scholarly journals MUKnine OPTIMUM protocol: a screening study to identify high-risk patients with multiple myeloma suitable for novel treatment approaches combined with a phase II study evaluating optimised combination of biological therapy in newly diagnosed high-risk multiple myeloma and plasma cell leukaemia

BMJ Open ◽  
2021 ◽  
Vol 11 (3) ◽  
pp. e046225
Author(s):  
Sarah Brown ◽  
Debbie Sherratt ◽  
Samantha Hinsley ◽  
Louise Flanagan ◽  
Sadie Roberts ◽  
...  

IntroductionMultiple myeloma (MM) is a plasma cell tumour with over 5800 new cases each year in the UK. The introduction of biological therapies has improved outcomes for the majority of patients with MM, but in approximately 20% of patients the tumour is characterised by genetic changes which confer a significantly poorer prognosis, generally termed high-risk (HR) MM. It is important to diagnose these genetic changes early and identify more effective first-line treatment options for these patients.Methods and analysisThe Myeloma UK nine OPTIMUM trial (MUKnine) evaluates novel treatment strategies for patients with HRMM. Patients with suspected or newly diagnosed MM, fit for intensive therapy, are offered participation in a tumour genetic screening protocol (MUKnine a), with primary endpoint proportion of patients with molecular screening performed within 8 weeks. Patients identified as molecularly HR are invited into the phase II, single-arm, multicentre trial (MUKnine b) investigating an intensive treatment schedule comprising bortezomib, lenalidomide, daratumumab, low-dose cyclophosphamide and dexamethasone, with single high-dose melphalan and autologous stem cell transplantation (ASCT) followed by combination consolidation and maintenance therapy. MUKnine b primary endpoints are minimal residual disease (MRD) at day 100 post-ASCT and progression-free survival. Secondary endpoints include response, safety and quality of life. The trial uses a Bayesian decision rule to determine if this treatment strategy is sufficiently active for further study. Patients identified as not having HR disease receive standard treatment and are followed up in a cohort study. Exploratory studies include longitudinal whole-body diffusion-weighted MRI for imaging MRD testing.Ethics and disseminationEthics approval London South East Research Ethics Committee (Ref: 17/LO/0022, 17/LO/0023). Results of studies will be submitted for publication in a peer-reviewed journal.Trial registration numberISRCTN16847817, May 2017; Pre-results.

Blood ◽  
1996 ◽  
Vol 88 (5) ◽  
pp. 1780-1787 ◽  
Author(s):  
TE Witzig ◽  
MA Gertz ◽  
JA Lust ◽  
RA Kyle ◽  
WM O'Fallon ◽  
...  

Abstract The purpose of this study was to quantitate the number and labeling index of monoclonal plasma cells in the blood of patients with newly diagnosed multiple myeloma (MM) to learn if these values were independent prognostic factors for survival. Patients were candidates for this study if they had untreated myeloma requiring therapy, were evaluated at our institution between 1984 and 1993, and had a sample of blood analyzed with a sensitive immunofluorescence technique for monoclonal plasma cells and the blood B-cell labelling index (BLI). The % blood monoclonal plasma cells (%BPC) and the BLI were analyzed along with stage, marrow plasma cell LI, % marrow plasma cells, calcium, creatinine, albumin, beta-2-microglobulin, and C-reactive protein as univariate and multivariate factors for survival. Eighty percent of the 254 patients accrued to this study had monoclonal BPC detected. The median % BPC was 6% and 57% (144 of 254) of patients had a high number (> or = 4%). Patients with > or = 4% BPC had a median survival of 2.4 years vs 4.4 years for those with < 4% BPC (P < .001). The BLI was also prognostic (P = .008). In a multivariate analysis, the % BPC, age, albumin, stage, marrow plasma cell LI, and the BLI were independent factors for survival. The %BPC and the marrow plasma cell LI best separated the group into low, intermediate, and high risk myeloma with median survivals of 52, 35, and 26 months, respectively. Patients with high %BPC were less likely to have lytic bone disease from their MM (P = .002). The %BPC and the BLI are independent prognostic factors for survival and are useful in identifying patients as low, intermediate, and high risk. Clonal cells in the blood should be quantified in future clinical trials for myeloma.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1871-1871 ◽  
Author(s):  
Christopher P Venner ◽  
Richard Leblanc ◽  
Irwindeep Sandhu ◽  
Darrell J White ◽  
Andrew Belch ◽  
...  

Background: Carfilzomib is effective in the treatment of relapsed and refractory multiple myeloma (RRMM). Questions remain regarding optimal dosing strategies and combinations. The MCRN-003/MYX.1 single arm phase II clinical trial of high-dose once weekly carfilzomib in combination with dexamethasone and cyclophosphamide (wCCD) in RRMM met its primary endpoint with an overall response rate (ORR) ≥ 80% after 4 treatment cycles [Venner, Blood 2018 132:1984]. This abstract focuses on previously unreported protocol specified secondary and exploratory endpoints including progression free (PFS) and overall survival (OS). Methods: This multi-centre clinical trial is run through the Myeloma Canada Research Network (MCRN) with support from the Canadian Cancer Trials Group (CCTG). Patients who had 1-3 prior lines of therapy and without proteasome inhibitor (PI) refractory disease were eligible. Treatment consists of carfilzomib (20 mg/m2 day 1 of first cycle then escalated to 70 mg/m2 for all subsequent doses) given on days 1, 8, and 15 of a 28-day cycle, plus weekly oral dexamethasone 40 mg and cyclophosphamide 300 mg/m2 capped at 500 mg. Treatment continues until progression or intolerance, except for cyclophosphamide which is discontinued after 12 cycles. Secondary endpoints included toxicity, depth of response, PFS and OS as defined by International Myeloma Working Group Uniform Response Criteria (2016). Exploratory endpoints included the impact of cytogenetics (CG) and prior PI or lenalidomide exposure on efficacy, and the novel endpoint of serum free light chain (sFLC) escape, defined as a > 25% change in the difference of involved to uninvolved light chain with the absolute rise > 100mg/L, in individuals with disease previously measurable by serum or urine protein electrophoresis. This analysis is based on the locked database of 19 June, 2019. Results: Of 76 patients accrued, 75 were included in the analysis. One was ineligible due to prior bortezomib refractoriness. Thirty-nine percent received 1 prior line, 44% two prior lines and 17% three prior lines of therapy. High-risk cytogenetics (t(4;14), t(14;16) and/or del P53, considered positive at any level above local accepted threshold) were identified in 32%. Twenty percent had ISS stage III disease. The majority of participants were previously exposed to PI (87%) and lenalidomide (83%). The median duration of follow-up was 25 months. The ORR at any time was 85% (1 patient achieved a response after 4 cycles) with ≥ VGPR achieved in 68% and ≥ CR in 29%. The presence of high-risk CG conferred a worse ORR (75% vs 97% respectively, p = 0.013). Thirty-one patients have died with a median OS and median PFS of 27 months and 17 months respectively (figure 1). High risk CG conferred a worse median OS (18 months vs NR, p = 0.002) and a trend toward a worse median PFS with high risk CG (14 months vs 22 months, p = 0.06; figure 1). For patients with prior PI exposure the median OS and PFS were 27 and 17 months respectively. For patients with prior lenalidomide exposure median OS and PFS were 26 and 16 months respectively. Free light chain escape events were noted in 11 patients (15%) but was the only progression event in 3 (4%). For the remaining 8 patients the sFLC rise was a harbinger of traditional relapse by electrophoresis. The median PFS when sFLC escape was included as a progression event was 17 months. With updated toxicity data the most common ≥ grade 3 non-hematologic events were infection (40%), cardiac (15%, including 5 dyspnea and 1 pulmonary edema) and vascular (17%, including 7 with hypertension and 3 with thrombotic microangiopathy). To date 57 (76%) patients have discontinued carfilzomib, including 34 due to disease progression and 14 due to toxicity. Conclusion: This phase II trial demonstrates that wCCD remains a safe and effective regimen for RRMM. The survival data presented here is comparable to current phase II and III studies examining the weekly dosing strategy. No new toxicity signals are observed but cardiovascular risks remain an important factor in the use of carfilzomib-based therapies. Using sFLC escape does not negatively affect PFS outcomes but likely better characterizes progression as a harbinger of more traditional events detected by electrophoresis. This regimen will be a useful triplet-based option for RRMM especially in patients refractory to lenalidomide and otherwise ineligible for the carfilzomib-lenalidomide-dexamethasone combination. Disclosures Venner: J&J: Research Funding. Leblanc:Amgen: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees. Sandhu:Takeda: Honoraria; Amgen: Honoraria; Celgene: Honoraria; Pfizer: Honoraria; Janssen: Honoraria; gilead: Honoraria. White:Celgene: Consultancy, Honoraria; Sanofi: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Janssen: Consultancy, Honoraria. Reece:Takeda: Consultancy, Honoraria, Research Funding; Amgen: Consultancy, Honoraria, Research Funding; BMS: Research Funding; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Karyopharm: Membership on an entity's Board of Directors or advisory committees, Research Funding; Otsuka: Research Funding; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Merck: Research Funding. Chen:Celgene: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Amgen: Honoraria. Louzada:Celgene: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Bayer: Honoraria. McCurdy:Janssen: Honoraria; Celgene: Honoraria. Hay:Janssen: Research Funding; Novartis: Research Funding; AbbVie: Research Funding; Kite: Research Funding; Takeda: Research Funding; Roche: Research Funding; Celgene: Research Funding; Seattle Genetics: Research Funding; MorphoSys: Research Funding; Gilead: Research Funding. OffLabel Disclosure: While Carfilzomib is approved for use in relapsed and refractory myeloma the combination with cyclophosphamide is not approved.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2874-2874 ◽  
Author(s):  
Norbert Grzasko ◽  
Marek Hus ◽  
Marta Morawska ◽  
Roman Hajek ◽  
Anna Dmoszynska

Abstract Abstract 2874 Introduction Cytogenetic abnormalities are concerned the major novel prognostic factors in patients with newly diagnosed multiple myeloma (MM). Chromosome 1 abnormalities, mainly 1q gain and 1p loss, are regarded as major prognostic factor in MM and their presence was shown to be associated with unfavourable disease course, although they often coexist with other genetic changes and amp1q21 alone was not showed to be an adverse prognostic factor in all studies. Methods We explored the prognostic value of amp1q21 alone and in coexistence with del17p13, del13q14 and t(4, 14) detected by fluorescence in situ hybridization (FISH) in newly diagnosed MM patients. The study was conducted in a cohort of 79 newly diagnosed MM patients upon the local Ethics Committee approval and according to guidelines of Declaration of Helsinki. All patients received first line regimens including thalidomide in a daily dose of 100 mg: 59 (75%) patients were treated with CTD (cyclophosphamide, thalidomide, dexamethasone) and 20 (25%) patients with MPT (melphalan, prednisone, thalidomide). Then 28 (35%) patients previously treated with CTD were given high-dose therapy with autologous stem cell support (HDT/ASCT). Results FISH analysis detected amp1q21 in 39 (49%), del13q14 in 38 (48%), t(4;14) in 16 (20%) and del17p13 in 13 (16%) patients. In 24 (30%) patients amp1q21 was combined with del13q14, in 12 (15%) with t(4;14) and in 5 (6%) with del17p13. The presence of amp1q21 correlated significantly with del13q14 and t(4;14). In the whole cohort of patients, the median PFS was 32.3 months in amp1q21-negative and 14.4 months in amp1q21-positive patients (p=0.049); the median OS was accordingly 40.0 and 26.2 months (p=0.027). In amp1q21-positive patients, the coexistence of additional aberrations made the outcome worse: del13q14 significantly shortened both PFS (22.8 vs 8.5 months, p=0.018) and OS (not reached vs 27 months, p=0.033), del17p13 significantly shortened OS (27.6 vs 11.5 months, p=0.048) and t(4;14) also significantly shortened OS (not reached vs 18.9 months, p=0.012). Moreover, analysis of patients with isolated amp1q21 showed that they have similar prognosis as amp1q21-negative patients (PFS not reached vs 22.7 months, p>0.05 and OS not reached for both, p>0.05). On the other hand, the amp1q21-positive patients with any of additional aberrations have significantly shortened survival: the median PFS of 8.5 months was significantly shortened in comparison both with amp1q21-negative patients (p=0.006) and with patients with isolated amp1q21 (p=0.018); the median OS of 16.7 months was significantly shortened in comparison with both of abovementioned groups (accordingly p=0.032 and p=0.033). The Kaplan-Meier estimates of PFS and OS are illustrated in Figure 1. Conclusions We demonstrate that isolated amp1q21 is not associated with poor prognosis in newly diagnosed MM patients treated with thalidomide. Our data clearly show that patients carrying amp1q21 accompanied by other genetic abnormalities, like del13q14, del17p13 or t(4;14), have significantly shortened PFS and OS than patients without amp1q21 or isolated amp1q21 and thalidomide-based regimens should not be recommended in this subset of patients. Acknowledgments The work was supported by a grant from the State Committee for Scientific Research, No. NN402287236.Figure 1Figure 1. Amp1q21-positive patients with additional aberrations had significantly shortened PFS and OS than amp1q21-negative patients (PFS: 8.5 months vs not reached, p=0.006: OS: 16.7 months vs not reached, p=0.032) and than patients with isolated amp1q21 (PFS: 8.5 vs 22.7 months, p=0.018; OS: 16.7 months vs not reached, p=0.033). The difference in PFS and OS between amp1q21-negative patients and patients with isolated amp1q21 was not statistically significant (p>0.05 for both). Disclosures: Dmoszynska: Mundipharma: ; Roche: Honoraria.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3069-3069 ◽  
Author(s):  
Antonio Palumbo ◽  
Federica Cavallo ◽  
Izhar Hardan ◽  
Barbara Lupo ◽  
Valter Redoglia ◽  
...  

Abstract Abstract 3069FN2 Background: High-dose chemotherapy with haemopoietic stem-cell improves outcome in multiple myeloma (MM). The introduction of novel agents questions the role of autologous stem-cell transplantation (ASCT) in MM patients. Aims: In this prospective randomized study, we compared conventional melphalan-prednisone-lenalidomide (MPR) with tandem high-dose melphalan (MEL200) in newly diagnosed MM patients younger than 65 years. Methods: All patients (N=402) received four 28-day cycles of lenalidomide (25 mg, d1-21) and low-dose dexamethasone (40 mg, d1, 8, 15, 22) (Rd) as induction. As consolidation, patients were randomized to MPR (N=202) consisting of six 28-day cycles of melphalan (0.18 mg/kg d1-4), prednisone (2 mg/kg d1-4) and lenalidomide (10 mg d1-21); or tandem melphalan 200 mg/m2 MEL200 (N=200) with stem-cell support. All patients enrolled were stratified according to International Staging System (stages 1 and 2 vs. stage 3) and age (<60 vs. ≥60 years). Progression-free survival (PFS) was the primary end point. Data were analyzed in intention-to-treat. Results: Response rates were similar: at least very good partial response (≥VGPR) rate was 60% with MPR vs. 58% with MEL200 (p=.24); the complete response (CR) rate was 20% with MPR vs. 25% with MEL200 (p=.49). After a median follow-up of 26 months, the 2-year PFS was 54% in MPR and 73% in MEL200 (HR=0.51, p<.001). The 2-year overall survival (OS) was similar in the two groups: 87% with MPR and 90% with MEL200 (HR 0.68, p=.19). In a subgroup analysis, MEL200 significantly prolonged PFS in both standard-risk patients without t(4;14) or t(14;16) or del17p abnormalities (2-year PFS was 46% in the MPR group vs. 78% in the MEL200 group, HR=0.57, p=.007) and high-risk patients with t(4;14) or t(14;16) or del17p abnormalities (2-year PFS was 27% for MPR vs. 71% for MEL200, HR=0.32, p=.004). In patients who achieved CR, the 2-year PFS was 66% for MPR vs. 87% for MEL200 (HR 0.26; p<.001); in those who achieved a partial response (PR), the 2-year PFS was 56% for MPR vs. 77% for MEL200 (HR 0.45; p<.001). In the MPR and MEL200 groups, G3-4 neutropenia was 55% vs. 89% (p<.001); G3-4 infections were 0% vs. 17% (p<.001); G3-4 gastrointestinal toxicity was 0% vs. 21% (p<.001); the incidence of second tumors was 0.5% in MPR patients and 1.5% in MEL200 patients (p=.12). Deep vein thrombosis rate was 2.44% with MPR vs. 1.13% with MEL200 (p=.43). Conclusions: PFS was significantly prolonged in the MEL200 group compared to MPR. This benefit was maintained in the subgroup of patients with standard- or high-risk cytogenetic features. Toxicities were significantly higher in the MEL200 group. This is the first report showing a PFS advantage for ASCT in comparison with conventional therapies including novel agents. These data will be updated at the meeting. Disclosures: Palumbo: celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees. Cavallo:Celgene: Honoraria; Janssen-Cilag: Honoraria. Cavo:celgene: Honoraria. Ria:celgene: Consultancy. Caravita Di Toritto:Celgene: Honoraria, Research Funding. Di Raimondo:celgene: Honoraria. Boccadoro:celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3024-3024
Author(s):  
Sule Mine Bakanay ◽  
Klara Dalva ◽  
Berna Elif Koksoy ◽  
Erol Ayyildiz ◽  
Muhit Ozcan ◽  
...  

Abstract Abstract 3024 Introduction: High dose melphalan supported by autologous hematopoietic cell transplantation has been shown to prolong survival and decrease relapse rates compared to conventional chemotherapies in elligible patients with multiple myeloma (MM) but unfortunately relapses remain a problem. Various factors have been shown to affect the relapse after transplantation. Malignant plasma cells (PCs) have been documented to contaminate blood stem cell harvest products. Conflicting results have been reported regarding the levels of PC contamination in apheresis products from patients with MM undergoing transplantation (Tx) and their impact on relapse and survival. Aim of this study was to analyze post induction residual myeloma by flow cytometric detection of the graft and by whole body PET imaging. Method: Standard panel was set up with CD138FITC/CD38PE/CD45ECD/CD56PC5. CD45negCD56pos plasma cells were identified as abnormal plasma cells. If any aberrant expression [such as CD20(loss) CD27(loss) CD28 (gain), CD33(loss), CD34 (gain), CD81(loss), CD117(gain)] is detected at diagnosis, the corresponding antibody was also added to the panel. Univariate and multivariate analysis were performed by SPSS.16. Results: Apheresis products from 118 patients (female/male=53/65; median age at diagnosis= 55 (35–69)) were tested for the presence of abnormal PCs and the number of normal PCs. The number of patients in MM subtypes IgG/IgA/Light chain/non-secretory were 70/20/26/2. International scoring system I/II/III:50/38/30. Most patients (n=108) had received one or two lines of induction and 52 had bortezomib based therapies before transplantation. In univariate analysis age (> 55; ≤ 55), ISS (ISS1 vs ISS2 + ISS3), beta2 mcg (>3.5 and ≤ 3.5) and MM Subtype (light chain vs others) were significantly associated with response to Tx. Presence of abnormal PCs in the harvests did not significantly affect response to Tx or OS. However, both the presence of abnormal PCs and the amount of normal PCs > 7.6 ×105/kg was associated with shorter PFS. Fifty-five of the patients had pre and post-Tx PET-imaging done. On the other hand, post-Tx PET positivity negatively affected the PFS. Post-Tx PET positivity was also significantly associated with relapse at 12 months after transplantation. Pre and post-Tx disease status significantly influenced relapse after transplantation. In multivariate analysis, age, beta2 mcg and light chain myeloma continued to be significantly associated with response to transplantation. Abnormal PC contamination and the amount of normal PCs> 7.6 ×105/kg were not associated with pre-Tx PET positivity. Conclusions: Age, ISS, beta2 mcg, light chain disease are predictors of response to transplant. Response to the treatments before mobilization, response to Tx, post Tx PET positivity, graft plasma cell (abnormal or normal) content, serum LDH are predictors of PFS. Response (≥VGPR) during mobilization correlates with less abnormal plasma cells in the apheresis product. PC contamination of the graft does not prevent response to transplant. However, presence of abnormal PC in the apheresis product as well as PET positivity predicts shorter DFS compared to lack of residual disease by flow or PET imaging (11 vs 18 months). Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document