scholarly journals Benefits of additional cycles of bortezomib/thalidomide/dexamethasone (VTD) induction therapy compared to four cycles of VTD for newly diagnosed multiple myeloma

2019 ◽  
Vol 54 (12) ◽  
pp. 2051-2059 ◽  
Author(s):  
Yoo Jin Lee ◽  
◽  
Joon Ho Moon ◽  
Sang Kyun Sohn ◽  
Seok Jin Kim ◽  
...  
2021 ◽  
Vol 39 (15_suppl) ◽  
pp. TPS8052-TPS8052
Author(s):  
Shaji Kumar ◽  
Zihan Wei ◽  
Michael A. Thompson ◽  
Bradley Snyder ◽  
Matthias Weiss ◽  
...  

TPS8052 Background: The monoclonal antibody (MoAb) daratumumab (dara) has been approved for treatment of newly diagnosed Multiple Myeloma (NDMM) in combination with lenalidomide (len) and dexamethasone (DRd) in patients who are not eligible to undergo stem cell transplantation (SCT). Ongoing trials are examining the role of adding bortezomib (Btz) to DRd, but it remains unclear if all patients benefit from a quadruplet regimen. Availability of sensitive assays to detect measurable/minimal residual disease (MRD) in MM and emerging data demonstrating significant prognostic value for attaining MRD negativity, offers an unprecedented opportunity to develop individualized treatment approaches. An important question is to identify who benefits from adding a fourth drug to the MoAb-IMiD triplet, thus individualizing therapy based on depth of response. We hypothesize that prolonged intensive therapy with the addition of Btz for consolidation and maintenance after DRd induction therapy for NDMM will improve survival outcomes with a more pronounced effect when used in MRD positive patients. Methods: Patients with NDMM, R-ISS Stage I or II, who are not eligible to undergo SCT or those willing to defer SCT to first relapse and have not received more than 1 cycle of any NDMM therapy will be enrolled, provided they have measurable disease, adequate organ and marrow function, have received no more than once cycle of therapy for MM and significant peripheral neuropathy or chronic obstructive pulmonary disease. Importantly, a dominant clone should be identified by lymphotrack assay for future MRD monitoring. Once enrolled, induction therapy will be in 28 day cycles consisting of daraSC (1800 mg) weekly for 2 cycles, every other week for cycles 3-6 and then every 4 weeks for 9 cycles, along with len 25 mg days 1-21 of each cycle and dex 40 mg (20 mg for those > 75 years) weekly. At end of 9 cycles (induction), patients will undergo MRD testing by next generation sequencing and will be classified into MRD positive or negative subgroups. Using MRD as an integral biomarker, the trial employs a randomized biomarker-stratified design as proposed by Freidlin et al. to determine efficacy for each MRD subgroup. Patients will be stratified by MRD status and R-ISS stage and randomized to receive 9 cycles of consolidation with DRd, without (control arm) or with (experimental arm) Btz (1.3 mg/m2 weekly for 3 of 4 weeks), followed by DR maintenance until progression The primary endpoint is consolidation OS. Sample size considerations rest on estimates of MRD subgroup prevalence at the end of induction and operating characteristics establishing the treatment effect within the MRD positive subgroup as primary and MRD negative subgroup as key secondary. The total accrual goal is 1450 patients. Clinical trial information: NCT04566328.


2021 ◽  
Vol 1 (2) ◽  
pp. 35-42
Author(s):  
YURIKA NOGUCHI ◽  
NORIYOSHI IRIYAMA ◽  
HIROMICHI TAKAHASHI ◽  
YOSHIHITO UCHINO ◽  
MASARU NAKAGAWA ◽  
...  

Background/Aim: Here, we investigated whether bortezomib as a maintenance therapy affected outcomes in transplant-ineligible patients with multiple myeloma (MM). Patients and Methods: Following induction therapy with bortezomib, maintenance therapy with bortezomib (1.3 mg/m2) and dexamethasone (20 mg) was administered once or twice every 4 weeks until disease progression. The endpoints of this study were time to next treatment and overall survival. Results: Seventy-six newly diagnosed, transplant-ineligible patients were treated with a bortezomib-based regimen; 28 discontinued induction therapy, 27 did not receive maintenance therapy after induction therapy (the non-maintenance group), and 21 did (the maintenance group). In the three groups, the median times to the next required treatment were 3, 14, and 37 months, respectively. The 3-year overall survival rates were 55%, 69%, and 85%, respectively. There were no significant differences in patient characteristics between the non-maintenance and maintenance groups, except for poorer estimated glomerular filtration rates in the maintenance group. Conclusion: Bortezomib maintenance therapy may be a useful option for transplant-ineligible patients with MM.


2019 ◽  
Vol 19 (10) ◽  
pp. e361
Author(s):  
Lalit Kumar ◽  
Santosh kumar Chellapuram ◽  
Ranjit Sahoo ◽  
Ritu Gupta

2019 ◽  
Vol 145 (2) ◽  
pp. 559-568
Author(s):  
Evangelos Terpos ◽  
Eirini Katodritou ◽  
Argiris Symeonidis ◽  
Flora Zagouri ◽  
Antonis Gerofotis ◽  
...  

2014 ◽  
Vol 167 (4) ◽  
pp. 563-565 ◽  
Author(s):  
Craig B. Reeder ◽  
Donna E. Reece ◽  
Vishal Kukreti ◽  
Joseph R. Mikhael ◽  
Christine Chen ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3017-3017 ◽  
Author(s):  
Michele Cavo ◽  
Antonio Palumbo ◽  
Sara Bringhen ◽  
Antonietta Falcone ◽  
Pellegrino Musto ◽  
...  

Abstract Thalidomide-containing regimens are currently being used as standard initial therapy for both younger and elderly pts with multiple myeloma (MM), but are associated with an increased risk of venous thromboembolism (VTE) which necessitates routine thromboprophylaxis. Controversies exist concerning the best thromboprophylactic regimen to be used in these pts. To address this issue, the Italian Myeloma Network GIMEMA designed a phase III sub-study aimed at prospectively investigating the efficacy and safety of low molecular weight heparin (LMWH) or fixed low-dose warfarin (WAR) or low-dose aspirin (ASA) as prophylaxis against VTE in newly diagnosed MM pts who were randomized to receive primary induction therapy with thalidomide-containing regimens in the context of 2 phase III studies conducted by the same group. In one of these studies, pts with ≤65 years of age were randomly assigned to receive Velcade-Thalidomide-Dexamethasone (VTD) or Thalidomide-Dexamethasone (TD) before autologous transplantation. In the other study, Velcade-Melphalan-Prednisone (VMP) was compared with VMP plus thalidomide (VMPT) for elderly patients aged >65 years. The daily dose of Thalidomide was 200 mg in both VTD and TD, and 50 mg in VMPT. Pts randomized to VTD or TD received a total Dexamethasone dose of 320 mg/cycle, while those assigned to VMP or VMPT were given a total Prednisone dose of 240 mg/m2/cycle. By sub-study design, pts treated on VTD or TD or VMPT were randomly assigned to receive thromboprophylaxis with LMWH (Enoxaparin, 40 mg/d) or WAR (1.25 mg/d) or ASA (100 mg/d) for the duration of induction therapy. At the opposite, pts randomized to VMP did not receive any prophylaxis and were used as controls. Sub-study end points included incidence of VTE, acute cardiovascular events, sudden death, bleeding and any other serious adverse events. At the time of the present analysis, 703 pts who received at least 3 cycles of induction therapy were evaluated. Of these pts, 164 treated on VMP were the control group, while the remaining 539 pts (of whom, 209 treated on VTD, 211 on TD and 119 on VMPT) were randomized to receive either LMWH (n=178) or WAR (n=180) or ASA (n=181). Baseline pts characteristics and risk factors for VTE were comparable in all sub-groups. Overall, the risk of VTE was 3.9% with WAR vs 4.5% with LMWH vs 5.5% with ASA (P values not significant for comparisons between different sub-groups), whereas it was 1.8% among the controls. Median times to onset of VTE for pts treated on LMWH or WAR or ASA were 2.66 vs 2.96 vs 2.10 months, respectively. Pts receiving Velcade-containing regimens (VTD or VMPT) had a VTE frequency in the range of approximately 3%, as compared to 5.8% for pts on TD (P value not significant). The rates of cardiovascular events were 0.6% in each of sub-groups including LMWH, WAR and controls, vs 1.1% for pts treated on ASA. No sudden deaths were reported. The incidence of all grades bleeding was 0.6% with LMWH vs 1.1% with WAR vs 3.3% with ASA (P values not significant for comparisons between different sub-groups), while it was 3.7% among the controls. In conclusion, results of the present analysis show that the overall risk of VTE among sub-groups of pts treated with different thalidomide-containing regimens was not superior to that expected during the natural course of MM. No significant relationship was found between the frequency of VTE and thromboprophylactic regimens, induction treatments (e.g. containing or not Velcade) and age of pts (e.g. young vs elderly). In comparison with LMWH and WAR, there was a higher, albeit marginal, risk of VTE and bleeding complications associated with ASA prophylaxis. Finally, a finding not previously well recognized, fixed low-dose WAR was not inferior to LMWH in reducing the risk of VTE among newly diagnosed MM pts receiving thalidomide-containing regimens. For these pts, LMWH, WAR and ASA are likely to be effective thromboprophylactic regimens.


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 ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5117-5117 ◽  
Author(s):  
Patrizia Tosi ◽  
Elena Zamagni ◽  
Paola Tacchetti ◽  
Giulia Perrone ◽  
Michela Ceccolini ◽  
...  

Abstract Bone disease occurs in approximately 80% of patients with newly diagnosed multiple myeloma (MM) and is caused by the interaction of the neoplastic clone with bone marrow microenvironment, ultimately resulting in an altered balance between bone resorption and bone formation. It has been previously reported that therapies aimed at eradicating the myeloma clone could contribute to decrease bone resorption, even though bone formation remains impaired even in responding patients, due to the use of high-dose steroids. It has been recently demonstrated, both in vitro and in animal models, that Bortezomib improves bone formation by stimulating osteoblasts. In order to test whether this activity was retained also in vivo, we evaluated markers of bone resorption (serum crosslaps) and bone formation (serum osteocalcin-OC and bone alkaline phosphatase - BAP) in a series of patients who were enrolled in the “Bologna 2005” phase III clinical trial at our Center. By study design, after registration patients were randomized to receive three 21-days courses of induction therapy with either VTD (Bortezomib, 1.3 mg/sqm on d 1, 4, 8, and 11, plus Dexamethasone, 40 mg on each day of and after Bortezomib administration plus Thalidomide 200 mg/d from d 1 to 63.) or TD (Thalidomide as in VTD and Dexamethasone 40 mg/d on d 1–4 and 9–12 of every 21-d cycle), prior to stem cell collection and double autologous stem cell transplantation. As of January 2008, 27 patients (19 male and 8 female, median age = 57.5 yrs) entered the sub-study; of these, 15 and 12 patients were randomized in the VTD and TD arm, respectively. At diagnosis, both groups of patients showed a marked increase in serum crosslaps as compared to upper baseline limit (7321±1445pmol/L in the VTD arm and 11140±2576pmol/L in the TD arm) while both OC and BAP were reduced as compared to lower baseline limits. After completion of the induction therapy, serum crosslaps were significantly decreased in both treatment groups (2747±319pmol/L in VTD arm, p=0.007; 3686±1084pmol/L in the TD arm, p=0.0015). In the TD group a significant further reduction in bone formation markers was also observed (42% reduction in serum OC and 30% in BAP, p=0.03 and 0.04 as compared to pre-treatment values); on the contrary, in the VTD arm both OC and BAP were not significantly decreased as compared to baseline values (15% and 11% for OC and BAP, respectively). These data suggest that incorporation of Bortezomib into induction therapy counteracts the inhibitory effects of high-dose steroids on osteoblastogenesis, thus sparing bone formation.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 42-42 ◽  
Author(s):  
Michele Cavo ◽  
Giulia Perrone ◽  
Silvia Buttignol ◽  
Elisabetta Calabrese ◽  
Monica Galli ◽  
...  

Abstract Abstract 42 We prospectively compared thalidomide-dexamethasone (TD) with bortezomib-thalidomide-dexamethasone (VTD) as induction therapy before, and consolidation after, double autologous stem-cell transplantation (ASCT) in patients with newly diagnosed multiple myeloma (MM). Three 21-d cycles of either VTD (V, 1.3 mg/m2 twice-weekly; T, 200 mg/d through d 1 to 63; D, 320 mg/cycle) or TD were given as induction therapy. Consolidation therapy comprised two 35-d cycles of VTD (V, 1.3 mg/m2 once-weekly; T, 100 mg/d through d 1 to 70; D, 320 mg/cycle) or TD. 474 patients randomized to the VTD (n=236) or TD (n=238) arm were analyzed on an intention-to-treat basis for response rate, PFS and OS. Centrally reassessed CR/nCR rate was significantly higher in the VTD compared with the TD arm after all treatment phases, including induction therapy (30% vs 10%, p<0.0001), double autotransplantation (54% vs 42%, p=0.008) and consolidation therapy (60% vs 44%, p=0.001). Best confirmed overall CR/nCR rate was 71% in the VTD arm compared with 52% in the TD arm (p<0.0001); the corresponding values for VGPR or better were 89% vs 72%, respectively (p<0.0001). To evaluate the role of consolidation therapy we performed a per-protocol analysis of 323 patients, 161 treated with VTD and 162 with TD. Overall, upgraded responses with VTD and TD as consolidation therapy were observed in 55% vs 37% of patients, respectively (p=0.01; OR:1.15-3.77). Furthermore, the probability to improve responses from less than CR before consolidation to CR after consolidation was 28% with VTD vs 15% with TD (p=0.02; OR:1.07-4.57) (p=0.003 using the Mc Nemar's test). Post-consolidation molecular detection of minimal residual disease was the objective of a substudy; detailed results are reported in a separate abstract. Briefly, both qualitative and quantitative analyses confirmed the statistically significant superiority of VTD over TD in effecting higher rates of molecular remissions and reducing the burden of residual myeloma cells after ASCT. Any grade 3–4 non-hematologic adverse events were 10% with VTD (peripheral neuropathy: 1.3%, skin rash: 0.6%) vs 12% with TD. With a median follow-up of 31 months, median PFS was 42 months in the TD arm and was not yet reached in the VTD arm (44-month projected rate: 61%) (HR: 0.62 [CI: 0.45–0.87], p=0.006). Superior PFS in the VTD vs TD arm was retained across patient subgroups with poor prognosis, including those with t(4;14) and/or del(17p). Randomization to VTD overcome the adverse influence of t(4;14) on PFS (40-month projected rates: 69% vs 67% according to the presence or absence of this abnormality, respectively; p=0.6). By the opposite, in the TD arm corresponding median PFS values were 24.5 vs 41.5 months, respectively (p=0.01). The small numbers of patients with del(17p) in both arms of the study precluded a statistical comparison with del(17p)-negative group. In a multivariate analysis, variables favorably influencing PFS were beta2-m lower than 3.5 mg/L (HR:0.47; p=0.000), absence of t(4;14) and/or del(17p) (HR:0.52; p=0.000), randomization to VTD arm (HR:0.57; p=0.002), attainment of at least VGPR (HR:0.50; p=0.009) and CR (HR:0.8; p=0.01). No statistically significant difference between the overall treatment protocols was seen in terms of OS, although curves seemed to initially diverge after 40 months (44-month projected rates: 84% vs 74% for VTD and TD arms, respectively). A multivariate analysis showed the independent role of absence of t(4;14) and/or del(17p) (HR:0.42; p=0.003), ISS stage1-2 (HR:0.49; p=0.02) and randomization to VTD (HR:0.53; p=0.04) in prolonging OS. When time-dependent CR entered the model, absence of t(4;14) and/or del(17p) and less advanced ISS stage retained their positive prognostic value; attainment of CR (strictly related to VTD randomization) was an additional favorable variable. In conclusion, in comparison with the TD arm of the study, 1) VTD induction emerges as a new standard of care for maximizing the degree and speedy of tumor reduction in preparation for ASCT; 2) VTD consolidation effected significantly higher rates of upgraded responses, including CR, and of molecular remissions; 3) double ASCT incorporating VTD as induction and consolidation therapy resulted in significantly longer PFS, a benefit confirmed in a multivariate regression analysis and maintained in the subgroup of patients with adverse cytogenetic abnormalities. Disclosures: Cavo: Janssen-Cilag: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Millennium Pharmaceuticals: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees. Off Label Use: Use of bortezomib and thalidomide as induction therapy before, and consolidation after, autologous transplantation in newly diagnosed multiple myeloma. Baccarani:NOVARTIS: Honoraria; BRISTOL MYERS SQUIBB: Honoraria.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3106-3106 ◽  
Author(s):  
Florence Magrangeas ◽  
Philippe Moreau ◽  
Loic Campion ◽  
Herve Avet-Loiseau ◽  
Catherine Guérin ◽  
...  

Abstract Purpose Activation of canonical and noncanonical NF-kB pathways plays a key role in multiple myeloma (MM) pathogenesis. Recent studies have shown that constitutive activation of NF-kB pathways is present in ∼15-20 % of newly diagnosed MM patients. Bortezomib is a potent selective inhibitor of NF-kB activation; however its cytotoxicity is mainly due to inhibition of the noncanonical NF-kB pathway. We have previously shown that the achievement of very good partial response (VGPR) after induction therapy prior to frontline autologous stem cell transplantation (ASCT) was a favourable prognostic factor for progression-free survival (Moreau et al, Blood 2011;117:3041-3044). Our aim was to correlate NF-kB activity, reflected by a gene signature of well-known NF-kB targets, with the response rate achieved with a bortezomib-based induction regimen prior to high-dose therapy and ASCT performed as part of frontline treatment in patients with symptomatic multiple myeloma. Patients and methods One hundred and ninety-nine patients with symptomatic MM were enrolled in the prospective randomized IFM2007-02 trial comparing 4 cycles of bortezomib-dexamethasone (VD) induction therapy versus 4 cycles of the triplet combination bortezomib-thalidomide-dexamethasone (VTD) prior to ASCT. Among these 199 patients, 114 included in the present study were available for gene expression profile testing and therefore analysed for NF-kB activity. For each patient, the NF-kB(10) index, a reliable measure of NF-kB activity in MM tumor cells (Demchenko et al, Blood 2010; 115: 3541-3552) was calculated. NF-kB(10) index is based on a transcription signature of 10 genes: IL2RG, NFKB2,TNFAIP3, NFKBIE, NFKBIA, RELB, CD74, PLEK, MALT1 and WNT10. Gene expression signature was obtained from Affymetrix Exon1.0 normalized data. A high level of NF-kB activity was defined by the NF-kB(10) index found in a cohort of 20 MM patients with biallelic deletions, identified by using Affymetrix SNP6.0 data, that inactivate negative regulators (cIAP1/2) of NF-kB pathways. Response to induction therapy was evaluated according to the IMWG criteria. Results Responses to induction therapy are shown in Table 1. In this subgroup analysis, the CR plus VGPR rates were not statistically different in both arms of the trial: 24 / 54 (44%) in the VTD arm versus 21 / 60 (35%) in the VD arm, P = .35. We subsequently analyzed the correlation between NF-kB activity and response in the whole cohort of 114 patients regardless of induction treatment. We found that the level of NF-kB activity, based NF-kB(10) index, tested as a continuous variable, was strongly correlated with the quality of response, i.e. VGPR or better (P = .007 Wilcoxon test). We also investigated the cut-off value of NF-kB(10) index that could impact the response rate. Since high NF-kB(10) index was found in MM cells with known NF-kB mutations, we calculated the NF-kB(10) index in a control cohort of MM patients deleted in cIAP1/2. Patients of IFM2007-02 trial with a NF-kB(10) index (< 70.5) lower than the index of the control cohort were assigned to the low NF-kB activity group (50/114, 44%); therefore the remaining cases (64/114, 56%) presented with a high NF-kB activity. We found that patients with a reduced NF-kB(10) index displayed a significantly higher response rate (27/50; 54% vs 18/64; 28%, P= .007), as shown in Figure 1, indicating that the vast majority of patients with high NF-kB were not able to achieve at least VGPR. Conclusion Our results show that a low level NF-kB activity is associated with a higher response rate (>VGPR) to bortezomib-based induction regimen. Patients with low NF-kB(10) index represent 44% of the cohort studied. Since NK-kB activity is related to both canonical and noncanonical pathways, and knowing that bortezomib-induced cytotoxicity is mostly due to the inhibition of the noncanonical pathway only, our data strongly suggest that bortezomib should be combined with drug also targeting the canonical pathway in order to induce a high response rate in patients with increased NF-kB activity, as shown in preclinical studies (Fabre et al, Clin Cancer Res. 2012;18:4669-4681. Disclosures: Moreau: CELGENE: Honoraria, Speakers Bureau; JANSSEN: Honoraria, Speakers Bureau. Off Label Use: FRONTLINE TREATMENT WITH CARFIZOMIB. Attal:CELGENE: Honoraria, Speakers Bureau; JANSSEN: Honoraria, Speakers Bureau. Facon:Janssen and Celgene: Speakers Bureau; Millennium, Onyx, Novartis, BMS, Amgen: Membership on an entity's Board of Directors or advisory committees.


Sign in / Sign up

Export Citation Format

Share Document