Four phase 3 studies of the oral proteasome inhibitor (PI) ixazomib for multiple myeloma in the newly-diagnosed, relapsed/refractory, and maintenance settings: TOURMALINE-MM1, -MM2, -MM3, and -MM4

2015 ◽  
Vol 15 ◽  
pp. e174 ◽  
Author(s):  
J. San Miguel ◽  
P. Moreau ◽  
V. Rajkumar ◽  
A. Palumbo ◽  
T. Facon ◽  
...  
Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2386-2386
Author(s):  
Si-Tien Wang ◽  
Mei Sheng Duh ◽  
Hui Huang ◽  
Leigh Ann White ◽  
Eva Chang ◽  
...  

Abstract Background: Induction therapy for multiple myeloma (MM) patients who are eligible for autologous stem cell transplant (ASCT) has evolved with the introduction of novel agents, such as bortezomib (VELCADE®, Vel) and thalidomide (THALOMID®, Thal). Phase 3 trial data show the clinical effectiveness of front-line bortezomib-based therapies, but little is known about the comparative economic benefit of bortezomib-based regimens in newly diagnosed MM in the transplant setting. Here we report a pharmacoeconomic analysis of the IFM 2005-01, GIMEMA MMY-3006, and the Myeloma-Autogreffe Group (MAG) phase 3 trials, comparing the relative costs of Vel–dexamethasone (Vel/Dex), vincristine–doxorubicin–dexamethasone (VAD), Vel–Thal–Dex (VTD), and Thal–Dex (TD). Methods: Expanding on a previous budget impact model of bortezomib treatment for relapsed MM (Fullerton et al. Blood2007: Abstract 3324), an Excel-based model was developed to calculate the cost of therapy for MM patients eligible for ASCT, including costs incurred during induction therapy and single or double transplant. Per IFM 2005-01 protocol design, we assumed that patients who achieved at least very good partial response (≥VGPR) after their first transplant did not need a second transplant; the GIMEMA and MAG trials were based on a similar tandem transplant protocol. Costs were evaluated from a health system perspective and included: induction therapy costs (drugs, medical care, adverse events [AEs], and prophylaxis treatment), cost of the first ASCT, and cost of the second ASCT if required. Drug costs were calculated based on 2008 Average Wholesale Price and applied to trial regimens. Medical care costs were based on the Medicare physician fee schedule and assumptions drawn from National Comprehensive Cancer Network (NCCN) MM treatment guidelines. AE costs (grade ≥3 AEs reported in at least two studies) were estimated using available incidence data and reported as per-event costs or annual costs depending on the data source. The unit cost of a transplant ($139,220) was calculated based on data from 1,917 MM patients treated at >120 centers in the OptumHealth database. Results: In IFM 2005-01, estimated mean per-patient treatment costs from start of induction to completion of second transplant (if required) were lower for patients receiving Vel/Dex induction ($201,793) versus VAD ($217,526), due to better post-induction response with Vel/Dex. More patients achieved post-transplant ≥VGPR with Vel/Dex than with VAD (72% vs 52%), and therefore did not require a second transplant. Similarly, in GIMEMA MMY-3006, post-induction and posttransplant ≥VGPR achieved with VTD induction was greater than that achieved with TD (post-induction: 60% vs 27% and post-transplant: 77% vs 54%), thereby reducing the need for a second transplant among patients receiving VTD induction therapy. Consequently, estimated mean treatment costs were lower for patients receiving VTD induction ($200,093) versus TD ($218,886). Estimated mean treatment costs for patients receiving VD induction in IFM 2005-01 ($201,793) and VTD induction in GIMEMA MMY-3006 ($200,093) were both lower than those for patients receiving TD induction in the MAG phase 3 trial ($239,851). Conclusions: In summary, the model showed that total costs were lower for patients receiving bortezomib-based induction compared with TD induction in both the GIMEMA MMY-3006 and MAG trials. These health economic analyses also suggest that bortezomib-based induction results in lower mean per-patient treatment costs when compared with VAD or TD induction, primarily due to better response post-first ASCT. Table: Cost Summaries of ASCT Trials Induction costs VD (IFM) VTD (GIMEMA) TD (MAG) Drugs $15,940 $24,273 $16,596 Medical care $4,147 $3,578 $1,019 Adverse events $2,948 $1000 $5,610 Total induction costs (per patient) $23,035 $28,852 $23,225 Transplant costs 1st ASCT $139,220 $139,220 $139,220 2nd ASCT $39,538 $32,021 $77,406 Total transplant costs (per patient) $178,758 $171,241 $216,626 TOTAL costs $201,793 $200,093 $239,851


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5752-5752 ◽  
Author(s):  
Hiroshi Handa ◽  
Kenshi Suzuki ◽  
Takaaki Chou ◽  
Takafumi Matsushima

Background Ixazomib is the first oral proteasome inhibitor to be investigated clinically for the treatment of MM. Phase 1 studies have shown single-agent activity and manageable toxicities in RRMM (Kumar et al. Blood 2014) and phase 1/2 studies have suggested the feasibility and activity of weekly oral ixazomib plus Rd in previously untreated MM (Kumar et al. ASH 2012; Richardson et al. ASH 2013). These findings have led to ongoing phase 3 trials of weekly ixazomib 4 mg + Rd in RRMM and previously untreated MM. However, the early-phase studies were conducted in Western pts. This phase 1, open-label multicenter study aimed to determine the safety, tolerability, and pharmacokinetics (PK) of weekly ixazomib alone or with Rd in Japanese pts with RRMM (Japic Clinical Trials Information no. 121822). Methods Primary objectives were to evaluate the safety and tolerability, including dose-limiting toxicities (DLTs) and adverse events (AEs), and the PK of ixazomib alone or with Rd. A secondary objective was evaluation of antitumor activity. Japanese pts aged ≥20 years with RRMM who had received at least 2 prior regimens, which must have included bortezomib, thalidomide or lenalidomide, and corticosteroids, were eligible. All had measurable disease and ECOG performance status of 0–2. Pts with grade ≥2 peripheral neuropathy or grade ≥2 diarrhea at study entry were excluded. Pts received ixazomib 4 mg on days 1, 8, and 15 of 28-day cycles, alone or with Rd (lenalidomide 25 mg on days 1–21, dexamethasone 40 mg on days 1, 8, 15, and 22), per the regimen used in the ongoing phase 3 trials. AEs were graded per NCI-CTCAE v4.03. Blood samples for PK analysis were taken at multiple time points prior to and after dosing on days 1 and 15 of cycle 1. Responses were assessed per IMWG uniform response criteria. Results Fourteen pts were enrolled; 8 (57%) were male, median age was 62.5 yrs (range 53–71), 4 pts were aged ≥65 yrs, median number of prior therapies was 7. Seven pts received single-agent ixazomib and 7 received ixazomib + Rd. One pt in each cohort was excluded from the DLT-evaluable population. Two patients experienced DLTs in cycle 1: 1 pt receiving single-agent ixazomib had grade 4 thrombocytopenia and grade 3 diarrhea, hypertension, hypokalemia, hyponatremia, and nausea; 1 pt in the ixazomib + Rd cohort had grade 4 thrombocytopenia and neutropenia. All events were considered treatment-related. At data cut-off (Jan 6 2014), 6 pts remained on treatment and 8 had discontinued due to: progressive disease (PD; n=3), AEs (n=3), symptomatic deterioration, and protocol violation (each n=1). At data cut-off, pts (n=14) had received a median of 6 cycles of ixazomib (range 1–21); the 7 pts in the ixazomib + Rd cohort had received a median of 4 cycles (range 1–12) of ixazomib + Rd. Thirteen (93%) pts experienced treatment-related AEs; the most common were neutropenia (71%), thrombocytopenia (71%), leukopenia (64%), lymphopenia (57%), and diarrhea (50%). There were no cases of peripheral neuropathy. Nine (64%) pts had grade ≥3 AEs; the most common were lymphopenia (50%), neutropenia (43%), and thrombocytopenia (36%). Two (14%) pts (single-agent cohort) had serious AEs (grade 2 bronchitis in 1 pt, and grade 4 thrombocytopenia and grade 3 hypokalemia in 1 pt). Three pts discontinued due to AEs; 1 due to diarrhea in the single-agent cohort, and 1 due to neutropenia and 1 due to thrombocytopenia in the ixazomib + Rd cohort. There were no deaths. PK data showed ixazomib was rapidly absorbed with a Tmax at 1.08–1.83 hrs. Terminal half-life (geometric mean) was 5.7 days for single-agent ixazomib and 5.2 days for ixazomib + Rd. There were no substantial differences in the ixazomib PK profile between the two cohorts. Thirteen pts were response-evaluable. One pt (ixazomib + Rd cohort) had a partial response; at data cut-off, this pt remained in response with a 100% M-protein reduction (unconfirmed VGPR) and duration of response of ~10.8 months. Seven pts had stable disease (including 3 with M-protein reductions of 25–50%), 2 had PD, and 3 were not assessable. Conclusions These data suggest that ixazomib 4 mg alone or with Rd is feasible and tolerable in Japanese pts with RRMM. The AEs were manageable, reflecting the AE profile seen in Western populations, supporting the use of this dose and schedule in Japanese pts. Disclosures Handa: Celgene: Research Funding; Yakult: Research Funding; Kirin: Research Funding; Chugai: Research Funding. Off Label Use: Investigational agent ixazomib for the treatment of Japanese patients with relapsed and/or refractory multiple myeloma.. Matsushima:Takeda Pharmaceutical Company Limited : Employment.


2016 ◽  
Vol 34 (15_suppl) ◽  
pp. TPS8068-TPS8068 ◽  
Author(s):  
Antonio Palumbo ◽  
Gareth John Morgan ◽  
S.Vincent Rajkumar ◽  
Sagar Lonial ◽  
Wee Joo Chng ◽  
...  

2021 ◽  
Author(s):  
Xiaoyan Han ◽  
Chunxiang Jin ◽  
Gaofeng Zheng ◽  
Donghua He ◽  
Yi Zhao ◽  
...  

Abstract Background: According to different patients’ subgroups choose optimal maintenance therapy. Methods: 226 Newly Diagnosed Multiple Myeloma (NDMM) patients in our center were included, the patients’ characteristics, survival, response, subgroup analysis, adverse reactions were compared between the patients with or without maintenance, proteasome inhibitor (PI) or immunomodulators (IMiDs) maintenance. And the survival of different maintenance duration of bortezomib-based regimens was also analyzed.Results: The maintenance therapy not only upgraded more patients’ response (34.3 vs. 13.3%, p= 0.006), but also significantly prolonged the patients’ PFS (median PFS: 41.1 vs. 10.5 months, p < 0.001) and OS (median OS: not reached vs. 38.6 months, p < 0.001). Compared with IMiDs, the PFS (median PFS: 43.7 vs. 38.5 months, p = 0.034) and OS (median OS: not reached vs. 78.5 months, p = 0.041) can both benefit from bortezomib-based maintenance. The patients younger than 65 years old with bortezomib-based maintenance significantly prolonged the OS (p= 0.032). Patients achieving the only partial response (PR) after induction and consolidation therapy experienced a significantly longer PFS and OS with bortezomib-based maintenance compared to IMiDs (p= 0.007, 0.002). Besides, the high-risk patients (ISS 2-3, DS 2-3 and RISS 2-3) with bortezomib-based maintenance can benefit PFS (p= 0.002, 0.02, 0.06, respectively) and OS (p=0.059, 0.047, 0.044, respectively) compared with IMiDs. The OS was significantly prolonged in the patients who received ≥12 months of bortezomib-based maintenance than those with maintenance < 12 months (p< 0.001), but no difference was observed in OS between the patients who received the 12-24 or ≥ 24months of bortezomib-based maintenance (p= 0.292).Conclusion: Maintenance therapy can significantly improve the survival of NDMM patients. Bortezomib-based regimens maintenance was more superior to IMiDs in overall PFS and OS. The beneficial effect is most evident in patients achieving the only PR after induction and consolidation therapy, and the high-risk patients. Moreover, younger patients also could benefit from bortezomib-based maintenance in OS. The bortezomib-based maintenance duration lasting 12-24 months after induction and consolidation therapy can reach a satisfactory OS.


PLoS Medicine ◽  
2021 ◽  
Vol 18 (1) ◽  
pp. e1003454
Author(s):  
Graham H. Jackson ◽  
Charlotte Pawlyn ◽  
David A. Cairns ◽  
Ruth M. de Tute ◽  
Anna Hockaday ◽  
...  

Background Carfilzomib is a second-generation irreversible proteasome inhibitor that is efficacious in the treatment of myeloma and carries less risk of peripheral neuropathy than first-generation proteasome inhibitors, making it more amenable to combination therapy. Methods and findings The Myeloma XI+ trial recruited patients from 88 sites across the UK between 5 December 2013 and 20 April 2016. Patients with newly diagnosed multiple myeloma eligible for transplantation were randomly assigned to receive the combination carfilzomib, lenalidomide, dexamethasone, and cyclophosphamide (KRdc) or a triplet of lenalidomide, dexamethasone, and cyclophosphamide (Rdc) or thalidomide, dexamethasone, and cyclophosphamide (Tdc). All patients were planned to receive an autologous stem cell transplantation (ASCT) prior to a randomisation between lenalidomide maintenance and observation. Eligible patients were aged over 18 years and had symptomatic myeloma. The co-primary endpoints for the study were progression-free survival (PFS) and overall survival (OS) for KRdc versus the Tdc/Rdc control group by intention to treat. PFS, response, and safety outcomes are reported following a planned interim analysis. The trial is registered (ISRCTN49407852) and has completed recruitment. In total, 1,056 patients (median age 61 years, range 33 to 75, 39.1% female) underwent induction randomisation to KRdc (n = 526) or control (Tdc/Rdc, n = 530). After a median follow-up of 34.5 months, KRdc was associated with a significantly longer PFS than the triplet control group (hazard ratio 0.63, 95% CI 0.51–0.76). The median PFS for patients receiving KRdc is not yet estimable, versus 36.2 months for the triplet control group (p < 0.001). Improved PFS was consistent across subgroups of patients including those with genetically high-risk disease. At the end of induction, the percentage of patients achieving at least a very good partial response was 82.3% in the KRdc group versus 58.9% in the control group (odds ratio 4.35, 95% CI 3.19–5.94, p < 0.001). Minimal residual disease negativity (cutoff 4 × 10−5 bone marrow leucocytes) was achieved in 55% of patients tested in the KRdc group at the end of induction, increasing to 75% of those tested after ASCT. The most common adverse events were haematological, with a low incidence of cardiac events. The trial continues to follow up patients to the co-primary endpoint of OS and for planned long-term follow-up analysis. Limitations of the study include a lack of blinding to treatment regimen and that the triplet control regimen did not include a proteasome inhibitor for all patients, which would be considered a current standard of care in many parts of the world. Conclusions The KRdc combination was well tolerated and was associated with both an increased percentage of patients achieving at least a very good partial response and a significant PFS benefit compared to immunomodulatory-agent-based triplet therapy. Trial registration ClinicalTrials.gov ISRCTN49407852.


Sign in / Sign up

Export Citation Format

Share Document