Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Cell ; 28(1): 15-28, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-26175413

ABSTRACT

Although clinically tested JAK inhibitors reduce splenomegaly and systemic symptoms, molecular responses are not observed in most myeloproliferative neoplasm (MPN) patients. We previously demonstrated that MPN cells become persistent to type I JAK inhibitors that bind the active conformation of JAK2. We investigated whether CHZ868, a type II JAK inhibitor, would demonstrate activity in JAK inhibitor persistent cells, murine MPN models, and MPN patient samples. JAK2 and MPL mutant cell lines were sensitive to CHZ868, including type I JAK inhibitor persistent cells. CHZ868 showed significant activity in murine MPN models and induced reductions in mutant allele burden not observed with type I JAK inhibitors. These data demonstrate that type II JAK inhibition is a viable therapeutic approach for MPN patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Myeloproliferative Disorders/drug therapy , Protein Kinase Inhibitors/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Benzamides/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/administration & dosage , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism , Sequence Analysis, RNA , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
2.
Cancer Cell ; 28(1): 29-41, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-26175414

ABSTRACT

A variety of cancers depend on JAK2 signaling, including the high-risk subset of B cell acute lymphoblastic leukemias (B-ALLs) with CRLF2 rearrangements. Type I JAK2 inhibitors induce paradoxical JAK2 hyperphosphorylation in these leukemias and have limited activity. To improve the efficacy of JAK2 inhibition in B-ALL, we developed the type II inhibitor CHZ868, which stabilizes JAK2 in an inactive conformation. CHZ868 potently suppressed the growth of CRLF2-rearranged human B-ALL cells, abrogated JAK2 signaling, and improved survival in mice with human or murine B-ALL. CHZ868 and dexamethasone synergistically induced apoptosis in JAK2-dependent B-ALLs and further improved in vivo survival compared to CHZ868 alone. These data support the testing of type II JAK2 inhibition in patients with JAK2-dependent leukemias and other disorders.


Subject(s)
Aminopyridines/administration & dosage , Antineoplastic Agents/administration & dosage , Benzimidazoles/administration & dosage , Dexamethasone/administration & dosage , Drug Resistance, Neoplasm/drug effects , Janus Kinase 2/antagonists & inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/administration & dosage , Aminopyridines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis , Benzimidazoles/pharmacology , Cell Line, Tumor , Cytoprotection/drug effects , Drug Synergism , Humans , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , Mice , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
3.
Clin Cancer Res ; 19(22): 6230-41, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24081976

ABSTRACT

PURPOSE: The myeloproliferative neoplasm myelofibrosis is characterized by frequent deregulation of Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling, and JAK inhibitors were shown to reduce splenomegaly and ameliorate disease-related symptoms. However, the mutant clone and bone marrow fibrosis persist in the majority of patients. Using preclinical models, we explored whether JAK and pan-deacetylase inhibitor combination yielded additional benefits. EXPERIMENTAL DESIGN: The combination of the JAK1/2 inhibitor ruxolitinib and panobinostat was investigated using two different mouse models of JAK2(V617F)-driven disease. A Ba/F3 JAK2(V617F) cell-driven leukemic disease model was used to identify tolerated and efficacious doses. The drugs were then evaluated alone and in combination in a mouse model of myeloproliferative neoplasm-like disease based on transplantation of bone marrow transduced with a retrovirus expressing JAK2(V617F). Exposures were determined in blood and tissues, and phosphorylated STAT5 and acetylated histone H3 pharmacodynamic readouts were assessed in spleen and bone marrow. Histologic analysis was conducted on spleen and bone marrow, including staining of reticulin fibers in the latter organ. RESULTS: The combination of ruxolitinib and panobinostat was found to have a more profound effect on splenomegaly, as well as on bone marrow and spleen histology, compared with either agent alone, and the analysis of pharmacodynamic readouts showed that ruxolitinib and panobinostat have nonoverlapping and complementary effects. CONCLUSION: Combining JAK1/2 and pan-deacetylase inhibitors was fairly well tolerated and resulted in improved efficacy in mouse models of JAK2(V617F)-driven disease compared with the single agents. Thus, the combination of ruxolitinib and panobinostat may represent a promising novel therapeutic modality for myeloproliferative neoplasms.


Subject(s)
Hydroxamic Acids/therapeutic use , Indoles/therapeutic use , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Primary Myelofibrosis/drug therapy , Pyrazoles/therapeutic use , Acetylation , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Disease Models, Animal , Histone Deacetylase Inhibitors/adverse effects , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/drug effects , Histones/metabolism , Hydroxamic Acids/adverse effects , Indoles/adverse effects , Janus Kinase 1/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Nitriles , Panobinostat , Polycythemia Vera/drug therapy , Pyrazoles/adverse effects , Pyrimidines , Reticulin/analysis , STAT5 Transcription Factor/drug effects , STAT5 Transcription Factor/metabolism , Spleen/cytology , Spleen/metabolism , Splenomegaly/drug therapy , Thrombocytosis/drug therapy
4.
Blood ; 121(7): 1188-99, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23264594

ABSTRACT

To establish a preclinical animal model for testing drugs with potential effects on myeloproliferative neoplasms (MPNs), we first performed a detailed phenotypic characterization of Cre-inducible transgenic JAK2-V617F mice. Deleting the conditional mouse Jak2-knockout alleles increased erythropoiesis and accentuated the polycythemia vera phenotype, but did not alter platelet or granulocyte levels. In a transplantation assay, JAK2-V617F(+) BM cells had an advantage over wild-type competitor cells. Using this competitive repopulation assay, we compared the effects of INC424 (ruxolitinib), a dual Jak1/Jak2 inhibitor, and hydroxyurea (HU). HU led to weight loss, but did not reduce spleen weight. The hematologic parameters were lowered and a slight decrease of the mutant allele burden was noted. INC424 had little effect on body weight, but strongly decreased spleen size and rapidly normalized RBC and neutrophil parameters. No significant decrease in the mutant allele burden was observed. INC424 reduced the phospho-Stat5 levels, whereas HU strongly increased phospho-Stat5, most likely because of the elevated erythropoietin levels in response to the HU-induced anemia. This compensatory increase in JAK/STAT signaling may counteract the beneficial effects of cytoreduction at higher doses of HU and represents an adverse effect that should be avoided.


Subject(s)
Hydroxyurea/pharmacology , Janus Kinase 2/genetics , Polycythemia Vera/drug therapy , Polycythemia Vera/genetics , Pyrazoles/pharmacology , Alleles , Amino Acid Substitution , Animals , Bone Marrow Transplantation , Disease Models, Animal , Female , Hematopoiesis/drug effects , Hematopoiesis/genetics , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Nitriles , Phenotype , Polycythemia Vera/metabolism , Polycythemia Vera/pathology , Pyrimidines , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects
5.
Cancer Cell ; 22(6): 796-811, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23238015

ABSTRACT

Hyperactive PI3K/mTOR signaling is prevalent in human malignancies and its inhibition has potent antitumor consequences. Unfortunately, single-agent targeted cancer therapy is usually short-lived. We have discovered a JAK2/STAT5-evoked positive feedback loop that dampens the efficacy of PI3K/mTOR inhibition. Mechanistically, PI3K/mTOR inhibition increased IRS1-dependent activation of JAK2/STAT5 and secretion of IL-8 in several cell lines and primary breast tumors. Genetic or pharmacological inhibition of JAK2 abrogated this feedback loop and combined PI3K/mTOR and JAK2 inhibition synergistically reduced cancer cell number and tumor growth, decreased tumor seeding and metastasis, and also increased overall survival of the animals. Our results provide a rationale for combined targeting of the PI3K/mTOR and JAK2/STAT5 pathways in triple-negative breast cancer, a particularly aggressive and currently incurable disease.


Subject(s)
Breast Neoplasms/metabolism , Janus Kinase 2/antagonists & inhibitors , Neoplasm Metastasis/drug therapy , Phosphoinositide-3 Kinase Inhibitors , STAT5 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/drug therapy , Cell Death/drug effects , Cell Line, Tumor , Female , Humans , Insulin Receptor Substrate Proteins/metabolism , Interleukin-8/metabolism , Janus Kinase 2/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Interleukin-8A/metabolism , STAT5 Transcription Factor/metabolism , TOR Serine-Threonine Kinases/metabolism
6.
Cancer Discov ; 2(6): 512-523, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22684457

ABSTRACT

Janus kinase (JAK) inhibitors are being developed for the treatment of rheumatoid arthritis, psoriasis, myeloproliferative neoplasms, and leukemias. Most of these drugs target the ATP-binding pocket and stabilize the active conformation of the JAK kinases. This type I binding mode can lead to an increase in JAK activation loop phosphorylation, despite blockade of kinase function. Here we report that stabilizing the inactive state via type II inhibition acts in the opposite manner, leading to a loss of activation loop phosphorylation. We used X-ray crystallography to corroborate the binding mode and report for the first time the crystal structure of the JAK2 kinase domain in an inactive conformation. Importantly, JAK inhibitor-induced activation loop phosphorylation requires receptor interaction, as well as intact kinase and pseudokinase domains. Hence, depending on the respective conformation stabilized by a JAK inhibitor, hyperphosphorylation of the activation loop may or may not be elicited.


Subject(s)
Janus Kinases/antagonists & inhibitors , Janus Kinases/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Binding Sites , Cell Line, Tumor , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/chemistry , Mice , Phosphorylation/drug effects , Protein Binding , Protein Structure, Tertiary , STAT5 Transcription Factor/metabolism
7.
BMC Cancer ; 11: 24, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21247487

ABSTRACT

BACKGROUND: The JAK2V617F mutation plays a major role in the pathogenesis of myeloproliferative neoplasms and is found in the vast majority of patients suffering from polycythemia vera and in roughly every second patient suffering from essential thrombocythemia or from primary myelofibrosis. The V617F mutation is thought to provide hematopoietic stem cells and myeloid progenitors with a survival and proliferation advantage. It has previously been shown that activated JAK2 promotes cell survival by upregulating the anti-apoptotic STAT5 target gene Bcl-xL. In this study, we have investigated the role of additional apoptotic players, the pro-apoptotic protein Bim as well as the anti-apoptotic protein Mcl-1. METHODS: Pharmacological inhibition of JAK2/STAT5 signaling in JAK2V617F mutant SET-2 and MB-02 cells was used to study effects on signaling, cell proliferation and apoptosis by Western blot analysis, WST-1 proliferation assays and flow cytometry. Cells were transfected with siRNA oligos to deplete candidate pro- and anti-apoptotic proteins. Co-immunoprecipitation assays were performed to assess the impact of JAK2 inhibition on complexes of pro- and anti-apoptotic proteins. RESULTS: Treatment of JAK2V617F mutant cell lines with a JAK2 inhibitor was found to trigger Bim activation. Furthermore, Bim depletion by RNAi suppressed JAK2 inhibitor-induced cell death. Bim activation following JAK2 inhibition led to enhanced sequestration of Mcl-1, besides Bcl-xL. Importantly, Mcl-1 depletion by RNAi was sufficient to compromise JAK2V617F mutant cell viability and sensitized the cells to JAK2 inhibition. CONCLUSIONS: We conclude that Bim and Mcl-1 have key opposing roles in regulating JAK2V617F cell survival and propose that inactivation of aberrant JAK2 signaling leads to changes in Bim complexes that trigger cell death. Thus, further preclinical evaluation of combinations of JAK2 inhibitors with Bcl-2 family antagonists that also tackle Mcl-1, besides Bcl-xL, is warranted to assess the therapeutic potential for the treatment of chronic myeloproliferative neoplasms.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/physiology , Janus Kinase 2/metabolism , Membrane Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Proto-Oncogene Proteins/physiology , Amino Acid Substitution , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Blotting, Western , Caspase Inhibitors , Caspases/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/physiology , Flow Cytometry , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinoxalines/pharmacology , RNA Interference , Signal Transduction/drug effects
8.
Proc Natl Acad Sci U S A ; 104(52): 20926-31, 2007 Dec 26.
Article in English | MEDLINE | ID: mdl-18087038

ABSTRACT

Loss of imprinting (LOI) of the insulin-like growth factor-II gene (IGF2), leading to abnormal activation of the normally silent maternal allele, is a common human epigenetic population variant associated with a 5-fold increased frequency of colorectal neoplasia. Here, we show first that LOI leads specifically to increased expression of proliferation-related genes in mouse intestinal crypts. Surprisingly, LOI(+) mice also have enhanced sensitivity to IGF-II signaling, not simply increased IGF-II levels, because in vivo blockade with NVP-AEW541, a specific inhibitor of the IGF-II signaling receptor, showed reduction of proliferation-related gene expression to levels half that seen in LOI(-) mice. Signal transduction assays in microfluidic chips confirmed this enhanced sensitivity with marked augmentation of Akt/PKB signaling in LOI(+) cells at low doses of IGF-II, which was reduced in the presence of the inhibitor to levels below those found in LOI(-) cells, and was associated with increased expression of the IGF1 and insulin receptor genes. We exploited this increased IGF-II sensitivity to develop an in vivo chemopreventive strategy using the azoxymethane (AOM) mutagenesis model. LOI(+) mice treated with AOM showed a 60% increase in premalignant aberrant crypt foci (ACF) formation over LOI(-) mice. In vivo IGF-II blockade with NVP-AEW541 abrogated this effect, reducing ACF to a level 30% lower even than found in exposed LOI(-) mice. Thus, LOI increases cancer risk in a counterintuitive way, by increasing the sensitivity of the IGF-II signaling pathway itself, providing a previously undescribed epigenetic chemoprevention strategy in which cells with LOI are "IGF-II addicted" and undergo reduced tumorigenesis in the colon upon IGF-II pathway blockade.


Subject(s)
Genomic Imprinting , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Animals , Anticarcinogenic Agents/pharmacology , Azoxymethane/pharmacology , Cell Proliferation , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Pyrimidines/pharmacology , Pyrroles/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...