Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
2.
Am J Hematol ; 96(4): 428-435, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33421178

ABSTRACT

The maximum tolerated dose of the panobinostat and carfilzomib combination in patients with relapsed/refractory multiple myeloma (RRMM) was not reached in our previous dose-escalation study. We report additional dose levels in the phase I/II, single-arm, multicenter, standard 3 + 3 dose-escalation expansion-cohort study (NCT01496118). Patients with RRMM were treated with panobinostat 30 mg, carfilzomib 20/56 mg/m2 (N = 3), or panobinostat 20 mg, carfilzomib 20/56 mg/m2 (N = 33). Treatment cycles lasted 28 days; panobinostat: days 1, 3, 5, 15, 17, 19; carfilzomib: days 1, 2, 8, 9, 15, 16. For dose level 6 (DL 6), median age was 63 years (range, 49-91 years), 60.6% were male, 42.4% were high risk. Patients received a median of two prior therapies (range 1-7); proteasome inhibitors (PI; 100%), immunomodulatory imide drugs (IMiD; 78.8%), and stem cell transplant (36.4%); 48.5%, 51.1%, and 24.2% were refractory to prior PI or prior IMiD treatment or both, respectively. Patients completed a median of seven (range 1-40) treatment cycles. Overall response rate (primary endpoint) of evaluable patients in the expansion cohort (N = 32): 84.4%; clinical benefit rate: 90.6%. With a median follow-up of 26.1 months (range, 0-72.5 months), median (95% CI) progression-free survival, time-to-progression and overall survival of patients was 10.3 (6.1, 13.9), 11.7 (5.6, 14.5), and 44.6 (20.8, N/A) months, respectively. Common adverse events (AEs) included thrombocytopenia (78.8%), nausea (63.6%), fatigue (63.6%), diarrhea (51.5%), and vomiting (51.5%). Seven patients had serious treatment-related AEs. There was one treatment-related death. In conclusion, panobinostat plus carfilzomib is an effective steroid-sparing regimen for RRMM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Salvage Therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Gastrointestinal Diseases/chemically induced , Hematologic Diseases/chemically induced , Humans , Male , Maximum Tolerated Dose , Middle Aged , Oligopeptides/administration & dosage , Oligopeptides/adverse effects , Panobinostat/administration & dosage , Panobinostat/adverse effects , Premedication , Progression-Free Survival
3.
Biol Blood Marrow Transplant ; 24(2): 373-380, 2018 02.
Article in English | MEDLINE | ID: mdl-29051021

ABSTRACT

Imatinib has clinical activity in chronic graft-versus-host disease (cGVHD), a significant complication of allogeneic hematopoietic cell transplant. Nilotinib is a tyrosine kinase inhibitor that targets the same receptors as imatinib but with different affinities. We tested the hypothesis that nilotinib is safe and has clinical activity in cGVHD. Thirty-three participants were enrolled in a phase I/II dose escalation and dose extension clinical trial of nilotinib for the treatment of steroid-refractory or- dependent cGVHD (ClinicalTrials.gov, NCT01155817). We assessed safety, clinical response, and pretreatment anti-platelet-derived growth factor receptor alpha chain (anti-PDGFRA) antibody levels. The 200-mg dose was identified as the maximum tolerated dose and used for the phase II dose extension study. At 6 months the incidence of failure-free survival (FFS), cGVHD progression, and nilotinib intolerance resulting in its discontinuation was 50%, 23%, and 23%, respectively. cGVHD responses in skin, joints, and mouth were observed at 3 and 6 months based on improvement in respective National Institutes of Health organ severity scores. Pretreatment anti-PDGFRA antibody levels ≥ .150 optical density as measured by ELISA correlated with longer FFS time (P < .0005) and trended with time until cGVHD progression (P < .06) but not drug intolerance. Nilotinib may be effective for corticosteroid-resistant or -refractory cGVHD in some patients, but its use is limited by intolerable side effects. Selection of patients with high pretreatment anti-PDGFRA antibody levels might improve the risk-to-benefit ratio of nilotinib and better justify its side effects.


Subject(s)
Antibodies/blood , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/immunology , Adult , Aged , Disease-Free Survival , Female , Graft vs Host Disease , Humans , Male , Middle Aged , Patient Selection , Predictive Value of Tests , Pyrimidines/adverse effects , Pyrimidines/therapeutic use , Young Adult
4.
J Adv Pract Oncol ; 8(7): 705-720, 2017.
Article in English | MEDLINE | ID: mdl-30333933

ABSTRACT

Abstract Tumor lysis syndrome (TLS) is a potentially life-threatening emergency that can develop rapidly after the release of intracellular contents from lysed malignant cells. The advent of novel and targeted therapies that have improved tumor-killing efficacy has the potential to increase the risk of TLS when used as part of front-line therapy. A recent review of TLS risk in patients with hematologic malignancies treated with newer targeted agents highlighted the need to revisit TLS risk stratification and to describe the practical challenges of TLS prevention, treatment, and monitoring. Although this era of rapid development of novel cancer therapies provides new hope for patients with hematologic malignancies, it is essential to be prepared for TLS because monitoring and prophylaxis can almost always prevent severe and life-threatening consequences. Heightened awareness of the development of TLS with novel and targeted agents, accompanied by aggressive hydration and rational, risk-appropriate management, are the keys to successful outcomes.

5.
Ann Hematol ; 95(4): 563-73, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26758269

ABSTRACT

Effective new treatments are now available for patients with hematologic malignancies. However, their propensity to cause tumor lysis syndrome (TLS) has not been systematically examined. A literature search identified published Phase I-III clinical trials of monoclonal antibodies (otlertuzumab, brentuximab, obinutuzumab, ibritumomab, ofatumumab); tyrosine kinase inhibitors (alvocidib [flavopiridol], dinaciclib, ibrutinib, nilotinib, dasatinib, idelalisib, venetoclax [ABT-199]); proteasome inhibitors (oprozomib, carfilzomib); chimeric antigen receptor (CAR) T cells; and the proapoptotic agent lenalidomide. Abstracts from major congresses were also reviewed. Idelalisib and ofatumumab had no reported TLS. TLS incidence was ≤5 % with brentuximab vedotin (for anaplastic large-cell lymphoma), carfilzomib and lenalidomide (for multiple myeloma), dasatinib (for acute lymphoblastic leukemia), and oprozomib (for various hematologic malignancies). TLS incidences were 8.3 and 8.9 % in two trials of venetoclax (for chronic lymphocytic leukemia [CLL]) and 10 % in trials of CAR T cells (for B-cell malignancies) and obinutuzumab (for non-Hodgkin lymphoma). TLS rates of 15 % with dinaciclib and 42 and 53 % with alvocidib (with sequential cytarabine and mitoxantrone) were seen in trials of acute leukemias. TLS mitigation was employed routinely in clinical trials of alvocidib and lenalidomide. However, TLS mitigation strategies were not mentioned or stated only in general terms for many studies of other agents. The risk of TLS persists in the current era of novel and targeted therapy for hematologic malignancies and was seen to some extent with most agents. Our findings underscore the importance of continued awareness, risk assessment, and prevention to reduce this serious potential complication of effective anticancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Drug Delivery Systems/methods , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/drug therapy , Tumor Lysis Syndrome/diagnosis , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Clinical Trials as Topic/methods , Drug Delivery Systems/adverse effects , Humans , Risk Factors , Tumor Lysis Syndrome/etiology
6.
J Hematol Oncol ; 2: 23, 2009 Jun 02.
Article in English | MEDLINE | ID: mdl-19490647

ABSTRACT

Acute myeloid leukemia (AML) is a heterogenous disorder that results from a block in the differentiation of hematopoietic progenitor cells along with uncontrolled proliferation. In approximately 60% of cases, specific recurrent chromosomal aberrations can be identified by modern cytogenetic techniques. This cytogenetic information is the single most important tool to classify patients at their initial diagnosis into three prognostic categories: favorable, intermediate, and poor risk. Currently, favorable risk AML patients are usually treated with contemporary chemotherapy while poor risk AML patients receive allogeneic stem cell transplantation if suitable stem cell donors exist. The largest subgroup of AML patients (aproximately 40%) have no identifiable cytogenetic abnormalities and are classified as intermediate risk. The optimal therapeutic strategies for these patients are still largely unclear. Recently, it is becoming increasingly evident that it is possible to identify a subgroup of poorer risk patients among those with normal cytogenic AML (NC-AML). Molecular risk stratification for NC-AML patients may be possible due to mutations of NPM1, FLT3, MLL, and CEBPalpha as well as alterations in expression levels of BAALC, MN1, ERG, and AF1q. Further prospective studies are needed to confirm if poorer risk NC-AML patients have improved clinical outcomes after more aggressive therapy.


Subject(s)
Genetic Markers , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Adult , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Cytogenetics , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Genetic Markers/physiology , Humans , Leukemia, Myeloid, Acute/pathology , Neoplasm, Residual , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Nucleophosmin , Prognosis , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/physiology
SELECTION OF CITATIONS
SEARCH DETAIL