Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Hematol ; 98: 47-52.e6, 2021 06.
Article in English | MEDLINE | ID: mdl-33794295

ABSTRACT

Azacitidine and enasidenib are two therapies available for treatment of acute myelogenous leukemia (AML), and the mechanisms of action of these drugs involve alteration of aberrant DNA methylation. We hypothesized that a combination of these agents could have interactive effects on DNA methylation and enhance differentiation in mIDH2 cells. Combination treatment enhanced cellular differentiation in TF-1 cells overexpressing IDJ2R140Q through increased hemoglobinization and increased hemoglobin γ RNA expression compared with the effects of single agents. Furthermore, in primary AML samples (IDH2R140Q or R172K), combination treatment reduced CD34+ cells and increased CD15+ cells to a greater extent than attained with single agents. To explore the mechanism of enhanced differentiation with combination treatment, the TF-1 epigenome was analyzed by profiling 5-hydroxymethylcytosine (5hmC) and 5-methylcytosine (5mC) DNA methylation changes. Enasidenib treatment alone increased 5hmC, consistent with reactivation of ten-eleven-translocation (TET) enzyme activity. Compared with treatment with azacitidine alone, combination treatment reduced 5mC levels at greater numbers of sites and these loci were significantly enriched in regions with increased 5hMC (25.8% vs. 7.4%). Results are consistent with a model in which enasidenib-mediated reactivation of ten-eleven-translocation enzymes cooperates with azacitidine-mediated inhibition of DNA methyltransferase enzymes, leading to greater reductions in DNA methylation and enhanced erythroid differentiation.


Subject(s)
Aminopyridines/pharmacology , Azacitidine/pharmacology , DNA Methylation/drug effects , DNA, Neoplasm/metabolism , Leukemia, Myeloid, Acute/metabolism , Triazines/pharmacology , Cell Line, Tumor , Humans , Leukemia, Myeloid, Acute/drug therapy
2.
Clin Cancer Res ; 25(1): 90-98, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30201761

ABSTRACT

PURPOSE: Avadomide is a novel, small-molecule therapeutic agent that modulates cereblon E3 ligase activity and exhibits potent antitumor and immunomodulatory activities. This first-in-human phase I study (NCT01421524) evaluated the safety and clinical activity of avadomide in patients with advanced solid tumors, non-Hodgkin lymphoma (NHL), and multiple myeloma. PATIENTS AND METHODS: Thirty-four patients were treated with avadomide in 7 dose-escalation cohorts using a 3 + 3 design (0.5-3.5 mg, 28-day continuous dosing cycles). The primary objectives were to determine the dose-limiting toxicity (DLT), nontolerated dose (NTD), maximum tolerated dose (MTD), recommended phase II dose, and pharmacokinetics of avadomide. The secondary objective was to determine preliminary avadomide efficacy. Exploratory objectives included evaluation of pharmacodynamic effects of avadomide. RESULTS: DLTs were reported in 2 patients, and grade ≥3 treatment-emergent adverse events (TEAEs) occurred in 14 patients (41%). The most common TEAEs (≥15%) were fatigue, neutropenia, and diarrhea. The NTD and MTD were 3.5 and 3.0 mg, respectively. Of 5 patients with NHL, 1 achieved a complete response, and 2 had partial responses. Although no objective responses were observed in patients with solid tumors, 5 of 6 patients with brain cancer experienced nonprogression of ≥6 months. A dose-dependent relationship between Aiolos degradation in peripheral B and T cells occurred within 5 hours of the first dose of avadomide administered, starting at 0.5 mg. CONCLUSIONS: Avadomide monotherapy demonstrated acceptable safety and favorable pharmacokinetics in patients with solid tumors, NHL, and multiple myeloma. In addition, 3 objective responses were observed in NHL.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Lymphoma, Non-Hodgkin/drug therapy , Multiple Myeloma/drug therapy , Piperidones/administration & dosage , Quinazolinones/administration & dosage , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug-Related Side Effects and Adverse Reactions/classification , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/pathology , Male , Maximum Tolerated Dose , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Piperidones/adverse effects , Quinazolinones/adverse effects , Ubiquitin-Protein Ligases
3.
Proc Natl Acad Sci U S A ; 116(2): 695-700, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30584089

ABSTRACT

Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are diseases of abnormal hematopoietic differentiation with aberrant epigenetic alterations. Azacitidine (AZA) is a DNA methyltransferase inhibitor widely used to treat MDS and AML, yet the impact of AZA on the cell-surface proteome has not been defined. To identify potential therapeutic targets for use in combination with AZA in AML patients, we investigated the effects of AZA treatment on four AML cell lines representing different stages of differentiation. The effect of AZA treatment on these cell lines was characterized at three levels: the DNA methylome, the transcriptome, and the cell-surface proteome. Untreated AML cell lines showed substantial overlap at all three omics levels; however, while AZA treatment globally reduced DNA methylation in all cell lines, changes in the transcriptome and surface proteome were subtle and differed among the cell lines. Transcriptome analysis identified five commonly up-regulated coding genes upon AZA treatment in all four cell lines, TRPM4 being the only gene encoding a surface protein, and surface proteome analysis found no commonly regulated proteins. Gene set enrichment analysis of differentially regulated RNA and surface proteins showed a decrease in metabolic pathways and an increase in immune defense response pathways. As such, AZA treatment led to diverse effects at the individual gene and protein levels but converged to common responses at the pathway level. Given the heterogeneous responses in the four cell lines, we discuss potential therapeutic strategies for AML in combination with AZA.


Subject(s)
Azacitidine/pharmacology , DNA, Neoplasm , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute , Membrane Proteins , Neoplasm Proteins , Proteome , Up-Regulation/drug effects , DNA Methylation/drug effects , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Genomics , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Proteome/biosynthesis , Proteome/genetics
4.
Blood ; 131(24): 2661-2669, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29724899

ABSTRACT

Pinometostat (EPZ-5676) is a first-in-class small-molecule inhibitor of the histone methyltransferase disrupter of telomeric silencing 1-like (DOT1L). In this phase 1 study, pinometostat was evaluated for safety and efficacy in adult patients with advanced acute leukemias, particularly those involving mixed lineage leukemia (MLL) gene rearrangements (MLL-r) resulting from 11q23 translocations. Fifty-one patients were enrolled into 6 dose-escalation cohorts (n = 26) and 2 expansion cohorts (n = 25) at pinometostat doses of 54 and 90 mg/m2 per day by continuous intravenous infusion in 28-day cycles. Because a maximum tolerated dose was not established in the dose-escalation phase, the expansion doses were selected based on safety and clinical response data combined with pharmacodynamic evidence of reduction in H3K79 methylation during dose escalation. Across all dose levels, plasma pinometostat concentrations increased in an approximately dose-proportional fashion, reaching an apparent steady-state by 4-8 hours after infusion, and rapidly decreased following treatment cessation. The most common adverse events, of any cause, were fatigue (39%), nausea (39%), constipation (35%), and febrile neutropenia (35%). Overall, 2 patients, both with t(11;19), experienced complete remission at 54 mg/m2 per day by continuous intravenous infusion, demonstrating proof of concept for delivering clinically meaningful responses through targeting DOT1L using the single agent pinometostat in MLL-r leukemia patients. Administration of pinometostat was generally safe, with the maximum tolerated dose not being reached, although efficacy as a single agent was modest. This study demonstrates the therapeutic potential for targeting DOT1L in MLL-r leukemia and lays the groundwork for future combination approaches in this patient population. This clinical trial is registered at www.clinicaltrials.gov as NCT01684150.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Histones/metabolism , Leukemia, Myeloid, Acute/drug therapy , Methyltransferases/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Benzimidazoles/adverse effects , Female , Histone-Lysine N-Methyltransferase , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Methylation/drug effects , Methyltransferases/metabolism , Middle Aged , Young Adult
5.
Clin Cancer Res ; 24(17): 4072-4080, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29764853

ABSTRACT

Purpose: This large two-part, three-arm phase I study examined the safety and tolerability of CC-486 (an oral formulation of azacitidine, a hypomethylating agent) alone or in combination with the cytotoxic agents, carboplatin or nab-paclitaxel, in patients with advanced unresectable solid tumors.Patients and Methods: Part 1 (n = 57) was a dose escalation of CC-486 alone (arm C) or with carboplatin (arm A) or nab-paclitaxel (arm B). The primary endpoint was safety, MTD, and recommended part 2 dose (RP2D) of CC-486. In part 2 (n = 112), the primary endpoint was the safety and tolerability of CC-486 administered at the RP2D for each treatment arm, in tumor-specific expansion cohorts. Secondary endpoints included pharmacokinetics, pharmacodynamics, and antitumor activity of CC-486.Results: At pharmacologically active doses CC-486 in combination with carboplatin or nab-paclitaxel had a tolerable safety profile and no drug-drug interactions. The CC-486 RP2D was determined as 300 mg (every day, days 1-14/21) in combination with carboplatin (arm A) or as monotherapy (arm C); and 200 mg in the same dosing regimen in combination with nab-paclitaxel (arm B). Albeit limited by the small sample size, CC-486 monotherapy resulted in partial responses (three/eight) and stable disease (four/eight) in patients with nasopharyngeal cancer. Three of the stable disease responses lasted more than 150 days.Conclusions: CC-486 is well tolerated alone or in combination with carboplatin or nab-paclitaxel. Exploratory analyses suggest clinical activity of CC-486 monotherapy in nasopharyngeal cancer and provided the basis for an ongoing phase II clinical trial (ClinicalTrials.gov identifier: NCT02269943). Clin Cancer Res; 24(17); 4072-80. ©2018 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Azacitidine/administration & dosage , Neoplasms/drug therapy , Aged , Albumins/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Azacitidine/adverse effects , Carboplatin/administration & dosage , Disease-Free Survival , Drug Administration Schedule , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/classification , Neoplasms/genetics , Neoplasms/pathology , Paclitaxel/administration & dosage , Treatment Outcome
6.
Cancer Cell ; 13(6): 483-95, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18538732

ABSTRACT

Faithful modeling of mixed-lineage leukemia in murine cells has been difficult to achieve. We show that expression of MLL-AF9 in human CD34+ cells induces acute myeloid, lymphoid, or mixed-lineage leukemia in immunodeficient mice. Some leukemia stem cells (LSC) were multipotent and could be lineage directed by altering either the growth factors or the recipient strain of mouse, highlighting the importance of microenvironmental cues. Other LSC were strictly lineage committed, demonstrating the heterogeneity of the stem cell compartment in MLL disease. Targeting the Rac signaling pathway by pharmacologic or genetic means resulted in rapid and specific apoptosis of MLL-AF9 cells, suggesting that the Rac signaling pathway may be a valid therapeutic target in MLL-rearranged AML.


Subject(s)
Cell Lineage , Cell Transformation, Neoplastic/pathology , Fetal Stem Cells/pathology , Leukemia, Myeloid, Acute/pathology , Multipotent Stem Cells/pathology , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplastic Stem Cells/pathology , Oncogene Proteins, Fusion/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Aneuploidy , Animals , Antigens, CD34/analysis , Apoptosis , Cell Culture Techniques , Cell Line, Transformed , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromosomes, Human, Pair 11 , Environment , Fetal Blood/immunology , Fetal Blood/metabolism , Fetal Stem Cells/immunology , Fetal Stem Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genotype , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Multipotent Stem Cells/immunology , Multipotent Stem Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Phenotype , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Signal Transduction , Species Specificity , Stem Cell Transplantation , Time Factors , Transduction, Genetic , Translocation, Genetic , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
8.
Mol Cell Biol ; 24(12): 5324-31, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15169896

ABSTRACT

Pbx2 is one of four mammalian genes that encode closely related TALE homeodomain proteins, which serve as DNA binding partners for a subset of Hox transcription factors. The expression and contributions of Pbx2 to mammalian development remain undefined, in contrast to the essential roles recently established for family members Pbx1 and Pbx3. Here we report that Pbx2 is widely expressed during embryonic development, particularly in neural and epithelial tissues during late gestation. Despite wide Pbx2 expression, mice homozygous mutant for Pbx2 are born at the expected Mendelian frequencies and exhibit no detectable abnormalities in development and organogenesis or reduction of long-term survival. The lack of an apparent phenotype in Pbx2(-)/(-) mice likely reflects functional redundancy, since the Pbx2 protein is present at considerably lower levels than comparable isoforms of Pbx1 and/or Pbx3 in embryonic tissues. In postnatal bone marrow and thymus, however, Pbx2 is the predominant high-molecular-weight (MW)-isoform Pbx protein detectable by immunoblotting. Nevertheless, the absence of Pbx2 has no measurable effect on steady-state hematopoiesis or immune function in adult mice, suggesting possible compensation by low-MW-isoform Pbx proteins present in these tissues. We conclude that the roles of Pbx2 in murine embryonic development, organogenesis, hematopoiesis, immune responses, and long-term survival are not essential.


Subject(s)
Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Animals , Embryonic and Fetal Development/genetics , Embryonic and Fetal Development/physiology , Female , Gene Expression Regulation, Developmental , Gene Targeting , Genes, Homeobox , Hematopoiesis/genetics , Hematopoiesis/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pre-B-Cell Leukemia Transcription Factor 1 , Pregnancy , Proto-Oncogene Proteins/deficiency , Transcription Factors/genetics , Transcription Factors/physiology
9.
Blood ; 99(10): 3780-5, 2002 May 15.
Article in English | MEDLINE | ID: mdl-11986236

ABSTRACT

The t(10;11)(p12;q23) chromosomal translocation in human acute myeloid leukemia results in the fusion of the MLL and AF10 genes. The latter codes for a novel leucine zipper protein, one of many MLL fusion partners of unknown function. In this report, we demonstrate that retroviral-mediated transduction of an MLL-AF10 complementary DNA into primary murine myeloid progenitors enhanced their clonogenic potential in serial replating assays and led to their efficient immortalization at a primitive stage of myeloid differentiation. Furthermore, MLL-AF10-transduced cells rapidly induced acute myeloid leukemia in syngeneic or severe combined immunodeficiency recipient mice. Structure/function analysis showed that a highly conserved 82-amino acid portion of AF10, comprising 2 adjacent alpha-helical domains, was sufficient for immortalizing activity when fused to MLL. Neither helical domain alone mediated immortalization, and deletion of the 29-amino acid leucine zipper within this region completely abrogated transforming activity. Similarly, the minimal oncogenic domain of AF10 exhibited transcriptional activation properties when fused to the MLL or GAL4 DNA-binding domains, while neither helical domain alone did. However, transcriptional activation per se was not sufficient because a second activation domain of AF10 was neither required nor competent for transformation. The requirement for alpha-helical transcriptional effector domains is similar to the oncogenic contributions of unrelated MLL partners ENL and ELL, suggesting a general mechanism of myeloid leukemogenesis by a subset of MLL fusion proteins, possibly through specific recruitment of the transcriptional machinery.


Subject(s)
Cell Transformation, Neoplastic , Leukemia, Myeloid/etiology , Myeloid Progenitor Cells/metabolism , Oncogene Proteins, Fusion/physiology , Proto-Oncogenes , Transcription Factors/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , COS Cells , Cell Line , Cells, Cultured , Conserved Sequence , DNA-Binding Proteins/genetics , Histone-Lysine N-Methyltransferase , Leucine Zippers , Leukemia, Myeloid/pathology , Mice , Mice, Inbred C57BL , Mice, SCID , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Protein Structure, Tertiary , Sequence Alignment , Transcription Factors/chemistry , Transcriptional Activation , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...