Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Blood ; 125(3): 516-24, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25395418

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is an aggressive myeloproliferative neoplasm of childhood associated with a poor prognosis. Recently, massively parallel sequencing has identified recurrent mutations in the SKI domain of SETBP1 in a variety of myeloid disorders. These lesions were detected in nearly 10% of patients with JMML and have been characterized as secondary events. We hypothesized that rare subclones with SETBP1 mutations are present at diagnosis in a large portion of patients who relapse, but are below the limits of detection for conventional deep sequencing platforms. Using droplet digital polymerase chain reaction, we identified SETBP1 mutations in 17/56 (30%) of patients who were treated in the Children's Oncology Group sponsored clinical trial, AAML0122. Five-year event-free survival in patients with SETBP1 mutations was 18% ± 9% compared with 51% ± 8% for those without mutations (P = .006).


Subject(s)
Carrier Proteins/genetics , Leukemia, Myelomonocytic, Juvenile/genetics , Mutation/genetics , Nuclear Proteins/genetics , Child, Preschool , Female , Follow-Up Studies , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Leukemia, Myelomonocytic, Juvenile/pathology , Male , Neoplasm Staging , Prognosis , Survival Rate
3.
Cancer Res ; 79(9): 2339-2351, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30862722

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the most common cancer in children. The highest rates of treatment failure occur in specific genetic subsets of ALL, including hypodiploid B-cell ALL (B-ALL), for which effective alternative therapies to current intensive chemotherapy treatments have yet to be developed. Here, we integrated biochemical and genomic profiling with functional drug assays to select effective agents with therapeutic potential against hypodiploid B-ALL. ABT-199, a selective Bcl-2 inhibitor, was effective in reducing leukemic burden in vitro and in vivo in patient-derived xenograft models of hypodiploid B-ALL. Daily oral treatment with ABT-199 significantly increased survival in xenografted mice. The unexpected efficacy of ABT-199 observed in hypodiploid leukemias lacking BIM expression (the major reported mediator of ABT-199-induced apoptosis) led us to investigate the mechanism of action of ABT-199 in the absence of BIM. Treatment with ABT-199 elicited responses in a dose-dependent manner, from cell-cycle arrest at low nanomolar concentrations to cell death at concentrations above 100 nmol/L. Collectively, these results demonstrate the efficacy of Bcl-2 inhibition and potential therapeutic strategy in hypodiploid B-ALL. SIGNIFICANCE: These results demonstrate the efficacy of ABT-199 in vivo and provide encouraging preclinical data of Bcl-2 as a potential target for the treatment of hypodiploid B-ALL.


Subject(s)
Antineoplastic Agents/pharmacology , Diploidy , Leukemia, Experimental/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Apoptosis , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Lineage , Cell Proliferation , Humans , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Bioconjug Chem ; 19(8): 1673-83, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18637680

ABSTRACT

CD22 represents a promising target for antibody-drug conjugate therapy in the context of B cell malignancies since it rapidly internalizes, importing specifically bound antibodies with it. To determine the pharmacokinetic parameters of anti-CD22-MCC-DM1 and MC-MMAF conjugates, various approaches to quantifying total and conjugated antibody were investigated. Although the total antibody assay formats gave similar results for both conjugates, the mouse pharmacokinetic profile for the anti-CD22-MCC-DM1 and MC-MMAF appeared significantly different depending on the conjugated antibody assay format. Since these differences significantly impacted the PK parameters determination, we investigated the effect of the drug/antibody ratio on the total and conjugated antibody quantification using multiple assay formats. Our investigations revealed the limitations of some assay formats to quantify anti-CD22-MCC-DM1 and MC-MMAF with different drug load and in the context of a heterogeneous ADC population highlight the need to carefully plan the assay strategy for the total and conjugated antibody quantification in order to accurately determine the ADC PK parameters.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Immunoconjugates/metabolism , Immunoconjugates/pharmacokinetics , Maleimides/metabolism , Maytansine/analogs & derivatives , Maytansine/metabolism , Oligopeptides/metabolism , Sialic Acid Binding Ig-like Lectin 2/immunology , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Humans , Immunoconjugates/analysis , Immunoconjugates/immunology , Mice , Sensitivity and Specificity
5.
Nat Commun ; 8(1): 2127, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29259179

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is a myeloproliferative disorder of childhood caused by mutations in the Ras pathway. Outcomes in JMML vary markedly from spontaneous resolution to rapid relapse after hematopoietic stem cell transplantation. Here, we hypothesized that DNA methylation patterns would help predict disease outcome and therefore performed genome-wide DNA methylation profiling in a cohort of 39 patients. Unsupervised hierarchical clustering identifies three clusters of patients. Importantly, these clusters differ significantly in terms of 4-year event-free survival, with the lowest methylation cluster having the highest rates of survival. These findings were validated in an independent cohort of 40 patients. Notably, all but one of 14 patients experiencing spontaneous resolution cluster together and closer to 22 healthy controls than to other JMML cases. Thus, we show that DNA methylation patterns in JMML are predictive of outcome and can identify the patients most likely to experience spontaneous resolution.


Subject(s)
DNA Methylation , Genome, Human/genetics , Leukemia, Myelomonocytic, Juvenile/genetics , Neoplasm Regression, Spontaneous/genetics , Antineoplastic Agents/therapeutic use , Biopsy , Child , Child, Preschool , Disease-Free Survival , Female , Hematopoietic Stem Cell Transplantation , Humans , Infant , Kaplan-Meier Estimate , Leukemia, Myelomonocytic, Juvenile/blood , Leukemia, Myelomonocytic, Juvenile/mortality , Leukemia, Myelomonocytic, Juvenile/therapy , Male , Monocytes , Mutation , Prognosis , Prospective Studies
6.
J Clin Invest ; 123(1): 335-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23221337

ABSTRACT

Children with neurofibromatosis type 1 (NF1) are predisposed to juvenile myelomonocytic leukemia (JMML), an aggressive myeloproliferative neoplasm (MPN) that is refractory to conventional chemotherapy. Conditional inactivation of the Nf1 tumor suppressor in hematopoietic cells of mice causes a progressive MPN that accurately models JMML and chronic myelomonocytic leukemia (CMML). We characterized the effects of Nf1 loss on immature hematopoietic populations and investigated treatment with the MEK inhibitor PD0325901 (hereafter called 901). Somatic Nf1 inactivation resulted in a marked expansion of immature and lineage-committed myelo-erythroid progenitors and ineffective erythropoiesis. Treatment with 901 induced a durable drop in leukocyte counts, enhanced erythropoietic function, and markedly reduced spleen sizes in mice with MPN. MEK inhibition also restored a normal pattern of erythroid differentiation and greatly reduced extramedullary hematopoiesis. Remarkably, genetic analysis revealed the persistence of Nf1-deficient hematopoietic cells, indicating that MEK inhibition modulates the proliferation and differentiation of Nf1 mutant cells in vivo rather than eliminating them. These data provide a rationale for performing clinical trials of MEK inhibitors in patients with JMML and CMML.


Subject(s)
Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Erythropoiesis/drug effects , Hematopoiesis, Extramedullary/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelomonocytic, Juvenile/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neurofibromin 1 , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Child , Child, Preschool , Diphenylamine/pharmacology , Disease Models, Animal , Erythropoiesis/genetics , Hematopoiesis, Extramedullary/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelomonocytic, Juvenile/etiology , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/metabolism , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurofibromatosis 1/complications , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/genetics
7.
Sci Signal ; 6(304): ra105, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24300897

ABSTRACT

Oncogenic K-Ras proteins, such as K-Ras(G12D), accumulate in the active, guanosine triphosphate (GTP)-bound conformation and stimulate signaling through effector kinases. The presence of the K-Ras(G12D) oncoprotein at a similar abundance to that of endogenous wild-type K-Ras results in only minimal phosphorylation and activation of the canonical Raf-mitogen-activated or extracellular signal-regulated protein kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) signaling cascades in primary hematopoietic cells, and these pathways remain dependent on growth factors for efficient activation. We showed that phospholipase C-γ (PLC-γ), PI3K, and their generated second messengers link activated cytokine receptors to Ras and ERK signaling in differentiated bone marrow cells and in a cell population enriched for leukemia stem cells. Cells expressing endogenous oncogenic K-Ras(G12D) remained dependent on the second messenger diacylglycerol for the efficient activation of Ras-ERK signaling. These data raise the unexpected possibility of therapeutically targeting proteins that function upstream of oncogenic Ras in cancer.


Subject(s)
Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hematopoietic Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Amino Acid Substitution , Animals , Cells, Cultured , Cytokines/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Hematopoietic Stem Cells/pathology , Leukemia/genetics , Leukemia/metabolism , Leukemia/pathology , MAP Kinase Signaling System/genetics , Mice , Mutation, Missense , Neoplastic Stem Cells/pathology , Phosphatidylinositol 3-Kinases/genetics , Phospholipase C gamma/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Second Messenger Systems/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
8.
Sci Transl Med ; 3(76): 76ra27, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21451123

ABSTRACT

Chronic and juvenile myelomonocytic leukemias (CMML and JMML) are aggressive myeloproliferative neoplasms that are incurable with conventional chemotherapy. Mutations that deregulate Ras signaling play a central pathogenic role in both disorders, and Mx1-Cre, Kras(LSL-G12D) mice that express the Kras oncogene develop a fatal disease that closely mimics these two leukemias in humans. Activated Ras controls multiple downstream effectors, but the specific pathways that mediate the leukemogenic effects of hyperactive Ras are unknown. We used PD0325901, a highly selective pharmacological inhibitor of mitogen-activated or extracellular signal-regulated protein kinase kinase (MEK), a downstream component of the Ras signaling network, to address how deregulated Raf/MEK/ERK (extracellular signal-regulated kinase) signaling drives neoplasia in Mx1-Cre, Kras(LSL-G12D) mice. PD0325901 treatment induced a rapid and sustained reduction in leukocyte counts, enhanced erythropoiesis, prolonged mouse survival, and corrected the aberrant proliferation and differentiation of bone marrow progenitor cells. These responses were due to direct effects of PD0325901 on Kras mutant cells rather than to stimulation of normal hematopoietic cell proliferation. Consistent with the in vivo response, inhibition of MEK reversed the cytokine hypersensitivity characteristic of Kras(G12D) hematopoietic progenitor cells in vitro. Our data demonstrate that deregulated Raf/MEK/ERK signaling is integral to the growth of Kras-mediated myeloproliferative neoplasms and further suggest that MEK inhibition could be a useful way to ameliorate functional hematologic abnormalities in patients with CMML and JMML.


Subject(s)
Benzamides/therapeutic use , Diphenylamine/analogs & derivatives , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mutation , Myeloproliferative Disorders/drug therapy , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Benzamides/pharmacology , Cell Differentiation/drug effects , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Myeloproliferative Disorders/pathology , Myeloproliferative Disorders/physiopathology , Myxovirus Resistance Proteins , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/physiopathology , Random Allocation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , raf Kinases/genetics , raf Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL