Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Mol Cancer Ther ; 23(7): 949-960, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38507740

ABSTRACT

The activated B cell (ABC) subset of diffuse large B-cell lymphoma (DLBCL) is characterized by chronic B-cell receptor signaling and associated with poor outcomes when treated with standard therapy. In ABC-DLBCL, MALT1 is a core enzyme that is constitutively activated by stimulation of the B-cell receptor or gain-of-function mutations in upstream components of the signaling pathway, making it an attractive therapeutic target. We discovered a novel small-molecule inhibitor, ABBV-MALT1, that potently shuts down B-cell signaling selectively in ABC-DLBCL preclinical models leading to potent cell growth and xenograft inhibition. We also identified a rational combination partner for ABBV-MALT1 in the BCL2 inhibitor, venetoclax, which when combined significantly synergizes to elicit deep and durable responses in preclinical models. This work highlights the potential of ABBV-MALT1 monotherapy and combination with venetoclax as effective treatment options for patients with ABC-DLBCL.


Subject(s)
Drug Synergism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Proto-Oncogene Proteins c-bcl-2 , Xenograft Model Antitumor Assays , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Humans , Animals , Mice , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Cell Line, Tumor , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Cell Proliferation/drug effects , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Disease Models, Animal
2.
Blood Adv ; 7(18): 5294-5303, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37315225

ABSTRACT

We evaluated immune cell subsets in patients with chronic lymphocytic leukemia (CLL) who received first-line therapy with 3 cycles of ibrutinib then 13 cycles of ibrutinib plus venetoclax in the minimal residual disease (MRD) cohort of the CAPTIVATE study (NCT02910583). Patients with Confirmed undetectable MRD (uMRD) were randomly assigned to placebo or ibrutinib groups; patients without Confirmed uMRD were randomly assigned to ibrutinib or ibrutinib plus venetoclax groups. We compared immune cell subsets in samples collected at 7 time points with age-matched healthy donors. CLL cells decreased within 3 cycles after venetoclax initiation; from cycle 16 onward, levels were similar to healthy donor levels (HDL; ≤0.8 cells per µL) in patients with Confirmed uMRD and slightly above HDL in patients without Confirmed uMRD. By 4 months after cycle 16, normal B cells had recovered to HDL in patients randomly assigned to placebo. Regardless of randomized treatment, abnormal counts of T cells, classical monocytes, and conventional dendritic cells recovered to HDL within 6 months (median change from baseline -49%, +101%, and +91%, respectively); plasmacytoid dendritic cells recovered by cycle 20 (+598%). Infections generally decreased over time regardless of randomized treatment and were numerically lowest in patients randomly assigned to placebo within 12 months after cycle 16. Sustained elimination of CLL cells and recovery of normal B cells were confirmed in samples from patients treated with fixed-duration ibrutinib plus venetoclax in the GLOW study (NCT03462719). These results demonstrate promising evidence of restoration of normal blood immune composition with ibrutinib plus venetoclax.


Subject(s)
Immune Reconstitution , Leukemia, Lymphocytic, Chronic, B-Cell , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Piperidines/therapeutic use
3.
Am J Hematol ; 98(3): 449-463, 2023 03.
Article in English | MEDLINE | ID: mdl-36594167

ABSTRACT

The treatment of patients with relapsed or refractory lymphoid neoplasms represents a significant clinical challenge. Here, we identify the pro-survival BCL-2 protein family member MCL-1 as a resistance factor for the BCL-2 inhibitor venetoclax in non-Hodgkin lymphoma (NHL) cell lines and primary NHL samples. Mechanistically, we show that the antibody-drug conjugate polatuzumab vedotin promotes MCL-1 degradation via the ubiquitin/proteasome system. This targeted MCL-1 antagonism, when combined with venetoclax and the anti-CD20 antibodies obinutuzumab or rituximab, results in tumor regressions in preclinical NHL models, which are sustained even off-treatment. In a Phase Ib clinical trial (NCT02611323) of heavily pre-treated patients with relapsed or refractory NHL, 25/33 (76%) patients with follicular lymphoma and 5/17 (29%) patients with diffuse large B-cell lymphoma achieved complete or partial responses with an acceptable safety profile when treated with the recommended Phase II dose of polatuzumab vedotin in combination with venetoclax and an anti-CD20 antibody.


Subject(s)
Immunoconjugates , Lymphoma, Non-Hodgkin , Humans , Myeloid Cell Leukemia Sequence 1 Protein/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/pathology , Rituximab/therapeutic use , Immunoconjugates/therapeutic use
4.
JCI Insight ; 7(17)2022 09 08.
Article in English | MEDLINE | ID: mdl-36073548

ABSTRACT

Heterozygous mutations in FLT3ITD, TET2, and DNMT3A are associated with hematologic malignancies in humans. In patients, cooccurrence of mutations in FLT3ITD combined with TET2 (TF) or FLT3ITD combined with DNMT3A (DF) are frequent. However, in some rare complex acute myeloid leukemia (AML), all 3 mutations cooccur - i.e., FLT3ITD, TET2, and DNMT3A (TFD). Whether the presence of these mutations in combination result in quantitative or qualitative differences in disease manifestation has not been investigated. We generated mice expressing heterozygous Flt3ITD and concomitant for either heterozygous loss of Tet2 (TF) or Dnmt3a (DF) or both (TFD). TF and DF mice did not induce disease early on, in spite of similar changes in gene expression; during the same time frame, an aggressive form of transplantable leukemia was observed in TFD mice, which was mostly associated with quantitative but not qualitative differences in gene expression relative to TF or DF mice. The gene expression signature of TFD mice showed remarkable similarity to the human TFD gene signature at the single-cell RNA level. Importantly, TFD-driven AML responded to a combination of drugs that target Flt3ITD, inflammation, and methylation in a mouse model, as well as in a PDX model of AML bearing 3 mutations.


Subject(s)
DNA Methyltransferase 3A , DNA-Binding Proteins , Dioxygenases , Leukemia, Myeloid, Acute , fms-Like Tyrosine Kinase 3 , Animals , DNA Methyltransferase 3A/genetics , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Proto-Oncogene Proteins/genetics , fms-Like Tyrosine Kinase 3/genetics
5.
Haematologica ; 106(4): 1034-1046, 2021 04 01.
Article in English | MEDLINE | ID: mdl-32414851

ABSTRACT

FLT3 internal tandem duplication (FLT3-ITD) mutations account for ~25% of adult acute myeloid leukemia cases and are associated with poor prognosis. Venetoclax, a selective BCL-2 inhibitor, has limited monotherapy activity in relapsed/refractory acute myeloid leukemia with no responses observed in a small subset of FLT3-ITD+ patients. Further, FLT3-ITD mutations emerged at relapse following venetoclax monotherapy and combination therapy suggesting a potential mechanism of resistance. Therefore, we investigated the convergence of FLT3-ITD signaling on the BCL-2 family proteins and determined combination activity of venetoclax and FLT3-ITD inhibition in preclinical models. In vivo, venetoclax combined with quizartinib, a potent FLT3 inhibitor, showed greater anti-tumor efficacy and prolonged survival compared to monotherapies. In a patient-derived FLT3-ITD+ xenograft model, cotreatment with venetoclax and quizartinib at clinically relevant doses had greater anti-tumor activity in the tumor microenvironment compared to quizartinib or venetoclax alone. Use of selective BCL-2 family inhibitors further identified a role for BCL-2, BCL-XL and MCL-1 in mediating survival in FLT3-ITD+ cells in vivo and highlighted the need to target all three proteins for greatest anti-tumor activity. Assessment of these combinations in vitro revealed synergistic combination activity for quizartinib and venetoclax but not for quizartinib combined with BCL-XL or MCL-1 inhibition. FLT3-ITD inhibition was shown to indirectly target both BCL-XL and MCL-1 through modulation of protein expression, thereby priming cells toward BCL-2 dependence for survival. These data demonstrate that FLT3-ITD inhibition combined with venetoclax has impressive anti-tumor activity in FLT3-ITD+ acute myeloid leukemia preclinical models and provides strong mechanistic rational for clinical studies.


Subject(s)
Leukemia, Myeloid, Acute , Adult , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Protein Kinase Inhibitors , Sulfonamides/pharmacology , Tumor Microenvironment , fms-Like Tyrosine Kinase 3/genetics
6.
Stem Cell Reports ; 15(1): 95-109, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32502465

ABSTRACT

Studies of patients with acute myeloid leukemia (AML) have led to the identification of mutations that affect different cellular pathways. Some of these have been classified as preleukemic, and a stepwise evolution program whereby cells acquire additional mutations has been proposed in the development of AML. How the timing of acquisition of these mutations and their impact on transformation and the bone marrow (BM) microenvironment occurs has only recently begun to be investigated. We show that constitutive and early loss of the epigenetic regulator, TET2, when combined with constitutive activation of FLT3, results in transformation of chronic myelomonocytic leukemia-like or myeloproliferative neoplasm-like phenotype to AML, which is more pronounced in double-mutant mice relative to mice carrying mutations in single genes. Furthermore, we show that in preleukemic and leukemic mice there are alterations in the BM niche and secreted cytokines, which creates a permissive environment for the growth of mutation-bearing cells relative to normal cells.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation/genetics , Animals , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Proliferation , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Dioxygenases , Heterozygote , Homozygote , Humans , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins/metabolism , Severity of Illness Index , Tumor Microenvironment , fms-Like Tyrosine Kinase 3/metabolism
7.
Cell Stem Cell ; 23(6): 833-849.e5, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30526882

ABSTRACT

Inflammation is a risk factor for cancer development. Individuals with preleukemic TET2 mutations manifest clonal hematopoiesis and are at a higher risk of developing leukemia. How inflammatory signals influence the survival of preleukemic hematopoietic stem and progenitor cells (HSPCs) is unclear. We show a rapid increase in the frequency and absolute number of Tet2-KO mature myeloid cells and HSPCs in response to inflammatory stress, which results in enhanced production of inflammatory cytokines, including interleukin-6 (IL-6), and resistance to apoptosis. IL-6 induces hyperactivation of the Shp2-Stat3 signaling axis, resulting in increased expression of a novel anti-apoptotic long non-coding RNA (lncRNAs), Morrbid, in Tet2-KO myeloid cells and HSPCs. Expression of activated Shp2 in HSPCs phenocopies Tet2 loss with regard to hyperactivation of Stat3 and Morrbid. In vivo, pharmacologic inhibition of Shp2 or Stat3 or genetic loss of Morrbid in Tet2 mutant mice rescues inflammatory-stress-induced abnormalities in HSPCs and mature myeloid cells, including clonal hematopoiesis.


Subject(s)
Benzoquinones/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Hematopoietic Stem Cells/drug effects , Inflammation/drug therapy , Myeloid Cells/drug effects , Piperidines/pharmacology , Propionates/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrimidines/pharmacology , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Female , Hematopoiesis/drug effects , Hematopoietic Stem Cells/metabolism , Inflammation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Myeloid Cells/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
8.
JCI Insight ; 3(4)2018 02 22.
Article in English | MEDLINE | ID: mdl-29467326

ABSTRACT

Mutations in KIT and TET2 are associated with myeloid malignancies. We show that loss of TET2-induced PI3K activation and -increased proliferation is rescued by targeting the p110α/δ subunits of PI3K. RNA-Seq revealed a hyperactive c-Myc signature in Tet2-/- cells, which is normalized by inhibiting PI3K signaling. Loss of TET2 impairs the maturation of myeloid lineage-derived mast cells by dysregulating the expression of Mitf and Cebpa, which is restored by low-dose ascorbic acid and 5-azacytidine. Utilizing a mouse model in which the loss of TET2 precedes the expression of oncogenic Kit, similar to the human disease, results in the development of a non-mast cell lineage neoplasm (AHNMD), which is responsive to PI3K inhibition. Thus, therapeutic approaches involving hypomethylating agents, ascorbic acid, and isoform-specific PI3K inhibitors are likely to be useful for treating patients with TET2 and KIT mutations.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/genetics , Mast Cells/pathology , Mastocytosis/genetics , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , DNA Methylation/drug effects , DNA-Binding Proteins/genetics , Dioxygenases , Disease Models, Animal , Gain of Function Mutation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Knock-In Techniques , Humans , Mastocytosis/drug therapy , Mastocytosis/pathology , Mice , Mice, Knockout , Myeloid Cells/drug effects , Myeloid Cells/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-kit/genetics
9.
J Clin Invest ; 126(7): 2621-5, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27294524

ABSTRACT

Hyperactivation of the mTOR pathway impairs hematopoietic stem cell (HSC) functions and promotes leukemogenesis. mTORC1 and mTORC2 differentially control normal and leukemic stem cell functions. mTORC1 regulates p70 ribosomal protein S6 kinase 1 (S6K1) and eukaryotic initiation factor 4E-binding (eIF4E-binding) protein 1 (4E-BP1), and mTORC2 modulates AKT activation. Given the extensive crosstalk that occurs between mTORC1 and mTORC2 signaling pathways, we assessed the role of the mTORC1 substrate S6K1 in the regulation of both normal HSC functions and in leukemogenesis driven by the mixed lineage leukemia (MLL) fusion oncogene MLL-AF9. We demonstrated that S6K1 deficiency impairs self-renewal of murine HSCs by reducing p21 expression. Loss of S6K1 also improved survival in mice transplanted with MLL-AF9-positive leukemic stem cells by modulating AKT and 4E-BP1 phosphorylation. Taken together, these results suggest that S6K1 acts through multiple targets of the mTOR pathway to promote self-renewal and leukemia progression. Given the recent interest in S6K1 as a potential therapeutic target in cancer, our results further support targeting this molecule as a potential strategy for treatment of myeloid malignancies.


Subject(s)
Carrier Proteins/metabolism , Hematopoietic Stem Cells/cytology , Leukemia/blood , Multiprotein Complexes/metabolism , Phosphoproteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Disease Progression , Eukaryotic Initiation Factors , Humans , Leukemia/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , Stem Cells/metabolism
10.
Oncotarget ; 7(13): 16936-47, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26943578

ABSTRACT

Understanding mast cell development is essential due to their critical role in regulating immunity and autoimmune diseases. Here, we show how Rho kinases (ROCK) regulate mast cell development and can function as therapeutic targets for treating allergic diseases. Rock1 deficiency results in delayed maturation of bone marrow derived mast cells (BMMCs) in response to IL-3 stimulation and reduced growth in response to stem cell factor (SCF) stimulation. Further, integrin-mediated adhesion and migration, and IgE-mediated degranulation are all impaired in Rock1-deficient BMMCs. To understand the mechanism behind altered mast cell development in Rock1-/- BMMCs, we analyzed the activation of ROCK and its downstream targets including LIM kinase (LIMK). We observed reduced activation of ROCK, LIMK, AKT and ERK1/2 in Rock1-deficient BMMCs in response to SCF stimulation. Further, loss of either Limk1 or Limk2 also demonstrated altered BMMC maturation and growth; combined deletion of both Limk1 and Limk2 resulted in further reduction in BMMC maturation and growth. In passive cutaneous anaphylaxis model, deficiency of Rock1 or treatment with ROCK inhibitor Fasudil protected mice against IgE-mediated challenge. Our results identify ROCK/LIMK pathway as a novel therapeutic target for treating allergic diseases involving mast cells.


Subject(s)
Cell Differentiation/immunology , Lim Kinases/metabolism , Mast Cells/cytology , Mast Cells/metabolism , rho-Associated Kinases/metabolism , Actins/metabolism , Animals , Cell Adhesion/immunology , Cell Degranulation/immunology , Cell Movement/immunology , Hypersensitivity/immunology , Mice , Mice, Knockout
11.
Hum Mol Genet ; 25(6): 1129-39, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26740548

ABSTRACT

Persons with neurofibromatosis type 1 (NF1) have a predisposition for premature and severe arterial stenosis. Mutations in the NF1 gene result in decreased expression of neurofibromin, a negative regulator of p21(Ras), and increases Ras signaling. Heterozygous Nf1 (Nf1(+/-)) mice develop a marked arterial stenosis characterized by proliferating smooth muscle cells (SMCs) and a predominance of infiltrating macrophages, which closely resembles arterial lesions from NF1 patients. Interestingly, lineage-restricted inactivation of a single Nf1 allele in monocytes/macrophages is sufficient to recapitulate the phenotype observed in Nf1(+/-) mice and to mobilize proinflammatory CCR2+ monocytes into the peripheral blood. Therefore, we hypothesized that CCR2 receptor activation by its primary ligand monocyte chemotactic protein-1 (MCP-1) is critical for monocyte infiltration into the arterial wall and neointima formation in Nf1(+/-) mice. MCP-1 induces a dose-responsive increase in Nf1(+/-) macrophage migration and proliferation that corresponds with activation of multiple Ras kinases. In addition, Nf1(+/-) SMCs, which express CCR2, demonstrate an enhanced proliferative response to MCP-1 when compared with WT SMCs. To interrogate the role of CCR2 activation on Nf1(+/-) neointima formation, we induced neointima formation by carotid artery ligation in Nf1(+/-) and WT mice with genetic deletion of either MCP1 or CCR2. Loss of MCP-1 or CCR2 expression effectively inhibited Nf1(+/-) neointima formation and reduced macrophage content in the arterial wall. Finally, administration of a CCR2 antagonist significantly reduced Nf1(+/-) neointima formation. These studies identify MCP-1 as a potent chemokine for Nf1(+/-) monocytes/macrophages and CCR2 as a viable therapeutic target for NF1 arterial stenosis.


Subject(s)
Macrophages/pathology , Monocytes/pathology , Neointima/pathology , Neurofibromatosis 1/pathology , Receptors, CCR2/metabolism , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Genes, Neurofibromatosis 1 , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/genetics , Neointima/metabolism , Neurofibromatosis 1/genetics , Neurofibromatosis 1/metabolism , Neurofibromin 1/genetics , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/genetics , Signal Transduction
12.
Cell Rep ; 9(4): 1333-48, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25456130

ABSTRACT

Oncogenic mutations of FLT3 and KIT receptors are associated with poor survival in patients with acute myeloid leukemia (AML) and myeloproliferative neoplasms (MPNs), and currently available drugs are largely ineffective. Although Stat5 has been implicated in regulating several myeloid and lymphoid malignancies, how precisely Stat5 regulates leukemogenesis, including its nuclear translocation to induce gene transcription, is poorly understood. In leukemic cells, we show constitutive activation of focal adhesion kinase (FAK) whose inhibition represses leukemogenesis. Downstream of FAK, activation of Rac1 is regulated by RacGEF Tiam1, whose inhibition prolongs the survival of leukemic mice. Inhibition of the Rac1 effector PAK1 prolongs the survival of leukemic mice in part by inhibiting the nuclear translocation of Stat5. These results reveal a leukemic pathway involving FAK/Tiam1/Rac1/PAK1 and demonstrate an essential role for these signaling molecules in regulating the nuclear translocation of Stat5 in leukemogenesis.


Subject(s)
Carcinogenesis/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Leukemia, Myeloid, Acute/metabolism , Proto-Oncogene Proteins c-kit/metabolism , STAT5 Transcription Factor/metabolism , fms-Like Tyrosine Kinase 3/metabolism , p21-Activated Kinases/metabolism , Animals , Benzothiazoles/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , Mastocytosis, Systemic/pathology , Mice, Inbred C57BL , Mutant Proteins/metabolism , Mutation/genetics , Phenylurea Compounds/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Signal Transduction/drug effects , Survival Analysis , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , p21-Activated Kinases/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism
13.
J Med Chem ; 57(15): 6594-609, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25003231

ABSTRACT

The Src homology 2 domain containing protein tyrosine phosphatase-2 (SHP2) is an oncogenic phosphatase associated with various kinds of leukemia and solid tumors. Thus, there is substantial interest in developing SHP2 inhibitors as potential anticancer and antileukemia agents. Using a structure-guided and fragment-based library approach, we identified a novel hydroxyindole carboxylic acid-based SHP2 inhibitor 11a-1, with an IC50 value of 200 nM and greater than 5-fold selectivity against 20 mammalian PTPs. Structural and modeling studies reveal that the hydroxyindole carboxylic acid anchors the inhibitor to the SHP2 active site, while interactions of the oxalamide linker and the phenylthiophene tail with residues in the ß5-ß6 loop contribute to 11a-1's binding potency and selectivity. Evidence suggests that 11a-1 specifically attenuates the SHP2-dependent signaling inside the cell. Moreover, 11a-1 blocks growth factor mediated Erk1/2 and Akt activation and exhibits excellent antiproliferative activity in lung cancer and breast cancer as well as leukemia cell lines.


Subject(s)
Antineoplastic Agents/chemistry , Indoles/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Enzyme Activation , Humans , Indoles/chemical synthesis , Indoles/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Docking Simulation , Molecular Targeted Therapy , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Small Molecule Libraries , Structure-Activity Relationship
14.
Curr Opin Hematol ; 21(4): 271-5, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24867289

ABSTRACT

PURPOSE OF REVIEW: Rho kinases (ROCKs) are involved in regulating a variety of physiologic functions including cytoskeletal reorganization, migration, adhesion, survival and proliferation. They do so via activating several different downstream substrates such as myosin light chain phosphatase, LIM kinase and ezrin/radixin/moesin proteins. To date, most of the conclusions with regard to the function of ROCKs have involved the use of cell line models, pharmacologic inhibitors and dominant negative approaches. Importantly, the role of ROCK in hematopoiesis or leukemogenesis in the context of whole organism remains poorly understood. RECENT FINDINGS: Recent studies utilizing mice deficient in the expression of ROCK1 have begun to shed some light into the physiologic role(s) of ROCK in both normal and abnormal hematopoiesis. Findings, thus far, suggest that ROCK plays an essential role in regulating growth and survival in different hematopoietic lineages via distinct mechanisms, in part, by utilizing distinct downstream substrates including maintaining the activation of tumor-suppressor genes. SUMMARY: In blood cells, emerging data suggest that ROCK plays an essential role in negatively regulating inflammatory and erythropoietic stress and positively regulates the growth and survival of leukemic cells.


Subject(s)
Hematologic Diseases/genetics , Hematologic Diseases/metabolism , Hematopoiesis/physiology , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Erythropoiesis , Humans , Leukemia/genetics , Leukemia/metabolism , Myeloid Cells/physiology , Stress, Physiological
15.
Blood ; 123(18): 2838-42, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24553178

ABSTRACT

Although hyperactivation of the Ras-Erk signaling pathway is known to underlie the pathogenesis of juvenile myelomonocytic leukemia (JMML), a fatal childhood disease, the PI3K-Akt signaling pathway is also dysregulated in this disease. Using genetic models, we demonstrate that inactivation of phosphatidylinositol-3-kinase (PI3K) catalytic subunit p110δ, but not PI3K p110α, corrects gain-of-function (GOF) Shp2-induced granulocyte macrophage-colony-stimulating factor (GM-CSF) hypersensitivity, Akt and Erk hyperactivation, and skewed hematopoietic progenitor distribution. Likewise, potent p110δ-specific inhibitors curtail the proliferation of GOF Shp2-expressing hematopoietic cells and cooperate with mitogen-activated or extracellular signal-regulated protein kinase kinase (MEK) inhibition to reduce proliferation further and maximally block Erk and Akt activation. Furthermore, the PI3K p110δ-specific inhibitor, idelalisib, also demonstrates activity against primary leukemia cells from individuals with JMML. These findings suggest that selective inhibition of the PI3K catalytic subunit p110δ could provide an innovative approach for treatment of JMML, with the potential for limiting toxicity resulting from the hematopoietic-restricted expression of p110δ.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Leukemia, Myelomonocytic, Juvenile/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Proliferation/drug effects , Class Ia Phosphatidylinositol 3-Kinase/genetics , Disease Models, Animal , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Leukemia, Myelomonocytic, Juvenile/genetics , Mice , Mice, Knockout , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
16.
Am J Pathol ; 184(1): 79-85, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24211110

ABSTRACT

Neurofibromatosis type 1 (NF1) results from mutations in the NF1 tumor-suppressor gene, which encodes neurofibromin, a negative regulator of diverse Ras signaling cascades. Arterial stenosis is a nonneoplastic manifestation of NF1 that predisposes some patients to debilitating morbidity and sudden death. Recent murine studies demonstrate that Nf1 heterozygosity (Nf1(+/-)) in monocytes/macrophages significantly enhances intimal proliferation after arterial injury. However, the downstream Ras effector pathway responsible for this phenotype is unknown. Based on in vitro assays demonstrating enhanced extracellular signal-related kinase (Erk) signaling in Nf1(+/-) macrophages and vascular smooth muscle cells and in vivo evidence of Erk amplification without alteration of phosphatidylinositol 3-kinase signaling in Nf1(+/-) neointimas, we tested the hypothesis that Ras-Erk signaling regulates intimal proliferation in a murine model of NF1 arterial stenosis. By using a well-established in vivo model of inflammatory cell migration and standard cell culture, neurofibromin-deficient macrophages demonstrate enhanced sensitivity to growth factor stimulation in vivo and in vitro, which is significantly diminished in the presence of PD0325901, a specific inhibitor of Ras-Erk signaling in phase 2 clinical trials for cancer. After carotid artery injury, Nf1(+/-) mice demonstrated increased intimal proliferation compared with wild-type mice. Daily administration of PD0325901 significantly reduced Nf1(+/-) neointima formation to levels of wild-type mice. These studies identify the Ras-Erk pathway in neurofibromin-deficient macrophages as the aberrant pathway responsible for enhanced neointima formation.


Subject(s)
Carotid Stenosis/pathology , Macrophages/metabolism , Neointima/pathology , Neurofibromatosis 1/metabolism , Neurofibromin 1/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Carotid Stenosis/metabolism , Disease Models, Animal , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neointima/metabolism , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Neurofibromin 1/genetics , ras Proteins/physiology
17.
J Clin Invest ; 123(10): 4449-63, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24091327

ABSTRACT

An acquired somatic mutation at codon 816 in the KIT receptor tyrosine kinase is associated with poor prognosis in patients with systemic mastocytosis and acute myeloid leukemia (AML). Treatment of leukemic cells bearing this mutation with an allosteric inhibitor of p21-activated kinase (Pak) or its genetic inactivation results in growth repression due to enhanced apoptosis. Inhibition of the upstream effector Rac abrogates the oncogene-induced growth and activity of Pak. Although both Rac1 and Rac2 are constitutively activated via the guanine nucleotide exchange factor (GEF) Vav1, loss of Rac1 or Rac2 alone moderately corrected the growth of KIT-bearing leukemic cells, whereas the combined loss resulted in 75% growth repression. In vivo, the inhibition of Vav or Rac or Pak delayed the onset of myeloproliferative neoplasms (MPNs) and corrected the associated pathology in mice. To assess the role of Rac GEFs in oncogene-induced transformation, we used an inhibitor of Rac, EHop-016, which specifically targets Vav1 and found that EHop-016 was a potent inhibitor of human and murine leukemic cell growth. These studies identify Pak and Rac GTPases, including Vav1, as potential therapeutic targets in MPN and AML involving an oncogenic form of KIT.


Subject(s)
Aminoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/pharmacology , p21-Activated Kinases/physiology , rac GTP-Binding Proteins/physiology , Animals , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Enzyme Activation , Humans , Mastocytosis/drug therapy , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mutation, Missense , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-vav/metabolism , Xenograft Model Antitumor Assays , p21-Activated Kinases/antagonists & inhibitors , rac GTP-Binding Proteins/antagonists & inhibitors
19.
Hum Mol Genet ; 22(5): 977-88, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23197650

ABSTRACT

Mutations in the NF1 tumor suppressor gene cause Neurofibromatosis type 1 (NF1). Neurofibromin, the protein product of NF1, functions as a negative regulator of Ras activity. Some NF1 patients develop cardiovascular disease, which represents an underrecognized disease complication and contributes to excess morbidity and mortality. Specifically, NF1 patients develop arterial occlusion resulting in tissue ischemia and sudden death. Murine studies demonstrate that heterozygous inactivation of Nf1 (Nf1(+/-)) in bone marrow cells enhances neointima formation following arterial injury. Macrophages infiltrate Nf1(+/-) neointimas, and NF1 patients have increased circulating inflammatory monocytes in their peripheral blood. Therefore, we tested the hypothesis that heterozygous inactivation of Nf1 in myeloid cells is sufficient for neointima formation. Specific ablation of a single copy of the Nf1 gene in myeloid cells alone mobilizes a discrete pro-inflammatory murine monocyte population via a cell autonomous and gene-dosage dependent mechanism. Furthermore, lineage-restricted heterozygous inactivation of Nf1 in myeloid cells is sufficient to reproduce the enhanced neointima formation observed in Nf1(+/-) mice when compared with wild-type controls, and homozygous inactivation of Nf1 in myeloid cells amplified the degree of arterial stenosis after arterial injury. Treatment of Nf1(+/-) mice with rosuvastatin, a stain with anti-inflammatory properties, significantly reduced neointima formation when compared with control. These studies identify neurofibromin-deficient myeloid cells as critical cellular effectors of Nf1(+/-) neointima formation and propose a potential therapeutic for NF1 cardiovascular disease.


Subject(s)
Arterial Occlusive Diseases/genetics , Fluorobenzenes/administration & dosage , Neointima/drug therapy , Neointima/genetics , Neurofibromatosis 1/genetics , Neurofibromin 1/genetics , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Arterial Occlusive Diseases/complications , Arterial Occlusive Diseases/physiopathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Heterozygote , Humans , Macrophages/cytology , Macrophages/pathology , Metabolic Networks and Pathways/drug effects , Mice , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Neointima/physiopathology , Neurofibromatosis 1/complications , Neurofibromatosis 1/physiopathology , Neurofibromin 1/metabolism , Rosuvastatin Calcium
SELECTION OF CITATIONS
SEARCH DETAIL
...