD arm. Higher rates of grade ≥3 leukopenia (13% vs 0%) and grade ≥3 thrombocytopenia (34% vs 24%) were observed in the VCD arm; however, rates of clinically relevant infections were comparable (grade ≥3 infections 9 [19%] in the VCD arm vs 8 [17%] in the VD arm) and no clinically relevant bleeding events occurred. Conclusions These data indicate that both treatment regimens were similar in efficacy and safety. However, due to limited enrollment, the study is underpowered to draw firm conclusions. Further studies will be required to address the potentially enhanced neurotoxicity of VCD compared with VD. The dosing of cyclophosphamide also warrants further investigation, as other studies suggest that a dose intensified regimen may result in higher CR rates. Disclosures Kropff: Janssen: Consultancy, Speakers Bureau; Celgene: Consultancy, Speakers Bureau; Mundipharma: Consultancy, Speakers Bureau; Amgen: Consultancy, Speakers Bureau; Onyx: Consultancy, Speakers Bureau; BMS: Consultancy, Speakers Bureau. Vogel:Janssen: Employment. Knauf:Janssen: Consultancy, Honoraria, Research Funding, Speakers Bureau; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; Mundipharma: Consultancy, Honoraria, Research Funding, Speakers Bureau; Roche: Consultancy, Honoraria, Research Funding, Speakers Bureau. Fiechtner:Onkovis: Consultancy. Berdel:Janssen: Honoraria.


Sign in / Sign up

Export Citation Format

Share Document