8 results on '"Ailawadhi S"'
Search Results
2. Health-related quality of life in patients with triple-class exposed relapsed and refractory multiple myeloma treated with idecabtagene vicleucel or standard regimens: patient-reported outcomes from the phase 3, randomised, open-label KarMMa-3 clinical trial.
- Author
-
Delforge M, Patel K, Eliason L, Dhanda D, Shi L, Guo S, Marshall TS, Arnulf B, Cavo M, Nooka A, Manier S, Callander N, Giralt S, Einsele H, Ailawadhi S, Popa McKiver M, Cook M, and Rodríguez-Otero P
- Subjects
- Female, Humans, Male, Middle Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Dexamethasone therapeutic use, Patient Reported Outcome Measures, Quality of Life psychology, Aged, Multiple Myeloma drug therapy, Multiple Myeloma diagnosis, Receptors, Chimeric Antigen therapeutic use, Thalidomide analogs & derivatives
- Abstract
Background: Chimeric antigen receptor T-cell therapy idecabtagene vicleucel (ide-cel) showed significantly improved progression-free survival compared with standard regimens in adults with relapsed and refractory multiple myeloma who had received two to four previous regimens in the ongoing phase 3 KarMMa-3 trial (NCT03651128). This study analysed patient-reported outcomes (PROs), a KarMMa-3 secondary endpoint., Methods: In the randomised, open-label, phase 3 KarMMa-3 trial, 386 patients in hospitals (≥18 years of age, with measurable disease and an Eastern Cooperative Oncology Group performance status score of 0 or 1, who had received two to four previous regimens-including an immunomodulatory agent, a proteasome inhibitor, and daratumumab-and had documented disease progression after receiving their last dose of the last therapy) were randomly assigned to ide-cel (n=254) or standard regimens (daratumumab, pomalidomide, and dexamethasone; daratumumab, bortezomib, and dexamethasone; ixazomib, lenalidomide, and dexamethasone; carfilzomib and dexamethasone; or elotuzumab, pomalidomide, and dexamethasone; n=132). Patients were expected to complete the European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life C30 Questionnaire (QLQ-C30), Multiple Myeloma Module (QLQ-MY20), EQ 5 dimensions (EQ-5D), and EQ-5D visual analogue scale (VAS) at baseline and follow-up timepoints (data cutoff April 18, 2022). PROs included nine prespecified primary domains: EORTC QLQ-C30 GHS-quality of life (QoL), physical functioning, cognitive functioning, fatigue, and pain; QLQ-MY20 disease symptoms and side effects of treatment; and five-level EQ-5D (EQ-5D-5L) index score and EQ-5D visual VAS. Differences in overall least-squares mean changes from baseline to month 20 were analysed using post-hoc constrained longitudinal data analysis. Time to confirmed improvement or deterioration from baseline was analysed using Cox proportional hazard models., Findings: Patients were randomly assigned between May 6, 2019, and April 8, 2022. Overall, the median age was 63 years (IQR 55-68); 151 (39%) patients were female; and 250 (65%) patients were White, 36 (9%) Black or African American, 19 (5%) Hispanic or Latino, 12 (3%) Asian, and seven (2%) of other race. The median follow-up was 18·6 months (IQR 14·0-26·4). PRO compliance was higher than 75% throughout. Overall least-squares mean changes from baseline favoured ide-cel with Hedges' g effect sizes from 0·3 to 0·7 for most domains. Patients in the ide-cel group showed statistically significant and clinically meaningful improvements across the primary PRO domains of interest, with the exception of QLQ-MY20 disease symptoms, side effects of treatment, and EQ-5D-5L index score, which showed improvement across assessment visits but did not exceed the within-group minimally important difference thresholds. The ide-cel group had shorter times to clinically meaningful improvement than the standard regimens group in QLQ-C30 domains except in role functioning, diarrhoea, and financial difficulties; in QLQ-MY20 domains except body image; and in EQ-5D-VAS., Interpretation: Ide-cel offers improved health-related quality of life compared with standard regimens for patients with relapsed and refractory multiple myeloma after previous lines of therapy. The PRO data highlight the extended QoL benefits of a one-time infusion with ide-cel compared with continuous treatment with standard regimens in the treatment of triple-class exposed patients with relapsed and refractory multiple myeloma., Funding: 2seventy bio and Celgene, a Bristol Myers Squibb Company., Competing Interests: Declaration of interests MD reports payment or honoraria for lectures, presentations, speakers’ bureaus, manuscript writing, or educational events from Amgen, Bristol Myers Squibb, GSK, Janssen, Sanofi, Stemline, and Takeda. KP reports consulting fees from AbbVie, Arcellx, Bristol Myers Squibb, Caribou Science, Cellectis, Genentech, Janssen, Karvopharm, Legend Biotech, Merck, Oncopeptides, Pfizer, and Precision Biosciences. BA reports consulting fees from Amgen, Bristol Myers Squibb, and Janssen; and honoraria for presentations, support for attending meetings or travel, and participation on advisory boards for Bristol Myers Squibb, Janssen, and Sanofi. MC reports consulting fees from Bristol Myers Squibb, Janssen, and Sanofi; and payment or honoraria for lectures, presentations, speakers’ bureaus, manuscript writing, or educational events for AbbVie, Bristol Myers Squibb, and Takeda. AN reports honoraria and participation on advisory boards for Adaptative Biotechnologies, Amgen, BeyondSpring Pharmaceuticals, Bristol Myers Squibb, Cellectar Biosciences, GSK, Janssen, Karyopharm, Oncopeptides, ONK Therapeutics, Pfizer, Sanofi, Secura Bio, and Takeda; research funding to the institution from Aduro Biotech, Amgen, Arch Oncology, Bristol Myers Squibb, Cellectis, Genentech, GSK, Janssen, Karyopharm Therapeutics, Kite Pharmaceuticals, Merck, Pfizer, and Takeda; and grants and research support for investigator-initiated studies from Amgen, GSK, Janssen, Merck, and Takeda. SM reports participation on advisory boards for AbbVie, Amgen, Bristol Myers Squibb, GSK, Janssen, Novartis, Pfizer, Regeneron, Roche, Sanofi, and Takeda. SG reports grants or contracts from Amgen, Actinuum, Bristol Myers Squibb, Johnson & Johnson, Miltenyi, Omeros, and Takeda; leadership or fiduciary role on other board, society committee or advocacy group, paid or unpaid, for Amgen, Actinuum, Bristol Myers Squibb, Kite Pharmaceuticals, Janssen, Jazz Phamaceuticals, Johnson & Johnson, Novartis, Spectrum Pharmaceuticals, and Takeda; and ownership of stock in Bristol Myers Squibb. HE reports consulting fees, payment or honoraria for lectures, presentations, speakers’ bureaus, manuscript writing or educational events, support for attending meetings or travel, and participation on a data safety monitoring board or advisory board for Amgen, Bristol Myers Squibb, GSK, Janssen, Novartis, Sanofi, and Takeda; research funding to the institution from Amgen, Bristol Myers Squibb, GSK, Janssen, and Sanofi; and ownership of stock in Bristol Myers Squibb. SA reports consulting fees from BeiGene, Bristol Myers Squibb, Cellectar Biosciences, GSK, Janssen, Pfizer, Sanofi, and Takeda; and research funding to the institution from AbbVie, Amgen, Ascentage, Bristol Myers Squibb, Cellectar Biosciences, GSK, Janssen, Pharmacyclics, and Sanofi. PR-O reports consulting fees from AbbVie, Bristol Myers Squibb, GSK, H3 Pharmaceuticals, Janssen, Oncopeptides, Pfizer, Roche, and Sanofi; payment or honoraria for lectures, presentations, speakers’ bureaus, manuscript writing, or educational events from AbbVie, Bristol Myers Squibb, GSK, Janssen, and Sanofi; support for attending meetings or travel from Pfizer; and participation on a data safety monitoring board or advisory board for Bristol Myers Squibb and Janssen. LE, DD, TSM, and MPM are employee of Bristol Myers Squibb. LS and SG are employees of Evidera. NC declares no competing interests. MC is an employee of Celgene, a Bristol Myers Squibb Company., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Indatuximab ravtansine plus dexamethasone with lenalidomide or pomalidomide in relapsed or refractory multiple myeloma: a multicentre, phase 1/2a study.
- Author
-
Kelly KR, Ailawadhi S, Siegel DS, Heffner LT, Somlo G, Jagannath S, Zimmerman TM, Munshi NC, Madan S, Chanan-Khan A, Lonial S, Chandwani S, Minasyan A, Ruehle M, Barmaki-Rad F, Abdolzade-Bavil A, Rharbaoui F, Herrmann-Keiner E, Haeder T, Wartenberg-Demand A, and Anderson KC
- Subjects
- 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, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Dexamethasone therapeutic use, Immunoconjugates therapeutic use, Lenalidomide therapeutic use, Multiple Myeloma drug therapy, Thalidomide analogs & derivatives
- 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/m
2 , 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., Competing Interests: Declaration of interests KRK received honoraria from Incyte, Bayer, Janssen, Novartis, Celgene, Epizyme, Pharmacyclics, Karyopharm and Gilead, consulting fees from Takeda, AstraZeneca, Sanofi-Aventis, Denovo Biopharma, Verastem, and Amgen, and received research funding from Takeda. SA received consulting fees or honoraria from Celgene, Takeda, Amgen, Janssen, Beigene, GSK, Oncopeptides, and Novartis. DSS had a membership on an entity's advisory committee with Celgene, BMS, GSK, Takeda, Karyopharma, Cellularity, Janssen, Amgen, and Merck. SJ received honorarium and participated in advisory boards for Karyopharm Therapeutics, Legend Biotech, Takeda, Celgene, BMS, and Janssen. NCM was a consultant for Celgene, Merck, Pfizer, Takeda, OncoPep, Janssen and Biotest, and is a part owner of OncoPep. SM received speaker fees from Takeda, Janssen, Amgen, Karyopharm, BMS, and GSK, and received support for attending meetings or travel from Merck, and participated in advisory boards for Amgen, Sanofi, Bristol-Meyers Squibb, Janssen, GSK, and Takeda. AC-K participated in an advisory board for OncoPep. SL received honoraria from Celgene, Takeda, Novartis, Janssen, Amgen, BMS, and GSK, and receive consultancy fees from Celgene, Takeda, Novartis, Janssen, Amgen, BMS, and GSK. MR, FB-R., AA-B, FR, EH-K, TH and AW-D are or were employees of Biotest. KCA received advisory board fees from BMS, Celgene, Gilead, and Millennium Pharmaceuticals, holds a leadership or fiduciary role in Starton, Raqia, and NextRNA, and is the scientific founder of C4 Therapeutics and OncoPep. SC, AM, GS, and TZ have no interests to declare., (Copyright © 2021 Elsevier Ltd. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
4. Cost-effectiveness of Pomalidomide, Carfilzomib, and Daratumumab for the Treatment of Patients with Heavily Pretreated Relapsed-refractory Multiple Myeloma in the United States.
- Author
-
Pelligra CG, Parikh K, Guo S, Chandler C, Mouro J, Abouzaid S, and Ailawadhi S
- Subjects
- Antibodies, Monoclonal therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cost-Benefit Analysis, Dexamethasone therapeutic use, Disease-Free Survival, Drug Costs, Drug Resistance, Neoplasm, Humans, Models, Economic, Multiple Myeloma drug therapy, Oligopeptides therapeutic use, Quality-Adjusted Life Years, Recurrence, Thalidomide economics, Thalidomide therapeutic use, United States, Antibodies, Monoclonal economics, Antineoplastic Combined Chemotherapy Protocols economics, Dexamethasone economics, Multiple Myeloma economics, Oligopeptides economics, Thalidomide analogs & derivatives
- Abstract
Purpose: Pomalidomide plus low-dose dexamethasone (POM-d), daratumumab monotherapy (DARA), and carfilzomib monotherapy (CAR) have been approved for use in the treatment of patients with heavily pretreated relapsed-refractory multiple myeloma (RRMM) in the US, based on findings from the MM-002, SIRIUS, and PX-171-003-A1 studies, respectively. The objective of this study was to assess the cost-effectiveness of POM-d, DARA, and CAR in this patient population from a US payer's perspective., Methods: A cost-effectiveness model was developed to estimate the cost and health outcomes over a 3-year time horizon in 3 health states: progression-free, post-progression, and death. The main efficacy data source was a matching-adjusted indirect comparison using data from the aforementioned studies. Direct medical costs were considered, including: treatment acquisition and administration (initial line and subsequent line), pre- and post-medication, prophylaxis treatment, adverse event management, and health care resource utilization. Sensitivity analyses were conducted. A scenario analysis that assumed equal efficacy across all 3 treatments was conducted. Costs, life-years, and quality-adjusted life-years were estimated and discounted at 3% per annum., Findings: Over 3 years, the use of POM-d was associated with similar life-years and quality-adjusted life-years gained compared with DARA and CAR (incremental: life-years, +0.02 and +0.07, respectively; quality-adjusted life-years, +0.01 and +0.05), and with a cost less than that of DARA (-$8,919) and similar to that of CAR (-$195). Sensitivity analyses illustrated that the results were sensitive to progression-free survival, treatment duration, and drug costs. An equal efficacy scenario resulted in cost-savings relative to those of both DARA and CAR (-$11,779 and -$12,595)., Implications: POM-d may be a cost-effective treatment option relative to DARA or CAR in heavily pretreated patients with RRMM in the US., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
5. Pomalidomide, bortezomib, and dexamethasone for patients with relapsed lenalidomide-refractory multiple myeloma.
- Author
-
Paludo J, Mikhael JR, LaPlant BR, Halvorson AE, Kumar S, Gertz MA, Hayman SR, Buadi FK, Dispenzieri A, Lust JA, Kapoor P, Leung N, Russell SJ, Dingli D, Go RS, Lin Y, Gonsalves WI, Fonseca R, Bergsagel PL, Roy V, Sher T, Chanan-Khan AA, Ailawadhi S, Stewart AK, Reeder CB, Richardson PG, Rajkumar SV, and Lacy MQ
- Subjects
- Aged, Aged, 80 and over, Disease-Free Survival, Female, Humans, Lenalidomide, Male, Maximum Tolerated Dose, Middle Aged, Thalidomide therapeutic use, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Dexamethasone therapeutic use, Multiple Myeloma drug therapy, Thalidomide analogs & derivatives
- Abstract
This phase 1/2 trial evaluated the maximum tolerated doses, safety, and efficacy of pomalidomide, bortezomib, and dexamethasone (PVD) combination in patients with relapsed lenalidomide-refractory multiple myeloma (MM). In phase 1, dose level 1 consisted of pomalidomide (4 mg by mouth on days 1 to 21), IV or subcutaneous bortezomib (1.0 mg/m
2 on days 1, 8, 15, and 22), and dexamethasone (40 mg by mouth on days 1, 8, 15, and 22) given every 28 days. Bortezomib was increased to 1.3 mg/m2 for dose level 2 and adopted in the phase 2 expansion cohort. We describe the results of 50 patients. Objective response rate was 86% (95% confidence interval [CI], 73-94) among all evaluable patients (stringent complete response, 12%; complete response, 10%; very good partial response, 28%; and partial response, 36%) and 100% among high-risk patients. Within a median follow-up of 42 months, 20% remain progression free, 66% are alive, and 4% remain on treatment. Median progression-free survival was 13.7 months (95% CI, 9.6-17.7). The most common toxicities were neutropenia (96%), leukopenia (84%), thrombocytopenia (82%), anemia (74%), and fatigue (72%); however, the majority of these were grade 1 or 2. The most common grade ≥3 toxicities included neutropenia (70%), leukopenia (36%), and lymphopenia (20%). Deep vein thrombosis occurred in 5 patients. In conclusion, PVD is a highly effective combination in lenalidomide-refractory MM patients. Weekly administration of bortezomib enhanced tolerability and convenience. Toxicities are manageable, mostly consisting of mild cytopenias with no significant neuropathy. This trial was registered at www.clinicaltrials.gov as #NCT01212952., (© 2017 by The American Society of Hematology.)- Published
- 2017
- Full Text
- View/download PDF
6. Pharmacoeconomic implications of lenalidomide maintenance therapy in multiple myeloma.
- Author
-
Kim MY, Sposto R, Swaika A, Asano H, Alamgir A, Chanan-Khan A, and Ailawadhi S
- Subjects
- Antineoplastic Agents adverse effects, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Cost-Benefit Analysis, Disease-Free Survival, Economics, Pharmaceutical, Humans, Induction Chemotherapy, Lenalidomide, Maintenance Chemotherapy, Melphalan administration & dosage, Melphalan adverse effects, Prednisone administration & dosage, Prednisone adverse effects, Thalidomide administration & dosage, Thalidomide adverse effects, Thalidomide economics, Antineoplastic Agents administration & dosage, Antineoplastic Combined Chemotherapy Protocols economics, Multiple Myeloma drug therapy, Multiple Myeloma economics, Thalidomide analogs & derivatives
- Abstract
We compared the three arms of the MM-015 randomized phase III clinical trial [melphalan and prednisone (MP), MP plus lenalidomide (MPR), and MPR plus lenalidomide maintenance (MPR-R)] to determine whether the addition of lenalidomide maintenance therapy for primary treatment of multiple myeloma is cost-effective. We used progression-free survival and adverse event data from the MM-015 study for the analysis. Two novel measures of cost-effectiveness termed the Average Cumulative Cost per Patient (ACCP) and the Average Cumulative Cost per Progression-Free Survivor (ACCPFS) were developed for the purpose of this analysis. The ACCP of MP was USD 18,218, compared to USD 167,862 for MPR and USD 309,173 for MPR-R. The ACCPFS was highest with MPR at USD 1,555,443, while MP was USD 313,592 and MPR-R was USD 690,111. MPR-R is superior to MPR in terms of preventing the first progression after initial therapy. However, the addition of lenalidomide to MP in the induction and also in the maintenance setting leads to significant costs., (© 2014 S. Karger AG, Basel.)
- Published
- 2014
- Full Text
- View/download PDF
7. Downregulation of BCL2 by AT-101 enhances the antileukaemic effect of lenalidomide both by an immune dependant and independent manner.
- Author
-
Masood A, Chitta K, Paulus A, Khan AN, Sher T, Ersing N, Miller KC, Manfredi D, Ailawadhi S, Borrelo I, Lee KP, and Chanan-Khan A
- Subjects
- Antibodies, Monoclonal, Murine-Derived therapeutic use, Antineoplastic Agents, Phytogenic therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Down-Regulation drug effects, Down-Regulation immunology, Female, Gene Expression Regulation, Leukemic immunology, Gossypol pharmacology, Gossypol therapeutic use, Humans, Immunologic Factors therapeutic use, Lenalidomide, Male, Rituximab, Thalidomide pharmacology, Thalidomide therapeutic use, Tumor Cells, Cultured, Antineoplastic Agents, Phytogenic pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Gene Expression Regulation, Leukemic drug effects, Gossypol analogs & derivatives, Immunologic Factors pharmacology, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Proto-Oncogene Proteins c-bcl-2 biosynthesis, Proto-Oncogene Proteins c-bcl-2 immunology, Thalidomide analogs & derivatives
- Abstract
Over-expression of anti-apoptotic BCL2 has been reported in chronic lymphocytic leukaemia (CLL), but targeting BCL2 alone did not yield appreciable clinical results. However, it was demonstrated that BCL2 inhibitors enhanced the clinical efficacy of chemo and immunotherapeutics. Lenalidomide, an immunomodulator, is clinically effective in CLL and can enhance the anti-CLL effects of CD20 targeting monoclonal antibody, rituximab. Here, we investigated the mechanism of immune-directed killing of lenalidomide in CLL and evaluated if concurrent targeting of CD20 and BCL2 can enhance this effect. In vitro treatment with lenalidomide enhanced the antibody-mediated cellular cytotoxicity (ADCC) directed by rituximab in autologous leukaemic cells. Furthermore, peripheral blood mononuclear cells obtained from patients after treatment with lenalidomide and rituximab showed increased ADCC in vitro versus control (pre-treatment sample). This effect was further enhanced with pre-treatment of tumour cells with AT-101 (a BH3 mimetic that functions as BCL2 antagonist). Our data suggest that AT-101 in combination with lenalidomide can potentially be an effective therapeutic regimen for CLL., (© 2011 Blackwell Publishing Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
8. Bortezomib in combination with pegylated liposomal doxorubicin and thalidomide is an effective steroid independent salvage regimen for patients with relapsed or refractory multiple myeloma: results of a phase II clinical trial.
- Author
-
Chanan-Khan A, Miller KC, Musial L, Padmanabhan S, Yu J, Ailawadhi S, Sher T, Mohr A, Bernstein ZP, Barcos M, Patel M, Iancu D, Lee K, and Czuczman MS
- Subjects
- Adult, Aged, Bortezomib, Doxorubicin administration & dosage, Female, Humans, Male, Middle Aged, Remission Induction, Salvage Therapy methods, Steroids therapeutic use, Treatment Outcome, Antineoplastic Agents administration & dosage, Boronic Acids administration & dosage, Doxorubicin analogs & derivatives, Multiple Myeloma drug therapy, Polyethylene Glycols administration & dosage, Pyrazines administration & dosage, Thalidomide administration & dosage
- Abstract
Novel agents have demonstrated enhanced efficacy when combined with other antimyeloma agents especially dexamethasone. The steroid doses employed in myeloma regimens are often poorly tolerated. Therefore, in a phase II clinical trial we investigated the efficacy of a steroid-free combination including bortezomib, pegylated liposomal doxorubicin and thalidomide (VDT regimen). Twenty-three patients with relapsed or refractory myeloma or other plasma cell cancers were treated with the VDT regimen. Patient had a median of five prior therapies and 65.2% were refractory to their last regimen. The overall response rates were 55.5% and 22%, respectively. The median progression free survival was 10.9 months (95% CI: 7.3-15.8) and the median overall survival was 15.7 months (95% CI: 9.1-not reached). Fatigue and sensory neuropathy were the most common side effects noted. We observe that VDT is an effective steroid-free regimen with ability to induce durable remission even in patients with refractory myeloma.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.