Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Blood ; 136(22): 2513-2523, 2020 11 26.
Article in English | MEDLINE | ID: mdl-32735641

ABSTRACT

In this phase 2 multicenter study, we evaluated the incorporation of autologous stem cell transplantation (ASCT) into a carfilzomib-lenalidomide-dexamethasone (KRd) regimen for patients with newly diagnosed multiple myeloma (NDMM). Transplant-eligible patients with NDMM received 4 cycles of KRd induction, ASCT, 4 cycles of KRd consolidation, and 10 cycles of KRd maintenance. The primary end point was rate of stringent complete response (sCR) after 8 cycles of KRd with a predefined threshold of ≥50% to support further study. Seventy-six patients were enrolled with a median age of 59 years (range, 40-76 years), and 35.5% had high-risk cytogenetics. The primary end point was met, with an sCR rate of 60% after 8 cycles. Depth of response improved over time. On intent-to-treat (ITT), the sCR rate reached 76%. The rate of minimal residual disease (MRD) negativity using modified ITT was 70% according to next-generation sequencing (<10-5 sensitivity). After median follow-up of 56 months, 5-year progression-free survival (PFS) and overall survival (OS) rates were 72% and 84% for ITT, 85% and 91% for MRD-negative patients, and 57% and 72% for patients with high-risk cytogenetics. For high-risk patients who were MRD negative, 5-year rates were 77% and 81%. Grade 3 to 4 adverse events included neutropenia (34%), lymphopenia (32%), infection (22%), and cardiac events (3%). There was no grade 3 to 4 peripheral neuropathy. Patients with NDMM treated with KRd with ASCT achieved high rates of sCR and MRD-negative disease at the end of KRd consolidation. Extended KRd maintenance after consolidation contributed to deepening of responses and likely to prolonged PFS and OS. Safety and tolerability were manageable. This trial was registered at www.clinicaltrials.gov as #NCT01816971.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Hematopoietic Stem Cell Transplantation , Multiple Myeloma , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Autografts , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Disease-Free Survival , Female , Humans , Lenalidomide/administration & dosage , Lenalidomide/adverse effects , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/mortality , Multiple Myeloma/therapy , Oligopeptides/administration & dosage , Oligopeptides/adverse effects , Progression-Free Survival
2.
Br J Haematol ; 186(4): 549-560, 2019 08.
Article in English | MEDLINE | ID: mdl-31124580

ABSTRACT

Selinexor, an oral Selective Inhibitor of Nuclear Export, targets Exportin 1 (XPO1, also termed CRM1). Non-clinical studies support combining selinexor with proteasome inhibitors (PIs) and corticosteroids to overcome resistance in relapsed/refractory multiple myeloma (RRMM). We conducted a phase I dose-escalation trial of twice-weekly selinexor in combination with carfilzomib and dexamethasone (SKd) to determine maximum tolerated dose in patients with RRMM (N = 21), with an expansion cohort to assess activity in carfilzomib-refractory disease and identify a recommended phase II dose (RP2D). During dose escalation, there was one dose-limiting toxicity (cardiac failure). The RP2D of twice-weekly SKd was selinexor 60 mg, carfilzomib 20/27 mg/m2 and dexamethasone 20 mg. The most common grade 3/4 treatment-emergent adverse events included thrombocytopenia (71%), anaemia (33%), lymphopenia (33%), neutropenia (33%) and infections (24%). Rates of ≥minimal response, ≥partial response and very good partial response were 71%, 48% and 14%, respectively; similar response outcomes were observed for dual-class refractory (PI and immunomodulatory drug)/quad-exposed (carfilzomib, bortezomib, lenalidomide and pomalidomide) patients (n = 17), and patients refractory to carfilzomib in last line of therapy (n = 13). Median progression-free survival was 3·7 months, and overall survival was 22·4 months in the overall population. SKd was tolerable and re-established disease control in RRMM patients, including carfilzomib-refractory patients. Registered at ClinicalTrials.gov (NCT02199665).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cytogenetic Analysis , Dexamethasone/administration & dosage , Drug Resistance, Neoplasm , Female , Humans , Hydrazines/administration & dosage , Male , Middle Aged , Multiple Myeloma/mortality , Neoplasm Staging , Oligopeptides/administration & dosage , Prognosis , Recurrence , Retreatment , Treatment Outcome , Triazoles/administration & dosage
3.
Blood ; 127(22): 2693-700, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27009059

ABSTRACT

Marizomib (MRZ) is a novel, irreversible proteasome inhibitor in clinical development for the treatment of relapsed or relapsed and refractory multiple myeloma (RRMM). MRZ inhibits the 3 proteolytic activities of the 20S proteasome with specificity distinct from bortezomib and carfilzomib. Study NPI-0052-101 Part 1 enrolled relapsed or RRMM patients into an open-label, dose-escalation design to determine the maximum tolerated dose and recommended phase 2 dose (RP2D) of MRZ administered intravenously on 2 different schedules: schedule A (0.025-0.7 mg/m(2) once weekly on days 1, 8, and 15 of 4-week cycles) and schedule B (0.15-0.6 mg/m(2) twice weekly on days 1, 4, 8, and 11 of 3-week cycles; concomitant dexamethasone was allowed with schedule B). Patients had received an average of 4.9 and 7.3 prior treatment regimens (schedules A and B, respectively). MRZ schedule A was administered to 32 patients, and the RP2D was established as 0.7 mg/m(2) infused over 10 minutes. Schedule B was administered to 36 patients, and the RP2D was determined to be 0.5 mg/m(2) infused over 2 hours. The most common (>20% of patients) related adverse events were fatigue, headache, nausea, diarrhea, dizziness, and vomiting. Six patients achieved clinical benefit responses (defined as minimal response or better), including 5 partial responses (1 patient on schedule A and 4 on schedule B; 3 of these 4 patients received concomitant dexamethasone). MRZ was generally well tolerated, and results suggest activity in previously treated RRMM patients. Combination studies using pomalidomide and dexamethasone are now underway. The trial was registered at www.clinicaltrials.gov as #NCT00461045.


Subject(s)
Lactones/administration & dosage , Multiple Myeloma/drug therapy , Pyrroles/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Female , Humans , Lactones/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Multiple Myeloma/mortality , Pyrroles/adverse effects , Recurrence , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/analogs & derivatives
4.
Blood ; 124(7): 1047-55, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-24904120

ABSTRACT

Proteasome inhibition is an effective treatment strategy for multiple myeloma. With improving survival, attention is increasingly focusing on ease of administration and toxicity profile. Ixazomib is an investigational, orally bioavailable 20S proteasome inhibitor. Sixty patients with relapsed and/or refractory multiple myeloma were enrolled on this phase 1 trial to evaluate safety and tolerability and determine the maximum tolerated dose (MTD) of single-agent, oral ixazomib given weekly for 3 of 4 weeks. Upon MTD determination, patients were enrolled to 4 different cohorts based on relapsed/refractory status and prior bortezomib and carfilzomib exposure. The MTD was determined to be 2.97 mg/m(2). Dose-limiting toxicities were grade 3 nausea, vomiting, and diarrhea in 2 patients, and grade 3 skin rash in 1 patient. Common drug-related adverse events were thrombocytopenia (43%), diarrhea (38%), nausea (38%), fatigue (37%), and vomiting (35%). The observed rate of peripheral neuropathy was 20%, with only 1 grade 3 event reported. Nine (18%) patients achieved a partial response or better, including 8 of 30 (27%) evaluable patients treated at the MTD. Pharmacokinetic studies suggested a long terminal half-life of 3.6 to 11.3 days, supporting once-weekly dosing. This trial was registered at www.clinicaltrials.gov as #NCT00963820.


Subject(s)
Boron Compounds/therapeutic use , Drugs, Investigational/therapeutic use , Glycine/analogs & derivatives , Multiple Myeloma/drug therapy , Proteasome Inhibitors/therapeutic use , Administration, Oral , Adult , Aged , Area Under Curve , Boron Compounds/adverse effects , Boron Compounds/pharmacokinetics , Diarrhea/chemically induced , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance, Neoplasm/drug effects , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacokinetics , Fatigue/chemically induced , Female , Glycine/adverse effects , Glycine/pharmacokinetics , Glycine/therapeutic use , Humans , Male , Middle Aged , Nausea/chemically induced , Neoplasm Recurrence, Local , Proteasome Inhibitors/adverse effects , Proteasome Inhibitors/pharmacokinetics , Remission Induction , Thrombocytopenia/chemically induced , Treatment Outcome , Vomiting/chemically induced
5.
Blood ; 118(3): 535-43, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21596852

ABSTRACT

This phase 1/2 trial evaluated combination lenalidomide, bortezomib, pegylated liposomal doxorubicin, and dexamethasone (RVDD) in newly diagnosed multiple myeloma (MM) patients. Patients received RVDD at 4 dose levels, including the maximum tolerated dose (MTD). Patients with a very good partial response or better (≥ VGPR) after cycle 4 proceeded to autologous stem cell transplantation or continued treatment. The primary objectives were MTD evaluation and response to RVDD after 4 and 8 cycles. Seventy-two patients received a median of 4.5 cycles. The MTDs were lenalidomide 25 mg, bortezomib 1.3 mg/m(2), pegylated liposomal doxorubicin 30 mg/m(2), and dexamethasone 20/10 mg, as established with 3-week cycles. The most common adverse events were fatigue, constipation, sensory neuropathy, and infection; there was no treatment-related mortality. Response rates after 4 and 8 cycles were 96% and 95% partial response or better, 57% and 65% ≥ VGPR, and 29% and 35% complete or near-complete response, respectively. After a median follow-up of 15.5 months, median progression-free survival (PFS) and overall survival (OS) were not reached. The estimated 18-month PFS and OS were 80.8% and 98.6%, respectively. RVDD was generally well tolerated and highly active, warranting further study in newly diagnosed MM patients. This trial was registered at www.clinicaltrials.gov as NCT00724568.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Boronic Acids/administration & dosage , Dexamethasone/administration & dosage , Doxorubicin/administration & dosage , Multiple Myeloma/drug therapy , Pyrazines/administration & dosage , Thalidomide/analogs & derivatives , Adult , Aged , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/adverse effects , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Boronic Acids/adverse effects , Bortezomib , Dexamethasone/adverse effects , Disease-Free Survival , Doxorubicin/adverse effects , Female , Follow-Up Studies , Humans , Lenalidomide , Male , Middle Aged , Multiple Myeloma/diagnosis , Prospective Studies , Pyrazines/adverse effects , Survival Rate , Thalidomide/administration & dosage , Thalidomide/adverse effects
6.
Lancet Haematol ; 8(11): e794-e807, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34529955

ABSTRACT

BACKGROUND: Indatuximab ravtansine (BT062) is an antibody-drug conjugate that binds to CD138 and synergistically enhances the antitumor activity of lenalidomide in preclinical models of multiple myeloma. This phase 1/2a study was done to determine the safety, activity, and pharmacokinetics of indatuximab ravtansine in combination with immunomodulatory drugs in patients with relapsed or refractory multiple myeloma. METHODS: This open-label, phase 1/2a study took place at nine hospital sites in the USA. Eligible patients were aged 18 years or older, had relapsed or refractory multiple myeloma, and ECOG performance status or Zubrod score of 2 or below. Patients who received indatuximab ravtansine with lenalidomide and dexamethasone (indatuximab ravtansine plus lenalidomide) had failure of at least one previous therapy. Patients treated with indatuximab ravtansine with pomalidomide and dexamethasone (indatuximab ravtansine plus pomalidomide) had failure of at least two previous therapies (including lenalidomide and bortezomib) and had progressive disease on or within 60 days of completion of their last treatment. In phase 1, patients received indatuximab ravtansine intravenously on days 1, 8, and 15 of each 28-day cycle in escalating dose levels of 80 mg/m2, 100 mg/m2, and 120 mg/m2, with lenalidomide (25 mg; days 1 to 21 every 28 days orally) and dexamethasone (20-40 mg; days 1, 8, 15, and 22 every 28 days). In phase 2, the recommended phase 2 dose of indatuximab ravtansine was given to an expanded cohort of patients in combination with lenalidomide and dexamethasone. The protocol was amended to allow additional patients to be treated with indatuximab ravtansine plus pomalidomide (4 mg; days 1 to 21 every 28 days orally) and dexamethasone, in a more heavily pretreated patient population than in the indatuximab ravtansine plus lenalidomide group. The phase 1 primary endpoint was to determine the dose-limiting toxicities and the maximum tolerated dose (recommended phase 2 dose) of indatuximab ravtansine, and the phase 2 primary endpoint was to describe the objective response rate (ORR; partial response or better) and clinical benefit response (ORR plus minor response). All patients were analysed for safety and all patients with post-treatment response assessments were analysed for activity. This study is registered with ClinicalTrials.gov, number NCT01638936, and is complete. FINDINGS: 64 (86%) of 74 screened patients were enrolled between July 3, 2012, and June 30, 2015. 47 (73%) patients received indatuximab ravtansine plus lenalidomide (median follow-up 24·2 months [IQR 19·9-45·4]) and 17 (27%) received indatuximab ravtansine plus pomalidomide (24·1 months [17·7-36·7]). The maximum tolerated dose of indatuximab ravtansine plus lenalidomide was 100 mg/m2, and defined as the recommended phase 2 dose for indatuximab ravtansine plus pomalidomide. An objective response for indatuximab ravtansine plus lenalidomide was observed in 33 (71·7%) of 46 patients and in 12 (70·6%) of 17 patients in the indatuximab ravtansine plus pomalidomide group. The clinical benefit response for indatuximab ravtansine plus lenalidomide was 85% (39 of 46 patients) and for indatuximab ravtansine plus pomalidomide it was 88% (15 of 17 patients). The most common grade 3-4 adverse events in both groups were neutropenia (14 [22%] of 64 patients), anaemia (10 [16%]), and thrombocytopenia (seven [11%]). Treatment-emergent adverse events (TEAEs) that led to discontinuation occurred in 35 (55%) of the 64 patients. Five (8%) patients with a TEAE had a fatal outcome; none was reported as related to indatuximab ravtansine. INTERPRETATION: Indatuximab ravtansine in combination with immunomodulatory drugs shows preliminary antitumor activity, is tolerated, and could be further evaluated in patients with relapsed or refractory multiple myeloma. FUNDING: Biotest AG.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dexamethasone/therapeutic use , Immunoconjugates/therapeutic use , Lenalidomide/therapeutic use , Multiple Myeloma/drug therapy , Thalidomide/analogs & derivatives , Adult , Aged , Aged, 80 and over , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dexamethasone/adverse effects , Female , Humans , Immunoconjugates/adverse effects , Lenalidomide/adverse effects , Male , Maximum Tolerated Dose , Maytansine/adverse effects , Maytansine/analogs & derivatives , Maytansine/therapeutic use , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Thalidomide/adverse effects , Thalidomide/therapeutic use
7.
J Immunother Cancer ; 8(1)2020 01.
Article in English | MEDLINE | ID: mdl-31940591

ABSTRACT

BACKGROUND: Progression after high-dose melphalan with autologous stem cell transplantation (ASCT) in multiple myeloma (MM) may be due in part to immune dysfunction. Regulatory T (Treg) cells reconstitute rapidly after ASCT and inhibit immune responses against myeloma cells. METHODS: We performed a randomized study to evaluate two methods of Treg depletion in patients with MM undergoing ASCT. No Treg depletion was performed in the control ASCT arm. An anti-CD25 monoclonal antibody (basiliximab 20 mg IV) was administered on day +1 post-ASCT in the in vivo Treg depletion (IVTRD) arm. Tregs were depleted from autologous stem cell (ASC) grafts with anti-CD25 microbeads and the CliniMACS device in the ex vivo Treg depletion (EVTRD) arm. RESULTS: Fifteen patients were enrolled, five in each arm. The conditioning regimen was melphalan 200 mg/m2. Primary objectives included assessments of efficiency of IVTRD/EVTRD, kinetics of Treg depletion and recovery following ASCT, and safety. EVTRD removed 90% of CD4+CD25+ cells from ASC grafts. IVTRD and EVTRD led to reductions in Treg frequency between days +7 and +90 post-transplant compared with the control (p=0.007 and p<0.001, respectively). CONCLUSIONS: IVTRD and EVTRD are feasible and significantly reduce and delay Treg recovery post-ASCT for MM, and serve as a platform for using post-transplant immunotherapies to improve post-ASCT outcomes. TRIAL REGISTRATION NUMBER: NCT01526096.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Multiple Myeloma/therapy , T-Lymphocytes, Regulatory/immunology , Humans , Middle Aged , Multiple Myeloma/immunology , Pilot Projects , Transplantation, Autologous
8.
Clin Lymphoma Myeloma Leuk ; 19(6): 372-380, 2019 06.
Article in English | MEDLINE | ID: mdl-30930134

ABSTRACT

BACKGROUND: Indatuximab ravtansine (BT062) is an antibody-drug conjugate that binds to CD138, which is overexpressed on multiple myeloma (MM) cells. PATIENTS AND METHODS: We report from 2 clinical studies of patients with relapsed and/or refractory MM previously treated with an immunomodulatory drug and a proteasome inhibitor. Single- and multi-dosing schedules were investigated to define dose-limiting toxicities, maximum tolerated dose (MTD), recommended phase II dose, and to describe safety, efficacy, and pharmacokinetics. RESULTS: In the first-in-human study, indatuximab ravtansine was administered to 32 patients on day 1 of each 21-day cycle. The MTD was 160 mg/m2. In the phase I/IIa study, indatuximab ravtansine was administered to 35 patients on days 1, 8, and 15 of each 28-day cycle, and the MTD/recommended phase II dose was 140 mg/m2. Most (88%) adverse events were grade 1 or 2, the most common being diarrhea and fatigue. There was rapid clearance of indatuximab ravtansine and no relevant accumulation. Over 75% of heavily pretreated patients achieved stable disease or better. With the multi-dose schedule, minor and partial responses occurred in 14.7% of patients, the median time to progression was 3 months, and the median overall survival was 26.7 months. CONCLUSION: Our data support further investigation of indatuximab ravtansine as part of a combination regimen for relapsed and/or refractory MM.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immunoconjugates/therapeutic use , Multiple Myeloma/drug therapy , Adult , Aged , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Drug Resistance, Neoplasm , Female , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/adverse effects , Male , Maytansine/administration & dosage , Maytansine/adverse effects , Maytansine/analogs & derivatives , Maytansine/therapeutic use , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/mortality , Neoplasm Grading , Neoplasm Staging , Recurrence , Treatment Outcome
9.
Clin Lymphoma Myeloma Leuk ; 17(9): 555-562, 2017 09.
Article in English | MEDLINE | ID: mdl-28735890

ABSTRACT

BACKGROUND: High risk and low risk multiple myeloma patients follow a very different clinical course as reflected in their PFS and OS. To be clinically useful, methodologies used to identify high and low risk disease must be validated in representative independent clinical data and available so that patients can be managed appropriately. A recent analysis has indicated that SKY92 combined with the International Staging System (ISS) identifies patients with different risk disease with high sensitivity. PATIENTS AND METHODS: Here we computed the performance of eight gene expression based classifiers SKY92, UAMS70, UAMS80, IFM15, Proliferation Index, Centrosome Index, Cancer Testis Antigen and HM19 as well as the combination of SKY92/ISS in an independent cohort of 91 newly diagnosed MM patients. RESULTS: The classifiers identified between 9%-21% of patients as high risk, with hazard ratios (HRs) between 1.9 and 8.2. CONCLUSION: Among the eight signatures, SKY92 identified the largest proportion of patients (21%) also with the highest HR (8.2). Our analysis also validated the combination SKY92/ISS for identification of three classes; low risk (42%), intermediate risk (37%) and high risk (21%). Between low risk and high risk classes the HR is >10.


Subject(s)
Gene Expression Profiling/methods , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Neoplasm Staging/methods , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Male , Middle Aged , Multiple Myeloma/mortality , Prognosis , Proportional Hazards Models
11.
J Clin Oncol ; 22(23): 4816-22, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15570084

ABSTRACT

PURPOSE: Tipifarnib, an orally bioavailable inhibitor of farnesyl transferase, has activity in hematologic malignancies, but the dose required to achieve the proposed biologic end point, inhibition of farnesylation, is unknown. PATIENTS AND METHODS: The impact on post-translational farnesylation was assessed in 42 patients with refractory hematologic malignancies and bone marrow involvement. Tipifarnib was taken orally for 21 days of a 28-day cycle. For cycle 1, patients were randomly assigned to one of four dose levels: 100 mg bid, 200 mg bid, 300 mg bid, and 600 mg bid. In cycle 1, peripheral blood and bone marrow mononuclear cells were analyzed for inhibition of HDJ2 prenylation by Western blot analysis at baseline and on day 21. RESULTS: Twenty-three patients were assessable for analysis of HDJ2 prenylation before and after therapy. Inhibition of farnesylation was noted at all dose levels, although the highest level of inhibition was noted at the 300-mg-bid dose. The inhibition of farnesylation in the peripheral blood correlated with the inhibition in the bone marrow (r = 0.62). Of the 26 patients assessable for clinical activity after cycle 1, three patients had a significant decrease in total blasts count (acute myeloid leukemia in two patients, and chronic myelogenous leukemia in one patient). The inhibition of farnesylation was greater in the three responders than the nonresponders (P = .03). CONCLUSION: Farnesylation as measured by HDJ2 analysis was inhibited at all dose levels administered. Clinical activity may correlate with the degree of farnesylation inhibition, rather than dose of tipifarnib, and escalation beyond 300 mg bid might not result in additional clinical activity.


Subject(s)
Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Quinolones/administration & dosage , Quinolones/pharmacokinetics , Salvage Therapy , Administration, Oral , Aged , Aged, 80 and over , Analysis of Variance , Biological Availability , Blotting, Western , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Hematologic Neoplasms/mortality , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/mortality , Quinolones/adverse effects , Risk Assessment , Statistics, Nonparametric , Survival Analysis , Treatment Outcome
13.
Cancer Chemother Pharmacol ; 54(6): 553-61, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15349753

ABSTRACT

PURPOSE: Gemcitabine and cladribine (2CdA) are nucleoside analogues that decrease DNA synthesis via inhibition of ribonucleotide reductase; the combination could be additive or synergistic. We conducted a dose escalation study to establish the maximum tolerable doses (MTD) of gemcitabine and 2CdA when given in combination in patients with advanced hematologic malignancies and to describe the toxicity profile of this combination. PATIENTS AND METHODS: A total of 45 patients with advanced hematologic diseases were enrolled into two groups. Group A had adequate baseline hematopoiesis, defined as absolute neutrophil count (ANC) >1 x 10(9)/l and platelet count >50 x 10(9)/l. Group B did not meet these criteria. Hematologic dose-limiting toxicity (DLT) for group A was defined as grade 4 neutropenia or thrombocytopenia lasting >28 days; group B was not evaluated for hematologic toxicity. Nonhematologic DLT was defined similarly for both groups. Death occurring during the first cycle of treatment was considered a DLT event only if it was related to drug toxicity. Gemcitabine was administered as a 4-h intravenous infusion once every 28 days. 2CdA was administered over 1 h daily for the first 3 days of each 28-day cycle. RESULTS: The MTD was not reached in either group. Myelosuppression was common, but not dose-limiting. Febrile neutropenia and infections were also common, particularly in group B, and judged in most cases to be due to bone marrow failure at baseline. Nonhematologic toxicities were generally mild, and skin rash, the most frequently observed, was dose-limiting in one patient enrolled in each group. Four deaths (three during the first cycle of treatment) occurred at the highest dose level tested in group B (gemcitabine 5000 mg/m2 and 2CdA 16 mg/m2). Although only one of these deaths was dose-limiting by stated criteria, this dose level did not appear to be safely tolerated in this patient population. Several responses were observed in patients with Hodgkin's disease. CONCLUSIONS: The combination of gemcitabine and 2CdA is feasible in patients with hematologic malignancies. Phase II studies of this combination should be considered, particularly in patients with Hodgkin's disease.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cladribine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/administration & dosage , Hematologic Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cladribine/adverse effects , Deoxycytidine/adverse effects , Drug Administration Schedule , Fatigue , Feasibility Studies , Female , Humans , Male , Middle Aged , Neutropenia/chemically induced , Thrombocytopenia/chemically induced , Gemcitabine
14.
Cancer Chemother Pharmacol ; 73(1): 35-42, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24149943

ABSTRACT

PURPOSE: A phase 1 study evaluated the QTc prolongation potential of siltuximab, a chimeric, anti-interleukin-6 mAb, in patients with monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM), or low-volume MM. METHODS: Patients with baseline QTcF and QTcB ≤ 500 ms, QRS < 100 ms, PR < 200 ms and no significant cardiac disease received siltuximab 15 mg/kg q3w, the highest dosage used in clinical studies, for 4 cycles. Twelve-lead ECGs obtained at multiple time points pre- and post-infusion at cycles 1 and 4 were evaluated by central cardiology laboratory. No effect on QTc interval was concluded if the upper limit of least square (LS) mean 90 % CI for QTc change from baseline at each time point was <20 ms. RESULTS: An effect on QTc prolongation was ruled out, as the upper bound of 90 % CI was <10 ms at each time point in 27 evaluable patients (13 MGUS, 13 SMM, 1 low-volume MM) with no differences between disease types. Maximum mean QTc increase from baseline occurred 3 h after cycle 1 infusion (QTcF = 3.2 [LS mean 90 % CI -0.01, 6.45] ms; QTcB = 2.7 [-0.69, 6.14] ms). At all other time points, mean QTcF and QTcB increase from baseline was ≤1.5 ms and upper bound 90 % CI was ≤5.1 ms. Twenty patients had mostly low-grade AEs, including nausea, fatigue (20 % each); thrombocytopenia, headache (each 13 %); dyspnea, leukopenia, neutropenia, paresthesia, abnormal hepatic function, URTI (each 10 %). Three MGUS patients achieved 50 % M-protein reduction. There was no association between siltuximab pharmacokinetics and QTc interval. CONCLUSIONS: Siltuximab did not affect the QTc interval. Overall safety was similar to other single-agent siltuximab studies.


Subject(s)
Antibodies, Monoclonal/adverse effects , Electrocardiography/drug effects , Monoclonal Gammopathy of Undetermined Significance/drug therapy , Multiple Myeloma/drug therapy , Adult , Aged , Antibodies, Monoclonal/pharmacokinetics , Humans , Middle Aged
15.
J Clin Oncol ; 30(16): 1960-5, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22291084

ABSTRACT

PURPOSE: To evaluate the maximum-tolerated dose (MTD), safety, and efficacy of elotuzumab in combination with bortezomib in patients with relapsed or relapsed and refractory multiple myeloma (MM). PATIENTS AND METHODS: Elotuzumab (2.5, 5.0, 10, or 20 mg/kg intravenously [IV]) and bortezomib (1.3 mg/m(2) IV) were administered on days 1 and 11 and days 1, 4, 8, and 11, respectively, in 21-day cycles by using a 3 + 3 dose-escalation design. Patients with stable disease or better after four cycles could continue treatment until disease progression or unexpected toxicity. Responses were assessed during each cycle by using European Group for Blood and Marrow Transplantation (EBMT) criteria. RESULTS: Twenty-eight patients with a median of two prior therapies were enrolled; three patients each received 2.5, 5.0, and 10 mg/kg of elotuzumab and 19 received 20 mg/kg (six during dose escalation and 13 during an expansion phase). No dose-limiting toxicities were observed during cycle 1 of the dose-escalation phase, and the MTD was not reached up to the maximum planned dose of 20 mg/kg. The most frequent grade 3 to 4 adverse events (AEs) were lymphopenia (25%) and fatigue (14%). Two elotuzumab-related serious AEs of chest pain and gastroenteritis occurred in one patient. An objective response (a partial response or better) was observed in 13 (48%) of 27 evaluable patients and in two (67%) of three patients refractory to bortezomib. Median time to progression was 9.46 months. CONCLUSION: The combination of elotuzumab and bortezomib was generally well-tolerated and showed encouraging activity in patients with relapsed/refractory MM.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boronic Acids/administration & dosage , Multiple Myeloma/drug therapy , Pyrazines/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bortezomib , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Recurrence
16.
Leuk Lymphoma ; 50(4): 559-65, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19373653

ABSTRACT

Upregulation of the Bcl-2 antiapoptotic protein is reported to be associated with aggressive clinical course in multiple myeloma. Oblimersen sodium is a bcl-2 antisense oligonucleotide complementary to the first six codons of the open-reading frame of bcl-2 mRNA that can decrease transcription of Bcl-2 protein and increase myeloma cell susceptibility to cytotoxic agents. In this phase III randomised trial, we investigated in patients with relapsed/refractory myeloma whether addition of oblimersen to dexamethasone improved clinical outcomes vs. dexamethasone alone. Two hundred and twenty-four patients were randomised to receive either oblimersen/dexamethasone (N = 110) or dexamethasone alone (N = 114). The primary endpoint was time to tumor progression (TTP). Final results of this study demonstrated no significant differences between the two groups in TTP or objective response rate. The oblimersen/dexamethasone regimen was generally well tolerated with fatigue, fever and nausea, the most common adverse events reported.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dexamethasone/therapeutic use , Multiple Myeloma/drug therapy , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Drug Administration Schedule , Fatigue/chemically induced , Female , Fever/chemically induced , Humans , Male , Middle Aged , Multiple Myeloma/pathology , Nausea/chemically induced , Oligonucleotides, Antisense/administration & dosage , Thionucleotides/administration & dosage , Treatment Outcome
17.
J Urol ; 170(4 Pt 1): 1141-5, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14501711

ABSTRACT

PURPOSE: Combination gemcitabine and 5-fluorouracil (5-FU) may have activity in metastatic renal cell cancer. To identify patient subgroups most likely to benefit and compare survival to that in previously described patient series long-term survival as a function of known and suspected prognostic variables was determined. MATERIALS AND METHODS: The survival status of 153 patients with metastatic renal cell cancer treated on 1 phase I and 4 phase II trials of gemcitabine/5-FU based regimens was updated. Univariate and multivariate Cox proportional hazards models were constructed using multiple imputation for missing values. RESULTS: Performance status, prior nephrectomy, number of metastatic sites, decreased albumin and elevated alkaline phosphatase were highly prognostic for survival. Sarcomatoid differentiation and hypercalcemia had borderline prognostic significance. Using a previous described prognostic model that divided patients into 3 risk groups survival in those treated with gemcitabine/5-FU in this series was consistently better than survival in similar patients treated in the Motzer series. CONCLUSIONS: Previously described prognostic factors for survival in metastatic renal cancer were confirmed. There is no clearly identifiable group that is most likely to benefit from a gemcitabine/5-FU regimen but there is a continued suggestion that this regimen provides a modest improvement over historical chemotherapy approaches.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/mortality , Deoxycytidine/analogs & derivatives , Kidney Neoplasms/drug therapy , Kidney Neoplasms/mortality , Antimetabolites, Antineoplastic/administration & dosage , Carcinoma, Renal Cell/pathology , Deoxycytidine/administration & dosage , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Humans , Kidney Neoplasms/pathology , Male , Prognosis , Survival Rate , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL