Thalidomide in Relapsed or Refractory Patients with Multiple Myeloma: Monotherapy or Combination Therapy? A Report from Systematic Reviews.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5125-5125 ◽  
Author(s):  
Axel Glasmacher ◽  
Corinna Hahn ◽  
Florian Hoffmann ◽  
Kerstin Furkert ◽  
Marie von Lilienfeld-Toal ◽  
...  

Abstract The activity of thalidomide monotherapy in relapsed or refractory disease is widely accepted but many haematologists have observed better response rates with combination therapies. This communication aims to give an overview on the efficacy of these alternatives. Methods: Our group has performed two systematic reviews (thalidomide monotherapy: Glasmacher et al., Brit. J. Haematol., 2005; 129 suppl 1: 24; thalidomide and dexamethasone combination therapy: submitted to ASH 2005). Furthermore a search of trials combining thalidomide, cyclophosphamide and dexamethasone (with or without further drugs) was performed. Only trials that included patients with relapsed or refractory multiple myeloma could be evaluated. Response was defined as a reduction of the monoclonal component by more than 50% in the absence of any sign of disease progression. Proportions and 95% confidence intervals were reported (Confidence Interval Analysis, Version 2.1.1, Southampton, UK, 2000). Results: Thalidomide monotherapy was administered in doses between 50–600 mg/d (median doses). Thalidomide in combination with dexamethasone was given in doses of 100–400 mg/d (median doses), dexamethasone was given 40 mg/d d1–4 every 3–4 weeks in 7 of 8 trials. Thalidomide dosage and combination therapy of trials adding Cy to thalidomide and dexamethasone are shown in Table 1. The response rates of the three different thalidomide applications are demonstrated Table 2. Conclusion: This compilation of data from phase II trials cannot replace a randomized trial and may be biased. With this limitation it seems that thalidomide combined with dexamethasone (response rate 51%) is more effective than monotherapy (response rate 29%) as the 95% confidence intervals for the response rates do not overlap. Whether a combination of thalidomide, dexamethasone and cyclophosphamide is more effective than thalidomide and dexamethasone alone cannot be decided from the current data. Table 1: Studies with thalidomide and combination chemotherapy Study Intervention No. of pts. Response (95%CI) Abb.: Cy, cyclophosphamide; Eto, etoposide; T, thalidomide; Dex, dexamethasone; Ida, idarubicin. Values are mg/m2 per course for Cy, Eto, Ida; mg per course for Dex, mg/d for T. Moehler et al. 2003 Cy 1600, Eto 160, T 400, Dex 160 119 55% (46–65) Kropff et al., 2003 Cy 1800, T 400, Dex 240 60 70% (57–81) Dimopoulos et al. 2003 Cy 1500, T 400, Dex 160 43 67% (51–81) Gracia-Sanz et al. 2004 Cy 50 daily, T 800, Dex 160 71 57% (45–67) Glasmacher et al. 2005 Cy 800, Ida 40, T 400, Dex 320 39 57% (41–73) Table 2: Response of different thalidomide combinations Intervention No. of trials No. of pts. Response (95%CI) Abb.: see Table 1 T monotherapy 42 1629 29% (27–32) T + Dex 8 283 51% (45–57) T + Cy + Dex 5 332 60% (55–65)

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4851-4851 ◽  
Author(s):  
Irena Hrusovsky ◽  
Hans-Heinrich Heidtmann

Bortezomib(Bo) was approved for therapy of relapsed Multiple Myeloma (MM) in 2003-for use in Germany 2004. There is growing evidence that combination of Bo with conventional chemotharapy agents could improve MM patients’ (pts) outcome(P.Richardson ASCO 2004). Bendamustin(Ben) is widely used for MM chemotherapy in Germany. In weekly low dose regimen it is well tolerated and highly effective in therapy of low grade lymphoma in elderly (K.Bremer ASCO2003, Abstract 2410). Ben has no cross resistances with other cytostatic drugs used in MM therapy, in low dose regimen it is well tolerated, has low bone marrow toxicity and no renal toxicity. We reported results of the combination therapy of the first 17 pts 2 years ago. Because of very good tolerability and high response rates we established the combination in our institution as a salvage-therapy for pts with relapsed MM after at least 2 previous chemotherapies. Till now we treated 40 pts - all white European-21 women, 19 men- median age 66 years (range 51–86). All pts had relapsed MM with clinical symptoms such as anemia, bone pain, progressive bone disease, several of the pts had renal failure (1 on hemodialysis). Previous therapies - median 4-(range 2–10)- included Melphalan, Dexamethason, VAD and sim. combinations, Cyclophosphamide, Bendamustin, Thalidomide, Bortezomib mono and in 3 pts tandem HD Melphalan. Therapy-regimen: Bortezomib 1–1,3 m/sqm d 1,4,8,11, Bendamustin 60mg/sqm d 1,8, Dexamethason 3x8mmg p.o. d 1–3 and 8–10 if tolerated, Ondansetron 8 mg i.v. d 1,4,8,11- q3w until best response- median number of cycles 4 (range 2–6). Early responses with symptom relief at begin of second cycle were frequently observed. Results (outcome according to SWOG criteria): ORR 85%, very good PR 25%(normal electrophoresis, normal level of free light chains in serum and urine), PR 47,5%, MR 12,5%. Remission duration in pts with at least PR- 8 Months (range 2–26 months)-19 pts are still alive. Toxicity: comprised fatigue and mostly mild thrombocytopenia without bleeding, reversible within 1 week. 8 pts had neuropathy and required symptomatic therapy with Gabapentine- most of them pretreated with Thalidomide. 9 pts had herpes zoster before aciclovir 3x400 mg daily was administered as prophylactic therapy. Conclusion: The therapy with combination Bortezomib- Bendamustin is highly effective, safe and well tolerated. The therapy is feasible in out-patient setting. The response rates suggest that the drugs act at least additive. We think therefore that further studies are warranted.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4076-4076
Author(s):  
Sallie Stradwick ◽  
Nick Freemantle ◽  
John Snowden ◽  
Felipe Rodrigues ◽  
Nic Brereton

Abstract Abstract 4076 OBJECTIVES: To evaluate the comparative effectiveness of lenalidomide (25 mg) plus dexamethasone (40 mg) (LEN/dex) for the treatment of relapsed/refractory multiple myeloma (RRMM) compared to thalidomide and bendamustine. Primary outcome of interest was time to progression (TTP). Secondary outcomes of interest were overall response rates (ORR) and overall survival (OS). METHODS: A comprehensive systematic literature review was conducted to identify any randomised controlled trials (RCTs) investigating the clinical efficacy of specified therapies for the treatment of RRMM. Specified therapies included lenalidomide, thalidomide and bendamustine. Of these therapies, only LEN/dex combination therapy is approved in RRMM but bendamustine and thalidomide (monotherapy or in combination with dexamethasone) have also shown activity in the treatment of myeloma. Electronic databases were searched from March 2002 to 2012 (language unrestricted. Randomized clinical trials were independently evaluated against predetermined criteria for inclusion and determined to be eligible for the meta-analysis prior to any outcome assessment. Fixed effects and random effects mixed-treatment comparisons (MTC) were carried out, adopting the methods described by Lu and Ades1. MTCs estimate the comparative effectiveness of multiple treatments using an evidence base of trials that individually do not compare all treatment options. Results for TTP were reported as hazard ratios (HR) and 95% confidence intervals and an associated probability of best treatment. Results for binary variables (ORR, OS) were reported as odds ratios (OR) and 95% confidence intervals. RESULTS: Sixteen original RCTs met the initial inclusion criteria; twelve (N = 3,590), nine (N = 3,350) and six (N = 2,295) of which were able to be connected to form a network of evidence which provided the heterogeneous trial base for MTC analysis of ORR, OS, and TTP comparison, respectively. Two trials directly investigated the efficacy of LEN/dex (N = 353) and three trials assessed the efficacy of thalidomide monotherapy (N = 785). No RCTs were identified that investigated the efficacy of bendamustine or thalidomide/dexamethasone combination therapy in RRMM. There were too few studies (investigating treatment arms of interest) relative to the number of trials in the network to formally estimate or test for heterogeneity. Therefore a fixed effects network meta-analysis was used instead of a random effects network meta-analysis. Comparability of trial populations was assessed and discussed in detail since these could not be adjusted for in the statistical model. Age, sex, baseline disease characteristics, time since diagnosis and medical history were similar between study populations. TTP analysis was statistically significant and favoured LEN/dex over thalidomide monotherapy: HR = 2.34 [1.31, 4.17]. The associated probability of LEN/dex being the best treatment within the evidence network is 97.9%. Secondary outcomes analysis were also statistically significant in favour of LEN/dex over thalidomide monotherapy (ORR: OR = 10.48 [4.75, 22.81]; OS: OR = 1.43 [1.12, 1.84]). CONCLUSIONS: Results demonstrated statistically significant superiority of lenalidomide plus dexamethasone therapy versus thalidomide monotherapy for the treatment of RRMM. No analyses were possible versus bendamustine or thalidomide/dexamethasone combination therapy due to the lack of RCTs investigating the efficacy of these therapeutic regimens in RRMM. Disclosures: Stradwick: Celgene International: Consultancy. Freemantle:BresMed: Consultancy. Off Label Use: Thalidomide monotherapy activity in the treatment of myeloma. Brereton:Celgene International: Consultancy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3301-3301
Author(s):  
Natalia Paola Schutz ◽  
Paola Ochoa ◽  
Patricio Duarte ◽  
Guillermina Remaggi ◽  
Sebastian Yantorno ◽  
...  

Abstract Introduction: There are scarce data regarding treatment outcomes and toxicity in Latin American countries. Argentina is the second largest country in the region and the fourth most populated one. National Guidelines from the Argentinean Society of Hematology (SAH) recommends the use of bortezomib based triplets for induction treatment in transplant eligible newly diagnosed Multiple Myeloma patients. Objective: To compare response rates and adverse events after induction treatment with Cyclophosphamide Bortezomib and Dexamethasone (CyBorD) or Bortezomib Thalidomide and Dexamethasone (VTD) outside of clinical trials in a Latin American country. Methods: Retrospective multicentric cohort study. All centers participating in the Argentinean Multiple Myeloma Study Group (GAMM) were invited to participate in the study. Eligible patients were 75 years of age or younger, with a diagnosis of Multiple Myeloma according to the IMWG 2014 criteria, transplant eligible, treated with at least one cycle of CyBorD or VTD as induction therapy in the time period from December 2012 until December 2017. Main exclusion criteria were amyloidosis, plasma cell leukemia and previous neuropathy. Patients were identified from local registries at each center and included consecutively in the study database. Epidemiological and clinical data were obtained from medical records and collected in a standardized clinical report form. Patients were followed from diagnosis until death or lost to follow up. Response was evaluated according to IMWG Response Criteria 2016. Adverse events were graded by CTCAE 4.3. Comparisons of response rates were performed using a Chi2 test and differences in rates were expressed as proportions with 95% confidence intervals (CI). Crude odds ratios (OR) and OR adjusted by potential confounders were calculated using a logistic regression model. Kaplan Meier method was used to estimate progression free survival (PFS) and overall survival (OS). Stata 13 software was used. Results: A total of 322 patients from 15 centers in Argentina were included in the study. The median age at diagnosis was 57 years (range 26-74), 52% (167) of the patients were male, 18% (58) had renal failure, 28% (85) ISS 3 , 7% (22) extramedullary disease, and 14% (46) high risk cytogenetics. Median time of follow up was 34 months (IQR 21-58). CyBorD was the most common treatment, indicated as induction therapy in 74% (238) of the cases. The characteristics of the patients were similar in both groups except age and LDH levels. The median number of cycles was 5 (range 1-12). Bortezomib was administered once per week in 85% (272) of the patients and subcutaneously in 86% (276) with no differences between both treatment arms. The median cumulative cyclophosphamide dose per month was 1.5 g (IQR 1.5-2.4) and thalidomide dose per day was 100 mg. In the VTD arm, 72,62% (61) of the patients achieved at least very good partial response (VGPR) vs 53.36% (127) with CyBorD [OR of 2.31 (CI 1.35 - 3.99) p=0.002]. The difference in VGPR was 19.26% (CI 15 - 24). Complete response rate (CR) was 35.92% in patients treated with VTD vs 22.55% with CyBorD [OR of 1.87 (CI 1.04 - 3.35) p=0.03). The difference in CR was 13,37% (CI 9.6 -17.53). There was no difference in overall response rate (ORR) with 94.05% vs 91.18% (p=0.406). Adverse events were more common with VTD (69.05% vs 55.46% p=0.030), especially neuropathy grade 3 - 4 (7.14% vs 1.26% p=0.005) and thrombosis (13.10 % vs 3.36 % p=0.001). Deep venous thrombosis prophylaxis was inadequate in 20.24% of the patients. Hematologic adverse events were more common with CyBorD, especially thrombocytopenia (5.95% vs 16.39% p=0.017). Autologous stem cell transplantation (ASCT) was performed in 78% (249) of patients. There was 5% (17) stem cell mobilization failure, all in the CyBorD arm. Response rates after ASCT with VTD and CyBorD induction treatment were: 76.19 vs 73.11% VGPR (p=0.580) and 48.53% vs 40% CR (p=0.20). Maintenance treatment was indicated in 67.86% (57) and 65.13% (155) patients respectively (p=0.650). The PFS at 24 months was 83% (CI 71-90) with VTD vs 72% (CI 66-78) [(HR 0.92 (CI 0.59 - 1.42) p 0.715] and OS 96% (CI 87-99) vs 91% (86-94) respectively [(HR 1.2 (CI 0.62 - 2.32) p 0.587]. Conclusions: VTD has better CR and VGPR compared to CyBorD. Nevertheless, CyBorD continues to be the preferred induction regimen in Argentina based on safety profile. The optimal number of induction treatment cycles remains to be determined. Disclosures Schutz: Takeda: Honoraria, Research Funding; Sanofi Aventis: Research Funding; Roche: Research Funding; Glaxo: Research Funding; Janssen: Honoraria, Research Funding; Varifarma: Honoraria. Shanley:Brystol Myers Squibb: Consultancy, Honoraria; Takeda: Consultancy, Honoraria. Fantl:Janssen: Consultancy, Honoraria, Research Funding; Varifarma/Amgen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria, Research Funding; Sanofi: Research Funding; Roche: Research Funding; Tecnofarma: Honoraria; BMS: Consultancy, Honoraria; Glaxo: Research Funding.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4948-4948
Author(s):  
Yongqing Zhang ◽  
Guangxun Gao ◽  
Jishi Wang ◽  
Xiequn Chen

Abstract Abstract 4948 Objective To investigate the efficacy and safety of PAD (bortezomib, doxorubicin and dexamethasone) combination therapy for Chinese relapsed or refractory multiple myeloma (MM). Methods 31 patients with relapsed or refractory MM received two to eight 21-days cycles of PAD: comprising an intravenous bolus of bortezomib 1.3 mg/m2 (P1,N=13) or 1.0 mg/m2 (P2,N=18) on days 1, 4, 8, and 11, doxorubicin 10mg per day on days 1 to 4, along with dexamethasone 40mg on days 1-4. Response to PAD was evaluated according to International Myeloma Working Group Criteria (IMWG 2006), toxicity was graded according to NCI CTCAE v3.0. Results 25 patients (80.6%) achieved at least a partial response (PR), including complete response (CR) in 9 patients (29%), very good partial response (VGPR) in 7 patients (22.6%), PR in 9 patients (29%) and stable disease (SD) in 4 patients(12.9%), progression of disease (PD) in 2 patients (6.5%); median time to progression was 9.2 months, the median courses to achieve at least PR was 1.6(1-3) cycles, all of 7 patients with extramedullary plasmacytoma achieved at least PR after the first cycle of PAD, extramedullary plasmacytoma disappeared with 1-2 cycles of PAD. The efficacy was independent of traditional prognostic factors such asβ2-MG, Albumin,LDH and Hemoglobin which have previously influenced response to traditional chemotherapy. 1.5 year OS (Overall survival)of CR+VGPR group and PR group were 87.5% vs 46.7% (P=0.09). ≥PR response rate (CR +VGPR +PR) of P1AD and P2AD were 84.6% VS 77.8% (P= 0.501), CR+VGPR rate of P1AD and P2AD were 53.8% vs 50.0% (P=0.561 ). 1 year PFS(Progession-free survival) of P1AD and P2AD were 61.2% vs 55.6%(P=0.638), there were not difference between P1AD and P2AD in response rate(P= 0.501) and 1 year OS (P=0.872). Adverse events included thrombocytopenia in 15 patients ( 48.4% ), leukopenia in 8 patients(25.8%), peripheral neuropathy in 6 patients (19.4% ), varicella herpes zoster in 7 patients (22.6%), fatigue in 11 patients (35.5%) and diarrhea in 5 patients (16.1%), Thrombocytopenia and peripheral neuropathy of P1AD and P2AD were 46.2% vs 11.1%( P=0.037)and 53.8% vs 11.1%(P= 0.014).Common adverse reactions could be controlled with routine supportive treatmemt, one patient (3.2% ) died from respiratory failure during his fifth P1>AD. Conclusions PAD should be considered an appropriate treatment for Chinese relapsed or refractory MM, especially for MM with extramedullary plasmacytoma, its efficacy were independent of traditional prognosis factors, bortezomib dose reduction may reduce toxicities of PAD while retaining the efficacy. Disclosures No relevant conflicts of interest to declare.


2008 ◽  
Vol 81 (4) ◽  
pp. 247-252 ◽  
Author(s):  
Marie von Lilienfeld-Toal ◽  
Corinna Hahn-Ast ◽  
Kerstin Furkert ◽  
Florian Hoffmann ◽  
Ralph Naumann ◽  
...  

2006 ◽  
Vol 24 (3) ◽  
pp. 431-436 ◽  
Author(s):  
S. Vincent Rajkumar ◽  
Emily Blood ◽  
David Vesole ◽  
Rafael Fonseca ◽  
Philip R. Greipp

Purpose To determine if thalidomide plus dexamethasone yields superior response rates compared with dexamethasone alone as induction therapy for newly diagnosed multiple myeloma. Patients and Methods Patients were randomly assigned to receive thalidomide plus dexamethasone or dexamethasone alone. Patients in arm A received thalidomide 200 mg orally for 4 weeks; dexamethasone was administered at a dose of 40 mg orally on days 1 to 4, 9 to 12, and 17 to 20. Cycles were repeated every 4 weeks. Patients in arm B received dexamethasone alone at the same schedule as in arm A. Results Two hundred seven patients were enrolled: 103 were randomly assigned to thalidomide plus dexamethasone and 104 were randomly assigned to dexamethasone alone; eight patients were ineligible. The response rate with thalidomide plus dexamethasone was significantly higher than with dexamethasone alone (63% v 41%, respectively; P = .0017). The response rate allowing for use of serum monoclonal protein levels when a measurable urine monoclonal protein was unavailable at follow-up was 72% v 50%, respectively. The incidence rates of grade 3 or higher deep vein thrombosis (DVT), rash, bradycardia, neuropathy, and any grade 4 to 5 toxicity in the first 4 months were significantly higher with thalidomide plus dexamethasone compared with dexamethasone alone (45% v 21%, respectively; P < .001). DVT was more frequent in arm A than in arm B (17% v 3%); grade 3 or higher peripheral neuropathy was also more frequent (7% v 4%, respectively). Conclusion Thalidomide plus dexamethasone demonstrates significantly superior response rates in newly diagnosed myeloma compared with dexamethasone alone. However, this must be balanced against the greater toxicity seen with the combination.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3552-3552 ◽  
Author(s):  
Edward Stadtmauer ◽  
Donna Weber ◽  
M. Dimopolous ◽  
Andrew Belch ◽  
Michel Attal ◽  
...  

Abstract Background: High-dose dexamethasone (Dex) remains a standard therapy for relapsed or refractory multiple myeloma (MM). Lenalidomide is a novel, orally administered, immunomodulatory drug (IMiD) that has single-agent activity against MM and additive effects when combined with Dex. At the interim analysis, lenalidomide/dexamethasone achieved a significant benefit over dexamethasone, providing a longer median time to progression (TTP), higher response rates, and higher CR rates. Aim: This prospective subgroup analysis of MM-009/010 was performed to determine the potential benefit of starting lenalidomide/dexamethasone at first relapse by analyzing outcomes with lenalidomide/dexamethasone versus dexamethasone among patients who had received only 1 versus > 1 prior line of therapy. Methods: Patients who had received at least 1 prior treatment and were not refractory to dexamethasone were randomized to either receive oral lenalidomide (25 mg daily for 3 weeks every 4 weeks) plus Dex (40 mg on Days 1–4, 9–12, 17–20 every 4 weeks for 4 months, then 40 mg on Days 1–4 every cycle thereafter) or placebo plus Dex. The European Blood and Marrow Transplantation criteria were used to evaluate response. Randomization was stratified at entry by number of prior therapies (1 versus > 1). Results: Among the 248 patients who had received only 1 prior therapy, those receiving second-line lenalidomide/dexamethasone had a significantly longer median TTP (71 vs. 20 wks) and a higher response rate (complete response [CR] + partial response [PR]; 65% vs. 26%) versus those receiving second-line dexamethasone. Among the 456 patients who had received > 1 prior line of therapy, the median TTP (41 vs. 20 wks), response rate (58% vs. 20%) were higher with lenalidomide/dexamethasone compared with dexamethasone. Comparing patients who received lenalidomide/dexamethasone as second-line versus as later salvage therapy, the median TTP appeared longer and the response rates higher in patients who received lenalidomide/dexamethasone earlier, although TTP and response rates were also significantly better with lenalidomide/dexamethasone than with dexamethasone in patients who received lenalidomide/dexamethasone later. Differences in the groups included prior stem cell transplant (66% vs. 54%), thalidomide (12.5% vs. 53.2%), and bortezomib (0.4% vs. 11.6%) in the second-line versus later salvage therapy groups. No difference was observed in grade ¾ adverse events or survival with a median follow-up of 16.8 months. Conclusions: Lenalidomide/dexamethasone provided higher response rates and improved TTP compared with dexamethasone at first relapse and beyond. Response to lenalidomide/dexamethasone was superior to that to dexamethasone regardless of the type of prior therapy. TTP and response rates appeared more favorable when lenalidomide/dexamethasone was administered earlier at first relapse compared with its use as later salvage therapy. These data support the use of lenalidomide/dexamethasone for patients as second-line therapy.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3210-3210 ◽  
Author(s):  
Shivlal Pandey ◽  
S. Vincent Rajkumar ◽  
Prashant Kapoor ◽  
Rhett P Ketterling ◽  
Martha Q Lacy ◽  
...  

Abstract Background Multiple myeloma (MM) is a heterogeneous disease with variable responses to different therapeutic regimens and wide spectrum of survival. Much of the heterogeneity in the outcomes appear to be related to the underlying primary genetic abnormality, which in the majority of patients consist of either translocations involving the heavy chain region on chromosome 14 (IgH translocation) or trisomies of odd numbered chromosomes. We hypothesized that the response to lenalidomide (Len) therapy would vary significantly based on the underlying molecular subtype of myeloma. Methods We examined a cohort of 518 patients with available FISH results, who had been exposed to Len-based regimens. Medical records were reviewed and data regarding the best response and time to next therapy following treatment with Len-based regimen was obtained. Data from the first use of Len was collected. Len was given in combination with dexamethasone (Dex) with or without alkylators in combination. Patients who received a combination of IMiD and bortezomib as their first exposure to Len were excluded. Patients were grouped according to whether FISH showed a trisomy or an IgH translocation. Results The median age was 62 (28-91), and the median estimated follow up from diagnosis was 52 months (95% CI; 50, 54) with 359 (69%) alive at the time of analysis. An IgH translocation was seen in 129 (30%) of patients and a trisomy in 268 (62%) of patients. IgH translocations included t(11;14) in 92 (18%), t(4;14) in 45 (9%), t(14;16) in 21 (4%); 34 (8%) had both translocations and trisomies. For the current analysis, we included only patients with either a translocation or trisomy (n=397) excluding those with neither or both of the abnormalities. The median time to start of Len from diagnosis was 0 months (range, 0-64). A PR or better was seen in 80% of patients with trisomy compared with 63% of the patients with translocation (p<0.001); and the response rate was similar among the different translocation types. The median TTNT was 28 months among trisomy pts compared with 17 months for translocated patients (p<0.001, figure). The median TTNT was similar across the different types of translocations (Figure). Among this group, 134 patients proceeded to an autologous SCT after Len induction. Among these patients, no difference was seen in terms of TTNT (29 months for patients with translocation vs. 28 months for those with trisomy (p=0.8). Finally, the TTNT was no different if Len was used with Dex or as part of an alkylator combination. Conclusion The current data supports the hypothesis that the underlying primary genetic abnormality can affect the response to a particular therapy. In this study response to Len was significantly higher in myeloma with trisomy compared with IgH translocated myeloma. Although the routine use of combinations of a proteasome inhibitor plus Len in frontline therapy in all patients with myeloma may overcome the lower response rate in IgH translocated patients, in most countries such a regimen is not approved or economically feasible. Based on our study, newly diagnosed patients with evidence of trisomy on FISH could be considered for a Len-based regimen such as lenalidomide-low dose dexamethasone. Additional studies should examine if use of bortezomib in patients with IgH translocation will lead to better outcomes compared with Len based therapies. Disclosures: Lacy: Celgene Corporation: Research Funding. Gertz:Celgene: Honoraria. Dispenzieri:Celgene, Millenium, Jansenn, Pfizer: Research Funding. Kumar:Merck: Consultancy, Honoraria; Celgene: Consultancy, Research Funding; Millennium: The Takeda Oncology Company: Research Funding; Novartis: Research Funding; Genzyme: Research Funding.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1742-1742 ◽  
Author(s):  
Paul Richardson ◽  
Sundar Jagannath ◽  
Andrzej Jakubowiak ◽  
Sagar Lonial ◽  
Noopur Raje ◽  
...  

Abstract Background: Bortezomib (VELCADE®, Bz) is approved for the treatment of multiple myeloma (MM). Lenalidomide (Revlimid®, Len) plus dexamethasone (Dex) is approved for the treatment of relapsed MM pts following ≥ 1 prior therapy. In a phase 1 study, Len/Bz (maximum tolerated dose [MTD] 15 mg/1.0 mg/m2) with or without Dex (20–40 mg) was well tolerated and resulted in a response rate (CR+PR+MR) of 58% in relapsed and/or refractory MM pts. The aim of this multi-center phase 2 study was to evaluate the efficacy and safety of Len/Bz/Dex (RVD) at the phase 1 MTD. Methods: Pts with relapsed or relapsed and refractory MM with 1–3 prior lines of therapy received up to eight 21-day cycles of Bz 1.0 mg/m2 (days 1, 4, 8, 11) Len 15 mg (days 1–14) and Dex 40/20 mg (cycles 1–4/5–8) on days of/after Bz dosing. After cycle 8, pts with stable or responding disease received maintenance therapy on a 21-day cycle; Bz (days 1 and 8) and Len (days 1–14) at the dose levels tolerated at the end of cycle 8, with Dex at 10 mg (days 1, 2, 8, 9) until disease progression or unacceptable toxicity. Pts received concomitant anti-thrombotic (aspirin 81 or 325 mg/day) and anti-viral prophylaxis. Pts with Grade (G) ≥ 2 peripheral neuropathy (PNY) were excluded. Response was assessed according to modified EBMT and Uniform Criteria with toxicities assessed using NCI CTCAE v3.0. Primary end point was time to progression (TTP). Results: 64 pts have been enrolled; 38 (59%) with relapsed and 26 (41%) with relapsed and refractory MM. Median number of prior therapies was two, including Len (8%), Bz (55%), Dex (92%), thalidomide (77%), and stem cell transplant (SCT; 36%). Pts received a median of 8 cycles; 31 (48%) completed 8 cycles, 24 continue on maintenance. 4 pts proceeded to stem cell collection (median CD34+ yield of 4.58x106/kg after median of 8 cycles of therapy) and 3 pts proceeded to stem cell transplant, with the 4th pt currently undergoing salvage therapy to restore disease control. 19 pts discontinued prior to completing 8 cycles due to: progressive disease (10), toxicities (3) or other reason (2). Toxicities were manageable, consisting primarily of G1–2 myelosuppression. Attributable non-hematologic toxicities included deep vein thrombosis (two pts; attributed to Len, both pts remain on-study after antithrombotic therapy), and two episodes of atrial fibrillation (G3) prompting Dex dose reduction. G3 PNY was reported in one pt attributed to Bz and leading to treatment discontinuation despite Bz dose reduction. Dose reductions were required for: Len (13 pts); Bz (9 pts) and Dex (26 pts). One on-study death (thought to be due to fungal pneumonia) was reported during cycle 3: this was not attributed to either Bz or Len, but possibly Dex. In 63 response-evaluable pts, the overall response rate (CR/nCR+VGPR+ PR+ MR) is currently 86%, including 24% CR/nCR and 67% CR/nCR/VGPR/PR. Response rates according to baseline cytogenetics, disease stage, and prior therapies showed no significant differences according to adverse risk (Table). Similarly, analysis of evaluable pts with chromosome 13 deletion by FISH showed no significant difference in response rate compared to those without. Median DOR in responding pts is 21 weeks; range 6–72 weeks. Median TTP and overall survival have not yet been reached. Conclusions: RVD is very active and well tolerated in pts with relapsed and/or refractory MM, including pts who have received prior Len, Bz, thalidomide, and SCT. Responses appeared independent of adverse cytogenetics, advanced disease stage at diagnosis, prior treatment, and being refractory to prior therapy. Durable responses have been observed. Surrogates and correlative studies are in progress. Response rates* by baseline cytogenetics, disease stage at diagnosis, and prior treatment Cytogenetics (n= 60) ORR (≥ MR) ≥ PR ≥ VGPR CR/nCR * Response assessed by Uniform Criteria. †Abnormal cytogenetics by metaphase analysis Normal (n=38) 84% 63% 29% 18% Abnormal† (n=22) 91% 73% 41% 32% ISS disease stage (n=44) Stage l (n=15) 80% 67% 40% 33% Stage ll (n=15) 87% 67% 20% 13% Stage lll (n=14) 86% 71% 57% 36% Response to RVD according to type of prior therapy Bortezomib (n=35) 77% 57% 31% 17% Len/Thalidomide (n=54) 81% 57% 22% 15% Response by Relapsed/Refractory Relapsed (n=38) 84% 66% 34% 26% Relapsed/Refractory (n=26) 85% 65% 31% 19%


Sign in / Sign up

Export Citation Format

Share Document