Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Blood ; 128(10): 1382-95, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27343252

ABSTRACT

The clinical success of the BCL-2-selective BH3-mimetic venetoclax in patients with poor prognosis chronic lymphocytic leukemia (CLL) highlights the potential of targeting the BCL-2-regulated apoptotic pathway in previously untreatable lymphoid malignancies. By selectively inhibiting BCL-2, venetoclax circumvents the dose-limiting, BCL-XL-mediated thrombocytopenia of its less selective predecessor navitoclax, while enhancing efficacy in CLL. We have previously reported the potent sensitivity of many high-risk childhood acute lymphoblastic leukemia (ALL) xenografts to navitoclax. Given the superior tolerability of venetoclax, here we have investigated its efficacy in childhood ALL. We demonstrate that in contrast to the clear dependence of CLL on BCL-2 alone, effective antileukemic activity in the majority of ALL xenografts requires concurrent inhibition of both BCL-2 and BCL-XL We identify BCL-XL expression as a key predictor of poor response to venetoclax and demonstrate that concurrent inhibition of both BCL-2 and BCL-XL results in synergistic killing in the majority of ALL xenografts. A notable exception is mixed lineage leukemia-rearranged infant ALL, where venetoclax largely recapitulates the activity of navitoclax, identifying this subgroup of patients as potential candidates for clinical trials of venetoclax in childhood ALL. Conversely, our findings provide a clear basis for progressing navitoclax into trials ahead of venetoclax in other subgroups.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Drug Resistance, Neoplasm/drug effects , Gene Rearrangement/genetics , Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Sulfonamides/therapeutic use , Animals , Apoptosis/genetics , Blotting, Western , Cell Proliferation/drug effects , Child , Drug Resistance, Neoplasm/genetics , Female , Flow Cytometry , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Nat Commun ; 6: 5794, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25565005

ABSTRACT

Early T-cell precursor leukaemia (ETP-ALL) is a high-risk subtype of human leukaemia that is poorly understood at the molecular level. Here we report translocations targeting the zinc finger E-box-binding transcription factor ZEB2 as a recurrent genetic lesion in immature/ETP-ALL. Using a conditional gain-of-function mouse model, we demonstrate that sustained Zeb2 expression initiates T-cell leukaemia. Moreover, Zeb2-driven mouse leukaemia exhibit some features of the human immature/ETP-ALL gene expression signature, as well as an enhanced leukaemia-initiation potential and activated Janus kinase (JAK)/signal transducers and activators of transcription (STAT) signalling through transcriptional activation of IL7R. This study reveals ZEB2 as an oncogene in the biology of immature/ETP-ALL and paves the way towards pre-clinical studies of novel compounds for the treatment of this aggressive subtype of human T-ALL using our Zeb2-driven mouse model.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Homeodomain Proteins/genetics , Leukemia, T-Cell/physiopathology , Repressor Proteins/genetics , Signal Transduction/physiology , Animals , Blotting, Western , Chromatin Immunoprecipitation , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Histological Techniques , Homeodomain Proteins/immunology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Janus Kinases/metabolism , Kaplan-Meier Estimate , Karyotyping , Luciferases , Mice , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-7/metabolism , Repressor Proteins/immunology , STAT Transcription Factors/metabolism , Signal Transduction/genetics , Zinc Finger E-box Binding Homeobox 2
3.
Mol Cancer Ther ; 14(2): 364-74, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25504635

ABSTRACT

Genome-wide studies have identified a high-risk subgroup of pediatric acute lymphoblastic leukemia (ALL) harboring mutations in the Janus kinases (JAK). The purpose of this study was to assess the preclinical efficacy of the JAK1/2 inhibitor AZD1480, both as a single agent and in combination with the MEK inhibitor selumetinib, against JAK-mutated patient-derived xenografts. Patient-derived xenografts were established in immunodeficient mice from bone marrow or peripheral blood biopsy specimens, and their gene expression profiles compared with the original patient biopsies by microarray analysis. JAK/STAT and MAPK signaling pathways, and the inhibitory effects of targeted drugs, were interrogated by immunoblotting of phosphoproteins. The antileukemic effects of AZD1480 and selumetinib, alone and in combination, were tested against JAK-mutated ALL xenografts both in vitro and in vivo. Xenografts accurately represented the primary disease as determined by gene expression profiling. Cellular phosphoprotein analysis demonstrated that JAK-mutated xenografts exhibited heightened activation status of JAK/STAT and MAPK signaling pathways compared with typical B-cell precursor ALL xenografts, which were inhibited by AZD1480 exposure. However, AZD1480 exhibited modest single-agent in vivo efficacy against JAK-mutated xenografts. Combining AZD1480 with selumetinib resulted in profound synergistic in vitro cell killing, although these results were not translated in vivo despite evidence of target inhibition. Despite validation of target inhibition and the demonstration of profound in vitro synergy between AZD1480 and selumetinib, it is likely that prolonged target inhibition is required to achieve in vivo therapeutic enhancement between JAK and MEK inhibitors in the treatment of JAK-mutated ALL.


Subject(s)
Janus Kinases/antagonists & inhibitors , Mutation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Animals , Benzimidazoles/pharmacology , Biopsy , Cell Survival/drug effects , Child , Gene Expression Regulation, Leukemic/drug effects , Genetic Association Studies , Humans , Janus Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , Molecular Targeted Therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Treatment Outcome , Xenograft Model Antitumor Assays
4.
Clin Cancer Res ; 20(17): 4520-31, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25013123

ABSTRACT

PURPOSE: Predictive biomarkers are required to identify patients who may benefit from the use of BH3 mimetics such as ABT-263. This study investigated the efficacy of ABT-263 against a panel of patient-derived pediatric acute lymphoblastic leukemia (ALL) xenografts and utilized cell and molecular approaches to identify biomarkers that predict in vivo ABT-263 sensitivity. EXPERIMENTAL DESIGN: The in vivo efficacy of ABT-263 was tested against a panel of 31 patient-derived ALL xenografts composed of MLL-, BCP-, and T-ALL subtypes. Basal gene expression profiles of ALL xenografts were analyzed and confirmed by quantitative RT-PCR, protein expression and BH3 profiling. An in vitro coculture assay with immortalized human mesenchymal cells was utilized to build a predictive model of in vivo ABT-263 sensitivity. RESULTS: ABT-263 demonstrated impressive activity against pediatric ALL xenografts, with 19 of 31 achieving objective responses. Among BCL2 family members, in vivo ABT-263 sensitivity correlated best with low MCL1 mRNA expression levels. BH3 profiling revealed that resistance to ABT-263 correlated with mitochondrial priming by NOXA peptide, suggesting a functional role for MCL1 protein. Using an in vitro coculture assay, a predictive model of in vivo ABT-263 sensitivity was built. Testing this model against 11 xenografts predicted in vivo ABT-263 responses with high sensitivity (50%) and specificity (100%). CONCLUSION: These results highlight the in vivo efficacy of ABT-263 against a broad range of pediatric ALL subtypes and shows that a combination of in vitro functional assays can be used to predict its in vivo efficacy.


Subject(s)
Aniline Compounds/administration & dosage , Neoplasm Proteins/biosynthesis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Sulfonamides/administration & dosage , Apoptosis/drug effects , Child , Gene Expression Regulation, Neoplastic/drug effects , Humans , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/biosynthesis , Xenograft Model Antitumor Assays
5.
J Allergy Clin Immunol ; 130(3): 735-742.e6, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22621957

ABSTRACT

BACKGROUND: Mutations in the SP110 gene result in infantile onset of the autosomal recessive primary immunodeficiency disease veno-occlusive disease with immunodeficiency syndrome (VODI), which is characterized by hypogammaglobulinemia, T-cell dysfunction, and a high frequency of hepatic veno-occlusive disease. OBJECTIVES: We sought to further characterize the clinical features, B-lineage cellular immunologic findings, and molecular pathogenesis of this disorder in 9 patients with new diagnoses, including 4 novel mutations from families of Italian, Hispanic, and Arabic ethnic origin. METHODS: Methods used include clinical review; Sanger DNA sequencing of the SP110 gene; determination of transfected mutant protein function by using immunofluorescent studies in Hep-2 cells; quantitation of B-cell subsets by means of flow cytometry; assessments of B-cell function after stimulation with CD40 ligand, IL-21, or both; and differential gene expression array studies of EBV-transformed B cells. RESULTS: We confirm the major diagnostic criteria and the clinical utility of SP110 mutation testing for the diagnosis of VODI. Analysis of 4 new alleles confirms that VODI is caused by reduced functional SP110 protein levels. Detailed B-cell immunophenotyping demonstrated that Sp110 deficiency compromises the ability of human B cells to respond to T cell-dependent stimuli and differentiate into immunoglobulin-secreting cells in vitro. Expression microarray studies have identified pathways involved in B-lymphocyte differentiation and macrophage function. CONCLUSION: These studies show that a range of mutations in SP110 that cause decreased SP110 protein levels and impaired late B-cell differentiation cause VODI and that the condition is not restricted to the Lebanese population.


Subject(s)
Hepatic Veno-Occlusive Disease/genetics , Immunologic Deficiency Syndromes/genetics , Nuclear Proteins/genetics , Adult , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Child , Child, Preschool , Hepatic Veno-Occlusive Disease/drug therapy , Hepatic Veno-Occlusive Disease/immunology , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunologic Deficiency Syndromes/drug therapy , Immunologic Deficiency Syndromes/immunology , Immunophenotyping , Infant , Minor Histocompatibility Antigens , Mutation , Nuclear Proteins/analysis
6.
Expert Rev Clin Immunol ; 6(5): 765-75, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20828285

ABSTRACT

B-cell development is characterized by the progressive maturation of hematopoietic stem cells through several stages to ultimately give rise to the mature B-cell pool that has been selected for reactivity against non-self antigens. Thus, the mature pool of naive B cells is capable of elicting high-affinity responses following natural infection with pathogens or vaccination and provides the host with protective long-lived humoral immunity. However, perturbations during the processes of B-cell development and differentiation can give rise to a diverse array of immunological diseases including autoimmunity, immunodeficiency and malignancy. While we have a very rich understanding of the processes underlying B-cell development in mice, our knowledge of the corresponding events occurring in human B cells is substantially less robust. Here, we overview the latest findings relating to human B cells in health and disease with a particular emphasis on the transitional stage of B-cell development.


Subject(s)
B-Lymphocyte Subsets/metabolism , B-Lymphocytes/metabolism , Immunologic Deficiency Syndromes/immunology , Animals , Autoimmunity , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Biomedical Research/trends , Cell Differentiation/genetics , Cell Differentiation/immunology , Humans , Immunologic Deficiency Syndromes/genetics , Mice , Mutation/genetics
7.
J Exp Med ; 207(1): 155-71, 2010 Jan 18.
Article in English | MEDLINE | ID: mdl-20048285

ABSTRACT

Engagement of cytokine receptors by specific ligands activate Janus kinase-signal transducer and activator of transcription (STAT) signaling pathways. The exact roles of STATs in human lymphocyte behavior remain incompletely defined. Interleukin (IL)-21 activates STAT1 and STAT3 and has emerged as a potent regulator of B cell differentiation. We have studied patients with inactivating mutations in STAT1 or STAT3 to dissect their contribution to B cell function in vivo and in response to IL-21 in vitro. STAT3 mutations dramatically reduced the number of functional, antigen (Ag)-specific memory B cells and abolished the ability of IL-21 to induce naive B cells to differentiate into plasma cells (PCs). This resulted from impaired activation of the molecular machinery required for PC generation. In contrast, STAT1 deficiency had no effect on memory B cell formation in vivo or IL-21-induced immunoglobulin secretion in vitro. Thus, STAT3 plays a critical role in generating effector B cells from naive precursors in humans. STAT3-activating cytokines such as IL-21 thus underpin Ag-specific humoral immune responses and provide a mechanism for the functional antibody deficit in STAT3-deficient patients.


Subject(s)
Cell Differentiation/physiology , Immunologic Memory/physiology , Interleukins/immunology , Plasma Cells/immunology , STAT3 Transcription Factor/immunology , Signal Transduction/physiology , Antibody Formation/physiology , Antigens/genetics , Antigens/immunology , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Interleukins/genetics , Plasma Cells/cytology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT3 Transcription Factor/genetics , Time Factors
8.
Blood ; 115(3): 519-29, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-19965666

ABSTRACT

The transitional stage of B-cell development represents an important step where autoreactive cells are deleted, allowing the generation of a mature functional B-cell repertoire. In mice, 3 subsets of transitional B cells have been identified. In contrast, most studies of human transitional B cells have focused on a single subset defined as CD24(hi)CD38(hi) B cells. Here, we have identified 2 subsets of human transitional B cells based on the differential expression of CD21. CD21(hi) transitional cells displayed higher expression of CD23, CD44, and IgD, and exhibited greater proliferation and Ig secretion in vitro than CD21(lo) transitional B cells. In contrast, the CD21(lo) subset expressed elevated levels of LEF1, a transcription factor highly expressed by immature lymphocytes, and produced higher amounts of autoreactive Ab. These phenotypic, functional, and molecular features suggest that CD21(lo) transitional B cells are less mature than the CD21(hi) subset. This was confirmed by analyzing X-linked agammaglobulinemia patients and the kinetics of B-cell reconstitution after stem cell transplantation, which revealed that the development of CD21(lo) transitional B cells preceded that of CD21(hi) transitional cells. These findings provide important insights into the process of human B-cell development and have implications for understanding the processes underlying perturbed B-cell maturation in autoimmune and immunodeficient conditions.


Subject(s)
Precursor Cells, B-Lymphoid/physiology , Receptors, Complement 3d/genetics , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation/physiology , Humans , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , Receptors, Complement 3d/metabolism
9.
Immunol Cell Biol ; 87(8): 590-600, 2009.
Article in English | MEDLINE | ID: mdl-19721453

ABSTRACT

T follicular helper (T(FH)) cells are a specialized subset of CD4(+) T cells that localize to B-cell follicles, where they are positioned to provide help for the induction of optimal humoral immune responses. Key features of T(FH) cells are the expressions of CXCR5, ICOS, interleukin (IL)-21 and BCL-6. The requirements for human T(FH) cell development are unknown. Here we show that IL-6, IL-12, IL-21 and IL-23 are capable of inducing IL-21 expression in naïve CD4(+) T cells isolated from human tonsils, peripheral blood and cord blood. However, only IL-12 induced sustained expressions of CXCR5 and ICOS on these activated naïve CD4(+) T cells, and endowed them with the ability to provide increased help to B cells for their differentiation into immunoglobulin-secreting cells. The effects of IL-12 were independent of interferon-gamma and T-bet, and associated with upregulation of BCL-6 expression. Thus, these cytokines, particularly IL-12, are likely to act at an early stage during dendritic cell-mediated priming of naïve CD4(+) T cells into a T(FH) cell fate, and thus underpin antibody-mediated immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Lineage , Immunity, Innate , Interleukin-12/immunology , Lymphoid Tissue/immunology , T-Lymphocytes, Helper-Inducer/immunology , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Humans , Interleukins/immunology , Phenotype , T-Lymphocytes, Helper-Inducer/cytology
10.
J Neuroimmunol ; 183(1-2): 96-103, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17240458

ABSTRACT

ThIL-17 (IL-17+/IFN-gamma-) cell lines are significantly more encephalitogenic than Th1 (IL-17-/IFN-gamma+) cell lines in adoptive transfer EAE models. In actively induced EAE short ex vivo peptide stimulation identifies an IL-17+/IFN-gamma+ population of CD4+ CNS-infiltrating MOG35-55-specific T cells, which outnumber IL-17+/IFN-gamma- cells by approximately 3:1 as disease develops. A decrease in numbers of IL-17+/IFN-gamma+ cells following in vitro culture is accompanied by an increase in IL-17-/IFN-gamma+ cell numbers. Together these ex vivo and in vitro observations imply that the Th1 lineage is more encephalitogenic than is suggested by adoptive transfer of Th1 (IL-17-/IFN-gamma+) cell lines which have been terminally differentiated in vitro.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Interferon-gamma/pharmacology , Interleukin-17/metabolism , Th1 Cells/drug effects , Animals , Cell Proliferation/drug effects , Central Nervous System/pathology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Flow Cytometry/methods , Glycoproteins , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , Th1 Cells/metabolism , Thymidine/metabolism , Tritium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...