Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
BMJ ; 364: l1097, 2019 Mar 13.
Article in English | MEDLINE | ID: mdl-30867145
2.
Am J Hematol ; 90(11): 1055-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26284823

ABSTRACT

Lenalidomide is manageable and effective in multiple myeloma, particularly in elderly patients. Surprisingly, the combination of lenalidomide with rituximab produced clinically significant anemia at 25 mg/day for 21/28 days, the highest possible dose, in Waldenström's Macroglobulinemia (WM). We aimed to determine the maximum tolerated dose (MTD) of single agent lenalidomide and determine its impact on WM. RV-WM-0426 is a multicenter dose escalation open label phase 1/2 study of lenalidomide in relapsed/refractory WM (RRWM). Lenalidomide was given orally 21/28 days per cycle for 1 year, at escalated dose of 15 to 20 mg during phase 1 to determine the MTD; the phase 2 part was conducted at the MTD. Seventeen RRWM patients were included. The MTD was established at 15 mg/day 21/28. By ITT analysis, the overall response rate was 29%. With a median follow-up of 36 months, median TTP was 16 months (95% CI 5.5-26), the 5-year OS was 91%. The most frequent adverse events ≥ grade 3 at 15 mg were 14% anemia and 43% neutropenia. The MTD of lenalidomide is 15 mg/day 21/28 days in RRWM. Lenalidomide is active in the treatment of RRWM and the safety profile appears manageable. Future studies may look into combinations of lenalidomide and continuous dosing.


Subject(s)
Antineoplastic Agents/administration & dosage , Immunologic Factors/administration & dosage , Thalidomide/analogs & derivatives , Waldenstrom Macroglobulinemia/drug therapy , Administration, Oral , Aged , Aged, 80 and over , Anemia/chemically induced , Anemia/pathology , Antineoplastic Agents/adverse effects , Drug Administration Schedule , Drug Dosage Calculations , Female , Humans , Immunologic Factors/adverse effects , Lenalidomide , Male , Maximum Tolerated Dose , Middle Aged , Neutropenia/chemically induced , Neutropenia/pathology , Recurrence , Survival Analysis , Thalidomide/administration & dosage , Thalidomide/adverse effects , Treatment Outcome , Waldenstrom Macroglobulinemia/mortality , Waldenstrom Macroglobulinemia/pathology
3.
Cytotherapy ; 15(7): 861-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23731764

ABSTRACT

BACKGROUND AIMS: This study aimed to characterize the immune effectors contained in apheresis samples obtained from patients with grafts mobilized with plerixafor and granulocyte colony-stimulating factor (G-CSF) (P+G) compared with grafts mobilized with G-CSF alone (G). METHODS: Aliquots of apheresis samples were obtained from 36 patients with malignant diseases after mobilization with G (n = 18) or P+G (n = 18). The phenotype and cytokine secretion profile of T cell and dendritic cell subsets were characterized by multicolor cytometry including intracellular cytokine staining. RESULTS: In grafts collected after mobilization with P+G, there was a significantly higher percentage of CD3(+) T cells compared with samples collected after mobilization with G alone. On a functional level, a significant increase of interferon-γ and tumor necrosis factor-α secreting CD8(+) T cells was observed in the P+G group compared with the G group. CD4(+)Foxp3(+) regulatory T cells were similar in both groups but exhibited a lower expression of inducible costimulatory molecule and a significantly higher expression of CD127 in the P+G group. Myeloid dendritic cells (MDCs) and BDCA3(+) dendritic cells were similar in both groups. In contrast, plasmacytoid dendritic cells (PDCs) (CD123(+)BDCA2(+)HLA-DR(+)) were significantly increased in the P+G grafts, leading to a higher PDC-to-MDC ratio. PDCs mobilized by P+G displayed different functional markers--a higher percentage of ILT7(+) PDCs and decreased expression of CD86--suggesting a potential regulatory capacity of PDCs mobilized by P+G. CONCLUSIONS: Grafts mobilized with P+G exhibited major different functional features compared with grafts mobilized with G alone, suggesting that such grafts may have an impact on patient outcome after autologous stem cell transplantation.


Subject(s)
Dendritic Cells/cytology , Granulocyte Colony-Stimulating Factor/metabolism , Heterocyclic Compounds/metabolism , Peripheral Blood Stem Cell Transplantation , Adolescent , Adult , Antigens, Surface/metabolism , Benzylamines , Blood Component Removal/methods , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Child , Child, Preschool , Cyclams , Dendritic Cells/metabolism , Female , Forkhead Transcription Factors/metabolism , Granulocyte Colony-Stimulating Factor/administration & dosage , HLA-DR Antigens/immunology , Heterocyclic Compounds/administration & dosage , Humans , Interferon-gamma/metabolism , Male , Middle Aged , Thrombomodulin , Transplants/drug effects , Transplants/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Clin Cancer Res ; 13(20): 6010-8, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17947462

ABSTRACT

PURPOSE: Intrinsic activation of nuclear factor kappaB (NF-kappaB) characterizes various hematologic malignancies. In this study, we specifically address the role of NF-kappaB blockade in mediated antimyeloma activity using the IkappaB kinase-2 pharmacologic inhibitor, AS602868. EXPERIMENTAL DESIGN: Human myeloma cell lines (n = 16) and primary myeloma cells (n = 10) were tested for their sensitivity to AS602868 in terms of proliferation and apoptosis. Both in vitro and in vivo experiments were conducted. Functional mechanisms regarding the apoptotic pathways triggered by AS602868 were studied. The potential proapoptotic synergy between AS602868 and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) was also evaluated. RESULTS: Our results show that AS602868 efficiently targeted the canonical NF-kappaB pathway in myeloma cells and potently inhibited their growth in inducing apoptosis through Bax and caspase-3 activation. AS602868 also induced apoptosis in primary myeloma cells even in the presence of bone marrow mononuclear cells. Moreover, the IkappaB kinase-2 inhibitor targeted the paracrine effect on the bone marrow environment. Indeed, it decreased the intrinsic and myeloma-induced secretion of interleukin-6 from bone marrow stromal cells. In addition, AS602868 inhibited myeloma cell growth in the MM.1S xenograft myeloma model. Of particular interest, AS602868 strongly increased myeloma sensitivity to TRAIL in blocking TRAIL-induced NF-kappaB activation and in decreasing the expression of antiapoptotic proteins such as cFLIP and cIAP-1/2. CONCLUSIONS: Taken together, our data point out the interest to inhibit the canonical NF-kappaB pathway in myeloma and clearly encourage clinical evaluation of novel therapies based on targeting NF-kappaB, especially in combination with TRAIL.


Subject(s)
Apoptosis , Gene Expression Regulation, Neoplastic , Multiple Myeloma/pathology , NF-kappa B/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Interleukin-6/metabolism , Mice , Models, Biological , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Neoplasm Transplantation , Pyrimidines/pharmacology , Receptors, Immunologic/metabolism , bcl-2-Associated X Protein/metabolism
5.
Proc Natl Acad Sci U S A ; 103(51): 19478-83, 2006 Dec 19.
Article in English | MEDLINE | ID: mdl-17164332

ABSTRACT

A critical role for vascular endothelial factor (VEGF) has been demonstrated in multiple myeloma (MM) pathogenesis. Here, we characterized the effect of the small-molecule VEGF receptor inhibitor pazopanib on MM cells in the bone marrow milieu. Pazopanib inhibits VEGF-triggered signaling pathways in both tumor and endothelial cells, thereby blocking in vitro MM cell growth, survival, and migration, and inhibits VEGF-induced up-regulation of adhesion molecules on both endothelial and tumor cells, thereby abrogating endothelial cell-MM cell binding and associated cell proliferation. We show that pazopanib is the first-in-class VEGF receptor inhibitor to inhibit in vivo tumor cell growth associated with increased MM cell apoptosis, decreased angiogenesis, and prolonged survival in a mouse xenograft model of human MM. Low-dose pazopanib demonstrates synergistic cytotoxicity with conventional (melphalan) and novel (bortezomib and immunomodulatory drugs) therapies. Finally, gene expression and signaling network analysis show transcriptional changes of several cancer-related genes, in particular c-Myc. Using siRNA, we confirm the role of c-Myc in VEGF production and secretion, as well as angiogenesis. These preclinical studies provide the rationale for clinical evaluation of pazopanib, alone and in combination with conventional and novel therapies, to increase efficacy, overcome drug resistance, reduce toxicity, and improve patient outcome in MM.


Subject(s)
Endothelial Cells/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Multiple Myeloma/metabolism , Pyrimidines/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/drug effects , Sulfonamides/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Immunoprecipitation , Indazoles , Mice , Microarray Analysis , Multiple Myeloma/drug therapy , Neovascularization, Pathologic/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL