Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Clin Lymphoma Myeloma Leuk ; 24(5): e205-e216, 2024 May.
Article in English | MEDLINE | ID: mdl-38453615

ABSTRACT

OBJECTIVES: To report healthcare resource utilization (HCRU) and safety outcomes in systemic light chain (AL) amyloidosis from the EMN23 study. MATERIALS AND METHODS: The retrospective, observational, multinational EMN23 study included 4,480 patients initiating first-line treatment for AL amyloidosis in 2004-2018 and assessed, among other objectives, HCRU and safety outcomes. HCRU included hospitalizations, examinations, and dialysis; safety included serious adverse events (SAEs) and adverse events of special interest (AESIs). Data were descriptively analyzed by select prognostic factors (e.g., cardiac staging by Mayo2004/European) for 2004-2010 and 2011-2018. A cost-of-illness analysis was conducted for the UK and Spain. RESULTS: HCRU/safety and dialysis data were extracted for 674 and 774 patients, respectively. Of patients with assessed cardiac stage (2004-2010: 159; 2011-2018: 387), 67.9% and 61.0% had ≥ 1 hospitalization, 56.0% and 51.4% had ≥ 1 SAE, and 31.4% and 28.9% had ≥ 1 AESI across all cardiac stages in 2004-2010 and 2011-2018, respectively. The per-patient-per-year length of hospitalization increased with disease severity (cardiac stage). Of patients with dialysis data (2004-2010: 176; 2011-2018: 453), 23.9% and 14.8% had ≥ 1 dialysis session across all cardiac stages in 2004-2010 and 2011-2018, respectively. The annual cost-of-illness was estimated at €40,961,066 and €31,904,386 for the UK and Spain, respectively; dialysis accounted for ∼28% (UK) and ∼35% (Spain) of the total AL amyloidosis costs. CONCLUSIONS: EMN23 showed that the burden of AL amyloidosis is substantial, highlighting the need for early disease diagnosis and effective treatments targeting the underlying pathology.


Subject(s)
Cost of Illness , Immunoglobulin Light-chain Amyloidosis , Humans , Retrospective Studies , Male , Female , Immunoglobulin Light-chain Amyloidosis/therapy , Immunoglobulin Light-chain Amyloidosis/economics , Aged , Europe , Middle Aged , Health Resources/statistics & numerical data , Health Resources/economics , Patient Acceptance of Health Care/statistics & numerical data , Health Care Costs/statistics & numerical data , Aged, 80 and over
2.
Am J Hematol ; 92(3): 244-250, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28006855

ABSTRACT

There are currently no direct head-to-head clinical trials evaluating bortezomib-melphalan-prednisone (VMP) versus lenalidomide and low-dose dexamethasone (Rd). VMP (257 cases) and Rd (222 cases) arms of two randomized phase III trials were employed to assess the treatment influence on outcome in untreated elderly MM patients. Progression free survival (PFS) and overall survival (OS) were the primary and secondary end-points, respectively, and were investigated according to treatments administered over a 60-months follow-up period. While VMP significantly reduced the disease progression rate between enrolment and 12 months of follow-up, no difference between the two schedules was found between 12 and 32 months. After 32 months, Rd-treated patients had a lower incidence of disease progression. A statistically significant higher OS rate was seen in the VMP arm, which was maintained after data adjustment for potential confounders. Both approaches showed acceptable toxicity profiles. The profound tumor reduction by VMP over Rd justifies the initial higher PFS rate in favor of the bortezomib schedule, while the Rd regimen overcomes this evident initial drawback in reducing the tumor burden by long-term drug administration, gaining a subsequent improved disease control. VMP is associated with a significant reduced risk of death. This study may help physicians make a more informed therapy choice.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Aged , Bortezomib/administration & dosage , Dexamethasone/administration & dosage , Disease-Free Survival , Female , Humans , Lenalidomide , Male , Melphalan/administration & dosage , Multiple Myeloma/mortality , Prednisone/administration & dosage , Prospective Studies , Survival Rate , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Treatment Outcome , Tumor Burden
3.
Lancet Haematol ; 3(4): e196-204, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27063978

ABSTRACT

BACKGROUND: The first-in-class small molecule inhibitor OTX015 (MK-8628) specifically binds to bromodomain motifs BRD2, BRD3, and BRD4 of bromodomain and extraterminal (BET) proteins, inhibiting them from binding to acetylated histones, which occurs preferentially at super-enhancer regions that control oncogene expression. OTX015 is active in haematological preclinical entities including leukaemia, lymphoma, and myeloma. We aimed to establish the recommended dose of OTX015 in patients with haematological malignancies. We report the results from a cohort of patients with lymphoma or multiple myeloma (non-leukaemia cohort). METHODS: In this dose-escalation, open-label, phase 1 study, we recruited patients from seven university hospital centres (in France [four], Switzerland [one], UK [one], and Italy [one]). Adult patients with non-leukaemia haematological malignancies who had disease progression on standard therapies were eligible to participate. Patients were treated with oral OTX015 once a day continuously over five doses (10 mg, 20 mg, 40 mg, 80 mg, and 120 mg), using a conventional 3 + 3 design, with allowance for evaluation of alternative administration schedules. The primary endpoint was dose-limiting toxicity (DLT) in the first treatment cycle (21 days). Secondary objectives were to evaluate safety, pharmacokinetics, and preliminary clinical activity of OTX015. The study is ongoing and is registered with ClinicalTrials.gov, number NCT01713582. FINDINGS: Between Feb 4, 2013, and Sept 5, 2014, 45 patients (33 with lymphoma and 12 with myeloma), with a median age of 66 years (IQR 55-72) and a median of four lines of prior therapy (IQR 3-5), were enrolled and treated. No DLTs were observed in the doses up to and including 80 mg once a day (first three patients). We then explored a schedule of 40 mg twice a day (21 of 21 days). DLTs were reported in five of six patients receiving OTX015 at this dose and schedule (all five patients had grade 4 thrombocytopenia). We explored various schedules at 120 mg once a day but none was tolerable, with DLTs of thrombocytopenia, gastrointestinal events (diarrhoea, vomiting, dysgeusia, mucositis), fatigue, and hyponatraemia in 11 of 18 evaluable patients. At this point, the Safety Monitoring Committee decided to establish the feasibility of 80 mg once a day on a continuous basis, and four additional patients were enrolled at this dose. DLTs (grade 4 thrombocytopenia) was noted in two of the patients. In light of these DLTs and other toxicities noted at 120 mg, the dose of 80 mg once a day was selected, although on a schedule of 14 days on, 7 days off. Common toxic effects reported in the study were thrombocytopenia (43 [96%] patients), anaemia (41 [91%]), neutropenia (23 [51%]), diarrhoea (21 [47%]), fatigue (12 [27%]), and nausea (11 [24%]). Grade 3-4 adverse events were infrequent other than thrombocytopenia (26 [58%]). OTX015 plasma peak concentrations and areas under the concentration versus time curve increased proportionally with dose. Trough concentrations increased less than proportionally at lower doses, but reached or exceeded the in-vitro active range at 40 mg twice a day and 120 mg once a day. Three patients with diffuse large B-cell lymphoma achieved durable objective responses (two complete responses at 120 mg once a day, and one partial response at 80 mg once a day), and six additional patients (two with diffuse large B-cell lymphoma, four with indolent lymphomas) had evidence of clinical activity, albeit not meeting objective response criteria. INTERPRETATION: The once-daily recommended dose for oral, single agent oral OTX015 in patients with lymphoma is 80 mg on a 14 days on, 7 days off schedule, for phase 2 studies. OTX015 is under evaluation in expansion cohorts using this intermittent administration (14 days every 3 weeks) to allow for recovery from toxic effects. FUNDING: Oncoethix GmbH (a wholly owned subsidiary of Merck Sharp & Dohme Corp).


Subject(s)
Acetanilides/therapeutic use , Antineoplastic Agents/therapeutic use , Heterocyclic Compounds, 3-Ring/therapeutic use , Lymphoma/drug therapy , Multiple Myeloma/drug therapy , Acetanilides/administration & dosage , Aged , Antineoplastic Agents/administration & dosage , Drug Administration Schedule , Female , France , Heterocyclic Compounds, 3-Ring/administration & dosage , Humans , Italy , Male , Maximum Tolerated Dose , Middle Aged , Switzerland , United Kingdom
4.
Blood ; 127(9): 1102-8, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26729895

ABSTRACT

Lenalidomide-dexamethasone improved outcome in newly diagnosed elderly multiple myeloma patients. We randomly assigned 662 patients who were age ≥65 years or transplantation-ineligible to receive induction with melphalan-prednisone-lenalidomide (MPR) or cyclophosphamide-prednisone-lenalidomide (CPR) or lenalidomide plus low-dose dexamethasone (Rd). The primary end point was progression-free survival (PFS) in triplet (MPR and CPR) vs doublet (Rd) lenalidomide-containing regimens. After a median follow-up of 39 months, the median PFS was 22 months for the triplet combinations and 21 months for the doublet (P = .284). The median overall survival (OS) was not reached in either arms, and the 4-year OS was 67% for the triplet and 58% for the doublet arms (P = .709). By considering the 3 treatment arms separately, no difference in outcome was detected among MPR, CPR, and Rd. The most common grade ≥3 toxicity was neutropenia: 64% in MPR, 29% in CPR, and 25% in Rd patients (P < .0001). Grade ≥3 nonhematologic toxicities were similar among arms and were mainly infections (6.5% to 11%), constitutional (3.5% to 9.5%), and cardiac (4.5% to 6%), with no difference among the arms. In conclusion, in the overall population, the alkylator-containing triplets MPR and CPR were not superior to the alkylator-free doublet Rd, which was associated with lower toxicity. This study was registered at www.clinicaltrials.gov as #NCT01093196.


Subject(s)
Multiple Myeloma/diagnosis , Multiple Myeloma/drug therapy , Thalidomide/analogs & derivatives , Aged , Aged, 80 and over , Demography , Disease-Free Survival , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Lenalidomide , Male , Middle Aged , Thalidomide/adverse effects , Thalidomide/therapeutic use , Treatment Outcome
5.
Leuk Lymphoma ; 57(3): 537-56, 2016.
Article in English | MEDLINE | ID: mdl-26445358

ABSTRACT

Novel agents, such as immunomodulantory drugs (IMiDs) and proteasome inhibitors (PI), have significantly improved overall survival of multiple myeloma (MM) patients. Yet, MM remains an incurable disease, relapse inevitably occurs and patients tend to become resistant to subsequent treatments. This led to the evaluation of new treatment strategies. The recent development of monoclonal antibodies is changing the treatment algorithm of MM by increasing the therapeutic armamentarium. Elotuzumab and Daratumumab were shown to be very effective and are likely to be soon approved by the FDA. Other antibodies are in pre-clinical or early clinical phases of evaluation and further investigation and more robust data are needed. This review will give an overview of the most active monoclonal antibodies against MM.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Multiple Myeloma/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Clinical Trials as Topic , Complement System Proteins/immunology , Humans , Immunomodulation/drug effects , Molecular Targeted Therapy , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Signal Transduction/drug effects , Tumor Escape/drug effects , Tumor Escape/immunology
6.
Lancet Oncol ; 16(16): 1617-29, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26596670

ABSTRACT

BACKGROUND: High-dose melphalan plus autologous stem-cell transplantation (ASCT) is the standard approach in transplant-eligible patients with newly diagnosed myeloma. Our aims were to compare consolidation with high-dose melphalan plus ASCT versus chemotherapy (cyclophosphamide and dexamethasone) plus lenalidomide, and maintenance with lenalidomide plus prednisone versus lenalidomide alone. METHODS: We did an open-label, randomised, multicentre, phase 3 study at 59 centres in Australia, Czech Republic, and Italy. We enrolled transplant-eligible patients with newly diagnosed myeloma aged 65 years or younger. Patients received a common induction with four 28-day cycles of lenalidomide (25 mg, days 1-21) and dexamethasone (40 mg, days 1, 8, 15, and 22) and subsequent chemotherapy with cyclophosphamide (3 g/m(2)) followed by granulocyte colony-stimulating factor for stem-cell mobilisation and collection. Using a 2 × 2 partial factorial design, we randomised patients to consolidation with either chemotherapy plus lenalidomide (six cycles of cyclophosphamide [300 mg/m(2), days 1, 8, and 15], dexamethasone [40 mg, days 1, 8, 15, and 22], and lenalidomide [25 mg, days 1-21]) or two courses of high-dose melphalan (200 mg/m(2)) and ASCT. We also randomised patients to maintenance with lenalidomide (10 mg, days 1-21) plus prednisone (50 mg, every other day) or lenalidomide alone. A simple randomisation sequence was used to assign patients at enrolment into one of the four groups (1:1:1:1 ratio), but the treatment allocation was disclosed only when the patient reached the end of the induction and confirmed their eligibility for consolidation. Both the patient and the treating clinician did not know the consolidation and maintenance arm until that time. The primary endpoint was progression-free survival assessed by intention-to-treat. The trial is ongoing and some patients are still receiving maintenance. This study is registered at ClinicalTrials.gov, number NCT01091831. FINDINGS: 389 patients were enrolled between July 6, 2009, and May 6, 2011, with 256 eligible for consolidation (127 high-dose melphalan and ASCT and 129 chemotherapy plus lenalidomide) and 223 eligible for maintenance (117 lenalidomide plus prednisone and 106 lenalidomide alone). Median follow-up was 52·0 months (IQR 30·4-57·6). Progression-free survival during consolidation was significantly shorter with chemotherapy plus lenalidomide compared with high-dose melphalan and ASCT (median 28·6 months [95% CI 20·6-36·7] vs 43·3 months [33·2-52·2]; hazard ratio [HR] for the first 24 months 2·51, 95% CI 1·60-3·94; p<0·0001). Progression-free survival did not differ between maintenance treatments (median 37·5 months [95% CI 27·8-not evaluable] with lenalidomide plus prednisone vs 28·5 months [22·5-46·5] with lenalidomide alone; HR 0·84, 95% CI 0·59-1·20; p=0·34). Fewer grade 3 or 4 adverse events were recorded with chemotherapy plus lenalidomide than with high-dose melphalan and ASCT; the most frequent were haematological (34 [26%] of 129 patients vs 107 [84%] of 127 patients), gastrointestinal (six [5%] vs 25 [20%]), and infection (seven [5%] vs 24 [19%]). Haematological serious adverse events were reported in two (2%) patients assigned chemotherapy plus lenalidomide and no patients allocated high-dose melphalan and ASCT. Non-haematological serious adverse events were reported in 13 (10%) patients assigned chemotherapy plus lenalidomide and nine (7%) allocated high-dose melphalan and ASCT. During maintenance, adverse events did not differ between groups. The most frequent grade 3 or 4 adverse events were neutropenia (nine [8%] of 117 patients assigned lenalidomide plus prednisone vs 14 [13%] of 106 allocated lenalidomide alone), infection (eight [8%] vs five [5%]), and systemic toxicities (seven [6%] vs two [2%]). Non-haematological serious adverse events were reported in 13 (11%) patients assigned lenalidomide plus prednisone versus ten (9%) allocated lenalidomide alone. Four patients died because of adverse events, three from infections (two during induction and one during consolidation) and one because of cardiac toxic effects. INTERPRETATION: Consolidation with high-dose melphalan and ASCT remains the preferred option in transplant-eligible patients with multiple myeloma, despite a better toxicity profile with chemotherapy plus lenalidomide. FUNDING: Celgene.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation , Multiple Myeloma/therapy , Prednisone/therapeutic use , Thalidomide/analogs & derivatives , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Australia , Chemotherapy, Adjuvant , Cyclophosphamide/therapeutic use , Czech Republic , Dexamethasone/therapeutic use , Disease Progression , Disease-Free Survival , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/mortality , Humans , Italy , Kaplan-Meier Estimate , Lenalidomide , Maintenance Chemotherapy , Male , Melphalan/therapeutic use , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/mortality , Prednisone/adverse effects , Proportional Hazards Models , Risk Factors , Thalidomide/adverse effects , Thalidomide/therapeutic use , Time Factors , Transplantation, Autologous , Treatment Outcome
7.
Blood ; 124(1): 63-9, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24855212

ABSTRACT

This multicenter, open-label phase 2 trial determined the safety and efficacy of carfilzomib, a novel and irreversible proteasome inhibitor, in combination with cyclophosphamide and dexamethasone (CCyd) in patients with newly diagnosed multiple myeloma (NDMM) ≥65 years of age or who were ineligible for autologous stem cell transplantation. Patients (N = 58) received CCyd for up to 9 28-day cycles, followed by maintenance with carfilzomib until progression or intolerance. After a median of 9 CCyd induction cycles (range 1-9), 95% of patients achieved at least a partial response, 71% achieved at least a very good partial response, 49% achieved at least a near complete response, and 20% achieved stringent complete response. After a median follow-up of 18 months, the 2-year progression-free survival and overall survival rates were 76% and 87%, respectively. The most frequent grade 3 to 5 toxicities were neutropenia (20%), anemia (11%), and cardiopulmonary adverse events (7%). Peripheral neuropathy was limited to grades 1 and 2 (9%). Fourteen percent of patients discontinued treatment because of adverse events, and 21% of patients required carfilzomib dose reductions. In summary, results showed high complete response rates and a good safety profile. This trial was registered at clinicaltrials.gov as #NCT01346787.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Aged , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Multiple Myeloma/mortality , Oligopeptides/administration & dosage , Oligopeptides/adverse effects , Treatment Outcome
8.
Haematologica ; 99(2): 232-42, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24497560

ABSTRACT

Multiple myeloma management has undergone profound changes in the past thanks to advances in our understanding of the disease biology and improvements in treatment and supportive care approaches. This article presents recommendations of the European Myeloma Network for newly diagnosed patients based on the GRADE system for level of evidence. All patients with symptomatic disease should undergo risk stratification to classify patients for International Staging System stage (level of evidence: 1A) and for cytogenetically defined high- versus standard-risk groups (2B). Novel-agent-based induction and up-front autologous stem cell transplantation in medically fit patients remains the standard of care (1A). Induction therapy should include a triple combination of bortezomib, with either adriamycin or thalidomide and dexamethasone (1A), or with cyclophosphamide and dexamethasone (2B). Currently, allogeneic stem cell transplantation may be considered for young patients with high-risk disease and preferably in the context of a clinical trial (2B). Thalidomide (1B) or lenalidomide (1A) maintenance increases progression-free survival and possibly overall survival (2B). Bortezomib-based regimens are a valuable consolidation option, especially for patients who failed excellent response after autologous stem cell transplantation (2A). Bortezomib-melphalan-prednisone or melphalan-prednisone-thalidomide are the standards of care for transplant-ineligible patients (1A). Melphalan-prednisone-lenalidomide with lenalidomide maintenance increases progression-free survival, but overall survival data are needed. New data from the phase III study (MM-020/IFM 07-01) of lenalidomide-low-dose dexamethasone reached its primary end point of a statistically significant improvement in progression-free survival as compared to melphalan-prednisone-thalidomide and provides further evidence for the efficacy of lenalidomide-low-dose dexamethasone in transplant-ineligible patients (2B).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Multiple Myeloma/diagnosis , Multiple Myeloma/therapy , Autografts , Clinical Trials, Phase III as Topic
9.
J Clin Oncol ; 32(7): 634-40, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24449241

ABSTRACT

PURPOSE: Bortezomib-melphalan-prednisone (VMP) has improved overall survival in multiple myeloma. This randomized trial compared VMP plus thalidomide (VMPT) induction followed by bortezomib-thalidomide maintenance (VMPT-VT) with VMP in patients with newly diagnosed multiple myeloma. PATIENTS AND METHODS: We randomly assigned 511 patients who were not eligible for transplantation to receive VMPT-VT (nine 5-week cycles of VMPT followed by 2 years of VT maintenance) or VMP (nine 5-week cycles without maintenance). RESULTS: In the initial analysis with a median follow-up of 23 months, VMPT-VT improved complete response rate from 24% to 38% and 3-year progression-free-survival (PFS) from 41% to 56% compared with VMP. In this analysis, median follow-up was 54 months. The median PFS was significantly longer with VMPT-VT (35.3 months) than with VMP (24.8 months; hazard ratio [HR], 0.58; P < .001). The time to next therapy was 46.6 months in the VMPT-VT group and 27.8 months in the VMP group (HR, 0.52; P < .001). The 5-year overall survival (OS) was greater with VMPT-VT (61%) than with VMP (51%; HR, 0.70; P = .01). Survival from relapse was identical in both groups (HR, 0.92; P = .63). In the VMPT-VT group, the most frequent grade 3 to 4 adverse events included neutropenia (38%), thrombocytopenia (22%), peripheral neuropathy (11%), and cardiologic events (11%). All of these, except for thrombocytopenia, were significantly more frequent in the VMPT-VT patients. CONCLUSION: Bortezomib and thalidomide significantly improved OS in multiple myeloma patients not eligible for transplantation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Boronic Acids/administration & dosage , Boronic Acids/adverse effects , Bortezomib , Disease-Free Survival , Female , Follow-Up Studies , Humans , Induction Chemotherapy , Kaplan-Meier Estimate , Maintenance Chemotherapy , Male , Melphalan/administration & dosage , Melphalan/adverse effects , Middle Aged , Neutropenia/chemically induced , Peripheral Nervous System Diseases/chemically induced , Prednisone/administration & dosage , Prednisone/adverse effects , Pyrazines/administration & dosage , Pyrazines/adverse effects , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thrombocytopenia/chemically induced , Treatment Outcome
10.
Br J Haematol ; 164(6): 841-50, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24450886

ABSTRACT

Abnormal activation of MET/HGF (Hepatocyte Growth Factor) pathway has been described in several tumours and increased HGF plasmatic levels have been detected in patients with aggressive multiple myeloma (MM). MET and HGF mRNA expression was investigated in 105 samples of purified plasma cells derived from newly diagnosed MM patients treated with bortezomib-based induction therapy. Gene expression was compared with response to therapy and clinical outcome. MET gene copy number was also evaluated. MET mRNA expression was higher in CD138(+) than in CD138(-) cells (median 76·90 vs. 11·24; P = 0·0009). Low MET mRNA expression characterized patients with better response (complete response or very good partial response) compared to other patients (median 56·10 vs. 134·83; P = 0·0006). After a median follow-up of 50 months, patients with high MET mRNA expression displayed a worse progression-free survival (PFS; P = 0·0029) and overall survival (OS; P = 0·0023) compared to those with low MET mRNA levels. Patients with both high MET mRNA expression and high ß2-microglobulin level (>5·5 mg/l) had further worse median PFS (P < 0·0001) and OS (P < 0·0001). Patients carrying 4 MET gene copies (8 out of 82, 9·8%) also had a short PFS. High MET mRNA expression identifies patients with dismal PFS and OS and the combination with high ß2-microglobulin further characterizes patients with worse outcome.


Subject(s)
Hepatocyte Growth Factor/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Female , Hepatocyte Growth Factor/biosynthesis , Hepatocyte Growth Factor/blood , Hepatocyte Growth Factor/genetics , Humans , Male , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Am J Hematol ; 89(4): 355-62, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24273190

ABSTRACT

Novel agents in combination with melphalan and prednisone (MP) significantly improved progression-free survival (PFS) and overall survival (OS) in multiple myeloma (MM). Randomized trials comparing MP plus bortezomib (VMP) versus MP plus thalidomide (MPT) are lacking. Nine hundred and fifty-six elderly (>65 years) newly diagnosed MM patients from six European randomized trials were retrospectively analyzed and matched for age, albumin, and beta2-microglobulin at diagnosis, 296 patients were selected from the VMP groups, and 294 from MPT. Complete response rate was 21% in the VMP patients and 13% in the MPT patients (P = 0.007). After a median follow-up of 34 months (range, 1-92), VMP significantly prolonged both PFS (median 32.5 vs. 22.9 months, HR 0.65; 95% CI 0.52-0.82; P < 0.001) and OS (median 79.7 vs. 45.1 months, HR 0.44; 95% CI 0.32-0.59; P < 0.001) in comparison with MPT. The benefit in terms of OS of the VMP group was quite similar among patients with different risk factors defined by sex, ISS, ECOG performance status, or serum creatinine but not among patients ≥ 75 years. Multivariate analysis confirmed that VMP was an independent predictor of longer PFS and OS. In a control-case matched analysis, PFS and OS were prolonged in patients who received VMP in comparison with those treated with MPT.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Boronic Acids/administration & dosage , Boronic Acids/adverse effects , Bortezomib , Case-Control Studies , Disease-Free Survival , Drug Evaluation , Female , Hematologic Diseases/chemically induced , Humans , Kaplan-Meier Estimate , Male , Melphalan/administration & dosage , Melphalan/adverse effects , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Nervous System Diseases/chemically induced , Prednisone/administration & dosage , Prednisone/adverse effects , Prognosis , Proportional Hazards Models , Pyrazines/administration & dosage , Pyrazines/adverse effects , Randomized Controlled Trials as Topic , Retrospective Studies , Thalidomide/administration & dosage , Thalidomide/adverse effects , Treatment Outcome
12.
Blood ; 122(16): 2799-806, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-23954889

ABSTRACT

We performed a phase 1/2 trial to determine the maximum tolerated dose (MTD) of pomalidomide and to explore its efficacy when combined with cyclophosphamide-prednisone in relapsed/refractory myeloma patients. Pomalidomide was given at 1 to 2.5 mg/d, cyclophosphamide at 50 mg every other day, prednisone at 50 mg every other day, for 6 28-day cycles, followed by pomalidomide-prednisone maintenance therapy. Thromboprophylaxis was recommended. Sixty-nine patients were enrolled, 55 received the MTD (2.5 mg/d) and were evaluated. Best responses included complete response in 3 patients (5%), very good partial response in 10 (18%), partial response in 15 (27%), minimal response in 11 (20%), stable disease in 15 (27%), and progressive disease in 1 (3%), for an overall response rate of 51%. The median time-to-response was 1.83 months. After a median follow-up of 14.8 months, median progression-free survival was 10.4 months and 1-year overall survival was 69%. At the MTD, grade 3 to 4 toxicities included anemia (9%), thrombocytopenia (11%), neutropenia (42%), neurologic events (7%), dermatologic events (7%), and thromboembolism (2%). Grade 3 to 5 infections occurred in 5 patients (9%). Five patients (9%) discontinued treatment for toxicity. New grade 3 to 4 adverse events were low during maintenance. Pomalidomide-cyclophosphamide-prednisone is safe and effective in relapsed/refractory myeloma patients. This trial was registered at www.clinicaltrials.gov as #NCT01166113.


Subject(s)
Cyclophosphamide/administration & dosage , Multiple Myeloma/drug therapy , Prednisone/administration & dosage , Thalidomide/analogs & derivatives , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Recurrence , Thalidomide/administration & dosage , Time Factors , Treatment Outcome
13.
Expert Opin Drug Metab Toxicol ; 9(10): 1371-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23834482

ABSTRACT

INTRODUCTION: The development of novel agents, such as immunomodulatory drugs and proteasome inhibitors, has led to a considerable increment in the response rate (RR) and outcomes for multiple myeloma (MM) patients. Unfortunately, MM patients will inevitably relapse and become resistant to new drugs. This led to the continuous development of novel agents. Carfilzomib is a second-generation proteasome inhibitor, demonstrating promising results in relapsed/refractory (RR) and newly diagnosed (ND) MM patients. AREAS COVERED: Herein, the authors review Phase I and II trials on carfilzomib for the treatment of MM. They also describe the profile of the drug during Phase I escalating doses and evaluate the efficacy of carfilzomib both alone and in combination. Finally, the authors also review and discuss the carfilzomib safety profile. EXPERT OPINION: Clinical trials (Phases I and II) with carfilzomib, used both as single agent or in combination with other therapies, established the maximum tolerated dose and recommended schedule of administration. Preliminary data showed that it had a high efficacy and a good safety profile both in RRMM and NDMM patients. Carfilzomib seems to be effective in patients previously treated with bortezomib. Future Phase II and III studies will better define the role of carfilzomib in the treatment of MM as well as its optimum dose.


Subject(s)
Multiple Myeloma/drug therapy , Oligopeptides/pharmacokinetics , Boronic Acids , Bortezomib , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Oligopeptides/administration & dosage , Proteasome Inhibitors , Pyrazines
14.
Blood ; 122(8): 1376-83, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23775712

ABSTRACT

A sequential approach including bortezomib induction, intermediate-dose melphalan, and autologous stem cell transplantation (ASCT), followed by lenalidomide consolidation-maintenance, has been evaluated. Efficacy and safety data have been analyzed on intention-to-treat and results updated. Newly diagnosed myeloma patients 65 to 75 years of age (n = 102) received 4 cycles of bortezomib-pegylated liposomal doxorubicin-dexamethasone, tandem melphalan (100 mg/m(2)) followed by ASCT (MEL100-ASCT), 4 cycles of lenalidomide-prednisone consolidation (LP), and lenalidomide maintenance (L) until disease progression. The complete response (CR) rate was 33% after MEL100-ASCT, 48% after LP and 53% after L maintenance. After a median follow-up of 66 months, median time-to-progression (TTP) was 55 months and median progression-free survival 48 months. Median overall survival (OS) was not reached, 5-year OS was 63%. In CR patients, median TTP was 70 months and 5-year OS was 83%. Median survival from relapse was 28 months. Death related to adverse events (AEs) occurred in 8/102 patients during induction or transplantation. Rate of death related to AEs was higher in patients ≥70 years compared with younger (5/26 vs 3/76, P = .024). Bortezomib-induction followed by ASCT and lenalidomide consolidation-maintenance is a valuable option for elderly myeloma patients, with the greatest benefit in those younger than 70 years of age.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boronic Acids/administration & dosage , Melphalan/administration & dosage , Multiple Myeloma/therapy , Pyrazines/administration & dosage , Stem Cell Transplantation/methods , Thalidomide/analogs & derivatives , Aged , Bortezomib , Dexamethasone/administration & dosage , Disease Progression , Disease-Free Survival , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Drug Administration Schedule , Female , Humans , Lenalidomide , Male , Middle Aged , Multiple Myeloma/mortality , Polyethylene Glycols/administration & dosage , Recurrence , Thalidomide/administration & dosage , Transplantation, Autologous/methods , Treatment Outcome
15.
Article in English | MEDLINE | ID: mdl-23714504

ABSTRACT

Hematologic malignancies are a common cause of morbidity and mortality among older adults, who represent the majority of patients diagnosed with these diseases. Treatment options and disease outcomes have improved in recent years because of the development of novel treatment strategies and the design of elderly-specific clinical trials. Despite this, extrapolation of clinical trial data to patients routinely seen in practice is challenging because of the presence of multimorbidity and functional impairments. Individualized treatment decision making requires not only an understanding of underlying tumor biology but also careful estimation of an older patient's anticipated ability to withstand the stresses of therapy. This article will discuss approaches to standardizing patient assessment strategies and tailoring therapeutic decisions for older adults with hematologic malignancies with a focus on acute myeloid leukemia (AML), allogeneic bone marrow transplantation, multiple myeloma (MM), and chronic lymphocytic leukemia (CLL).


Subject(s)
Bone Marrow Transplantation , Hematologic Neoplasms/therapy , Precision Medicine , Aged , Aged, 80 and over , Comorbidity , Geriatric Assessment , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/epidemiology , Humans , Induction Chemotherapy , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Leukemia, Myeloid, Acute/therapy , Molecular Diagnostic Techniques , Multiple Myeloma/therapy , Risk Factors , Transplantation, Homologous , Treatment Outcome
16.
J Clin Oncol ; 29(8): 986-93, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21282540

ABSTRACT

PURPOSE: In patients with myeloma, thalidomide significantly improves outcomes but increases the risk of thromboembolic events. In this randomized, open-label, multicenter trial, we compared aspirin (ASA) or fixed low-dose warfarin (WAR) versus low molecular weight heparin (LMWH) for preventing thromboembolism in patients with myeloma treated with thalidomide-based regimens. PATIENTS AND METHODS: A total of 667 patients with previously untreated myeloma who received thalidomide-containing regimens and had no clinical indication or contraindication for a specific antiplatelet or anticoagulant therapy were randomly assigned to receive ASA (100 mg/d), WAR (1.25 mg/d), or LMWH (enoxaparin 40 mg/d). A composite primary end point included serious thromboembolic events, acute cardiovascular events, or sudden deaths during the first 6 months of treatment. RESULTS: Of 659 analyzed patients, 43 (6.5%) had serious thromboembolic events, acute cardiovascular events, or sudden death during the first 6 months (6.4% in the ASA group, 8.2% in the WAR group, and 5.0% in the LMWH group). Compared with LMWH, the absolute differences were +1.3% (95% CI, -3.0% to 5.7%; P = .544) in the ASA group and +3.2% (95% CI, -1.5% to 7.8%; P = .183) in the WAR group. The risk of thromboembolism was 1.38 times higher in patients treated with thalidomide without bortezomib. Three major (0.5%) and 10 minor (1.5%) bleeding episodes were recorded. CONCLUSION: In patients with myeloma treated with thalidomide-based regimens, ASA and WAR showed similar efficacy in reducing serious thromboembolic events, acute cardiovascular events, and sudden deaths compared with LMWH, except in elderly patients where WAR showed less efficacy than LMWH.


Subject(s)
Anticoagulants/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aspirin/therapeutic use , Cardiovascular Diseases/prevention & control , Enoxaparin/therapeutic use , Fibrinolytic Agents/therapeutic use , Multiple Myeloma/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Thromboembolism/prevention & control , Warfarin/therapeutic use , Aged , Anticoagulants/adverse effects , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Aspirin/adverse effects , Cardiovascular Diseases/etiology , Cardiovascular Diseases/mortality , Enoxaparin/adverse effects , Female , Fibrinolytic Agents/adverse effects , Hemorrhage/chemically induced , Humans , Italy , Male , Middle Aged , Multiple Myeloma/complications , Multiple Myeloma/mortality , Platelet Aggregation Inhibitors/adverse effects , Risk Assessment , Risk Factors , Thalidomide/administration & dosage , Thromboembolism/etiology , Thromboembolism/mortality , Time Factors , Treatment Outcome , Warfarin/adverse effects
17.
Br J Haematol ; 152(5): 579-92, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21241278

ABSTRACT

Signalling through the interleukin (IL)-6 pathway induces proliferation and drug resistance of multiple myeloma cells. We therefore sought to determine whether the IL-6-neutralizing monoclonal antibody siltuximab, formerly CNTO 328, could enhance the activity of melphalan, and to examine some of the mechanisms underlying this interaction. Siltuximab increased the cytotoxicity of melphalan in KAS-6/1, INA-6, ANBL-6, and RPMI 8226 human myeloma cell lines (HMCLs) in an additive-to-synergistic manner, and sensitized resistant RPMI 8226.LR5 cells to melphalan. These anti-proliferative effects were accompanied by enhanced activation of drug-specific apoptosis in HMCLs grown in suspension, and in HMCLs co-cultured with a human-derived stromal cell line. Siltuximab with melphalan enhanced activation of caspase-8, caspase-9, and the downstream effector caspase-3 compared with either of the single agents. This increased induction of cell death occurred in association with enhanced Bak activation. Neutralization of IL-6 also suppressed signalling through the phosphoinositide 3-kinase/Akt pathway, as evidenced by decreased phosphorylation of Akt, p70 S6 kinase and 4E-BP1. Importantly, the siltuximab/melphalan regimen demonstrated enhanced anti-proliferative effects against primary plasma cells derived from patients with myeloma, monoclonal gammopathy of undetermined significance, and amyloidosis. These studies provide a rationale for translation of siltuximab into the clinic in combination with melphalan-based therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Interleukin-6/antagonists & inhibitors , Multiple Myeloma/pathology , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Melphalan/administration & dosage , Melphalan/pharmacology , Multiple Myeloma/metabolism , Neoplasm Proteins/metabolism , Plasma Cells/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
18.
Am J Blood Res ; 1(2): 190-204, 2011.
Article in English | MEDLINE | ID: mdl-22432080

ABSTRACT

The treatment of multiple myeloma has undergone important changes in the last few years. The use of novel agents, such as the immunomodulatory drugs thalidomide and lenalidomide, and the proteasome inhibitor bortezomib, has increased the treatment options available and significantly improved the outcome of this rare disease. Several trials have shown the advantages linked to the use of novel agents both in young patients, who are considered eligible for transplantation, and elderly patients, who are considered transplant ineligible. In the non-transplant setting, novel agent-containing regimens have replaced the traditional melphalan-prednisone approach. Preliminary data also support the role of consolidation and maintenance therapy to further improve outcomes. An appropriate management of side effects is fundamental for the success of the treatment, and outcome should always be balanced against the toxicity profile associated with the regimen used. This review provides an overview of the latest strategies including novel agents used to treat elderly patients with multiple myeloma.

19.
Haematologica ; 95(7): 1144-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20053869

ABSTRACT

BACKGROUND: Defibrotide is a novel orally bioavailable polydisperse oligonucleotide with anti-thrombotic and anti-adhesive effects. In SCID/NOD mice, defibrotide showed activity in human myeloma xenografts. This phase I/II study was conducted to identify the most appropriate dose of defibrotide in combination with melphalan, prednisone and thalidomide in patients with relapsed and relapsed/refractory multiple myeloma, and to determine its safety and tolerability as part of this regimen. DESIGN AND METHODS: This was a phase I/II, multicenter, dose-escalating, non-comparative, open label study. Oral melphalan was administered at a dose of 0.25 mg/kg on days 1-4, prednisone at a dose of 1.5 mg/kg also on days 1-4 and thalidomide at a dose of 50-100 mg/day continuously. Defibrotide was administered orally at three dose-levels: 2.4, 4.8 or 7.2 g on days 1-4 and 1.6, 3.2, or 4.8 g on days 5-35. RESULTS: Twenty-four patients with relapsed/refractory multiple myeloma were enrolled. No dose-limiting toxicity was observed. In all patients, the complete response plus very good partial response rate was 9%, and the partial response rate was 43%. The 1-year progression-free survival and 1-year overall survival rates were 34% and 90%, respectively. The most frequent grade 3-4 adverse events included neutropenia, thrombocytopenia, anemia and fatigue. Deep vein thrombosis was reported in only one patient. CONCLUSIONS: This combination of melphalan, prednisone and thalidomide together with defibrotide showed anti-tumor activity with a favorable tolerability. The maximum tolerated dose of defibrotide was identified as 7.2 g p.o. on days 1-4 followed by 4.8 g p.o. on days 5-35. Further trials are needed to confirm the role of this regimen and to evaluate the combination of defibrotide with new drugs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Dose-Response Relationship, Drug , Humans , Melphalan/administration & dosage , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Polydeoxyribonucleotides/administration & dosage , Prednisone/administration & dosage , Salvage Therapy/methods , Thalidomide/administration & dosage , Treatment Outcome
20.
Cancer J ; 15(6): 494-501, 2009.
Article in English | MEDLINE | ID: mdl-20010169

ABSTRACT

Maintenance therapy after remission achievement is a question still open in multiple myeloma (MM). Steroids and interferon-alpha failed to demonstrate a clear benefit in term of survival. Thalidomide, lenalidomide, and bortezomib have shown to be effective and safe drugs for the treatment of both newly diagnosed and relapsed MM, leading to explore their efficacy also in maintenance setting. Thalidomide seems to be a good choice for patients with low-risk MM and for those who achieved less than very good partial remission after induction treatment. Lenalidomide and bortezomib are still under investigation and a longer follow-up is needed for confirming their role as maintenance treatment. As shown by recent clinical trials, thalidomide and bortezomib are more indicated as consolidation agent, increasing the complete remission/very good partial remission rate. Considering their toxicity profile, first of all peripheral neuropathy and, in case of thalidomide, the lack of correlation between cumulative dose and outcome, a limited administration is suggested. In contrast, lenalidomide showed a low toxicity profile and a benefit from prolonged treatment, making the drug one of the best choices for maintenance treatment.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Multiple Myeloma/drug therapy , Pyrazines/therapeutic use , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Bortezomib , Dexamethasone/therapeutic use , Drug Therapy, Combination , Humans , Lenalidomide , Melphalan/therapeutic use , Prednisolone/therapeutic use , Protease Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL