Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Blood ; 115(15): 3109-17, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20130243

ABSTRACT

Constitutive JAK2 activation in hematopoietic cells by the JAK2V617F mutation recapitulates myeloproliferative neoplasm (MPN) phenotypes in mice, establishing JAK2 inhibition as a potential therapeutic strategy. Although most polycythemia vera patients carry the JAK2V617F mutation, half of those with essential thrombocythemia or primary myelofibrosis do not, suggesting alternative mechanisms for constitutive JAK-STAT signaling in MPNs. Most patients with primary myelofibrosis have elevated levels of JAK-dependent proinflammatory cytokines (eg, interleukin-6) consistent with our observation of JAK1 hyperactivation. Accordingly, we evaluated the effectiveness of selective JAK1/2 inhibition in experimental models relevant to MPNs and report on the effects of INCB018424, the first potent, selective, oral JAK1/JAK2 inhibitor to enter the clinic. INCB018424 inhibited interleukin-6 signaling (50% inhibitory concentration [IC(50)] = 281nM), and proliferation of JAK2V617F(+) Ba/F3 cells (IC(50) = 127nM). In primary cultures, INCB018424 preferentially suppressed erythroid progenitor colony formation from JAK2V617F(+) polycythemia vera patients (IC(50) = 67nM) versus healthy donors (IC(50) > 400nM). In a mouse model of JAK2V617F(+) MPN, oral INCB018424 markedly reduced splenomegaly and circulating levels of inflammatory cytokines, and preferentially eliminated neoplastic cells, resulting in significantly prolonged survival without myelosuppressive or immunosuppressive effects. Preliminary clinical results support these preclinical data and establish INCB018424 as a promising oral agent for the treatment of MPNs.


Subject(s)
Janus Kinases/antagonists & inhibitors , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/enzymology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/therapeutic use , Amino Acid Substitution/genetics , Animals , Apoptosis/drug effects , Blood Cell Count , Cell Proliferation/drug effects , Cell Survival/drug effects , Colony-Forming Units Assay , Cytokines/blood , Disease Models, Animal , Drug Screening Assays, Antitumor , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/genetics , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Mice , Myeloproliferative Disorders/blood , Myeloproliferative Disorders/pathology , Nitriles , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/pharmacology , Pyrimidines , Signal Transduction/drug effects , Spleen/drug effects , Spleen/pathology , Treatment Outcome
2.
Mol Cancer Ther ; 8(1): 26-35, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19139110

ABSTRACT

Protein tyrosine kinases of the Janus kinase (JAK) family are associated with many cytokine receptors, which, on ligand binding, regulate important cellular functions such as proliferation, survival, and differentiation. In multiple myeloma, JAKs may be persistently activated due to a constant stimulation by interleukin (IL)-6, which is produced in the bone marrow environment. INCB20 is a synthetic molecule that potently inhibits all members of the JAK family with a 100- to 1,000-fold selectivity for JAKs over >70 other kinases. Treatment of multiple myeloma cell lines and patient tumor cells with INCB20 resulted in a significant and dose-dependent inhibition of spontaneous as well as IL-6-induced cell growth. Importantly, multiple myeloma cell growth was inhibited in the presence of bone marrow stromal cells. The IL-6 dependent cell line INA-6 was particularly sensitive to the drug (IC50<1 micromol/L). Growth suppression of INA-6 correlated with an increase in the percentage of apoptotic cells and inhibition of signal transducer and activator of transcription 3 phosphorylation. INCB20 also abrogated the protective effect of IL-6 against dexamethasone by blocking phosphorylation of SHP-2 and AKT. In contrast, AKT phosphorylation induced by insulin-like growth factor-I remained unchanged, showing selectivity of the compound. In a s.c. severe combined immunodeficient mouse model with INA-6, INCB20 significantly delayed INA-6 tumor growth. Our studies show that disruption of JAKs and downstream signaling pathways may both inhibit multiple myeloma cell growth and survival and overcome cytokine-mediated drug resistance, thereby providing the preclinical rationale for the use of JAK inhibitors as a novel therapeutic approach in multiple myeloma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Janus Kinases/antagonists & inhibitors , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Bone Marrow Cells/drug effects , Bone Marrow Cells/enzymology , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Cytoprotection/drug effects , Dexamethasone/pharmacology , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Janus Kinases/genetics , Janus Kinases/metabolism , Mice , Multiple Myeloma/drug therapy , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , STAT3 Transcription Factor/metabolism , Stromal Cells/drug effects , Stromal Cells/enzymology , Substrate Specificity , Survival Rate , Xenograft Model Antitumor Assays
3.
Clin Cancer Res ; 13(6): 1892-902, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17363546

ABSTRACT

PURPOSE: ErbB receptor signaling pathways are important regulators of cell fate, and their dysregulation, through (epi)genetic alterations, plays an etiologic role in multiple cancers. ErbB ligands are synthesized as membrane-bound precursors that are cleaved by members of the ADAM family of zinc-dependent metalloproteases. This processing, termed ectodomain shedding, is essential for the functional activation of ErbB ligands. Recent studies suggest that elevated levels of ErbB ligands may circumvent the effectiveness of ErbB-targeted therapeutics. Here, we describe the discovery and preclinical development of potent, selective inhibitors of ErbB ligand shedding. EXPERIMENTAL DESIGN: A series of biochemical and cell-based assays were established to identify selective inhibitors of ErbB ligand shedding. The therapeutic potential of these compounds was assessed in multiple in vivo models of cancer and matrix metalloprotease-related toxicity. RESULTS: INCB3619 was identified as a representative selective, potent, orally bioavailable small-molecule inhibitor of a subset of ADAM proteases that block shedding of ErbB ligands. Administration of INCB3619 to tumor-bearing mice reduced ErbB ligand shedding in vivo and inhibited ErbB pathway signaling (e.g., phosphorylation of Akt), tumor cell proliferation, and survival. Further, INCB3619 synergized with clinically relevant cancer therapeutics and showed no overt or compounding toxicities, including fibroplasia, the dose-limiting toxicity associated with broad-spectrum matrix metalloprotease inhibitors. CONCLUSIONS: Inhibition of ErbB ligand shedding offers a potentially novel and well-tolerated therapeutic strategy for the treatment of human cancers and is currently being evaluated in the clinic.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Oncogene Proteins v-erbB/metabolism , Signal Transduction/drug effects , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Neoplasms/metabolism , Neoplasms/pathology , Rats , Xenograft Model Antitumor Assays
4.
J Med Chem ; 50(4): 603-6, 2007 Feb 22.
Article in English | MEDLINE | ID: mdl-17256836

ABSTRACT

The design, synthesis, evaluation, and identification of a novel class of (6S,7S)-N-hydroxy-6-carboxamide-5-azaspiro[2.5]octane-7-carboxamides as the first potent and selective inhibitors of human epidermal growth factor receptor-2 (HER-2) sheddase is described. Several compounds were identified that possess excellent pharmacodynamic and pharmacokinetic properties and were shown to decrease tumor size, cleaved HER-2 extracellular domain plasma levels, and potentiate the effects of the humanized anti-HER-2 monoclonal antibody (trastuzumab) in vivo in a HER-2 overexpressing cancer murine xenograft model.


Subject(s)
Amides/chemical synthesis , Antineoplastic Agents/chemical synthesis , Hydroxamic Acids/chemical synthesis , Piperidines/chemical synthesis , Receptor, ErbB-2/antagonists & inhibitors , Spiro Compounds/chemical synthesis , Administration, Oral , Amides/pharmacokinetics , Amides/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/pharmacology , Mice , Molecular Conformation , Piperidines/chemistry , Piperidines/pharmacology , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous , Trastuzumab
5.
Cancer Biol Ther ; 5(6): 648-56, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16627988

ABSTRACT

The HER-2 receptor tyrosine kinase is an important regulator of cell proliferation and survival, and it is a clinically validated target of therapeutic intervention for HER-2 positive breast cancer patients. Its extracellular domain (ECD) is frequently cleaved by protease(s) in HER-2 overexpressing breast cancer patients, rendering the remaining membrane-bound portion (p95) a constitutively activated kinase. The presence of both serum ECD and cellular p95 protein has been linked to poor clinical outcome as well as reduced effectiveness of some therapeutic treatments. We have identified a series of potent, selective small molecule inhibitors of ADAM proteases, exemplified here by INCB003619, and demonstrate that these inhibitors effectively block HER-2 cleavage in HER-2 overexpressing human breast cancer cell lines. Intriguingly, when used in combination, INCB003619 dramatically enhances the antiproliferative activity of suboptimal doses of the anti-HER-2 antibody, trastuzumab, in HER-2 overexpressing/shedding breast cancer cell lines, accompanied by reduced ERK and AKT phosphorylation. Furthermore, INCB003619, in combination with trastuzumab, augments the pro-apoptotic and antiproliferative effects of the chemotherapeutic agent paclitaxel. Consistent with these in vitro data, INCB003619 reduces serum ECD levels and enhances the antitumor effect of trastuzumab in a xenograft tumor model derived from the HER-2 overexpressing BT-474 breast cancer cell line. Collectively, these findings suggest that blocking HER-2 cleavage with selective ADAM inhibitors may represent a novel therapeutic approach for treating HER-2 overexpressing breast cancer patients.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/drug therapy , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Transplantation, Heterologous , Trastuzumab
6.
Neoplasia ; 12(1): 28-38, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20072651

ABSTRACT

Cytokines in the bone marrow of multiple myeloma patients activate Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathways in tumor cells and promote tumor growth, survival, and drug resistance. INCB16562 was developed as a novel, selective, and orally bioavailable small-molecule inhibitor of JAK1 and JAK2 markedly selective over JAK3. The specific cellular activity of the inhibitor was demonstrated by its potent and dose-dependent inhibition of cytokine-dependent JAK/STAT signaling and cell proliferation in the absence of effects on Bcr-Abl-expressing cells. Treatment of myeloma cells with INCB16562 potently inhibited interleukin-6 (IL-6)-induced phosphorylation of STAT3. Moreover, the proliferation and survival of myeloma cells dependent on IL-6 for growth, as well as the IL-6-induced growth of primary bone marrow-derived plasma cells from a multiple myeloma patient, were inhibited by INCB16562. Induction of caspase activation and apoptosis was observed and attributed, at least in part, to the suppression of Mcl-1 expression. Importantly, INCB16562 abrogated the protective effects of recombinant cytokines or bone marrow stromal cells and sensitized myeloma cells to cell death by exposure to dexamethasone, melphalan, or bortezomib. Oral administration of INCB16562 antagonized the growth of myeloma xenografts in mice and enhanced the antitumor activity of relevant agents in combination studies. Taken together, these data suggest that INCB16562 is a potent JAK1/2 inhibitor and that mitigation of JAK/STAT signaling by targeting JAK1 and JAK2 will be beneficial in the treatment of myeloma patients, particularly in combination with other agents.


Subject(s)
Azepines/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Multiple Myeloma/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Stromal Cells/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Azepines/administration & dosage , Azepines/chemistry , Blotting, Western , Boronic Acids/administration & dosage , Bortezomib , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Humans , Interleukin-6/pharmacology , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Melphalan/administration & dosage , Mice , Mice, SCID , Molecular Structure , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Pyrazines/administration & dosage , Pyridines/administration & dosage , Pyridines/chemistry , STAT3 Transcription Factor/metabolism , Stromal Cells/cytology , Treatment Outcome , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 15(22): 6891-900, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19887489

ABSTRACT

PURPOSE: Deregulation of the Janus kinase-signal transducers and activators of transcription (JAK-STAT) pathway is a hallmark for the Philadelphia chromosome-negative myeloproliferative diseases polycythemia vera, essential thrombocythemia, and primary myelofibrosis. We tested the efficacy of a selective JAK1/2 inhibitor in cellular and in vivo models of JAK2-driven malignancy. EXPERIMENTAL DESIGN: A novel inhibitor of JAK1/2 was characterized using kinase assays. Cellular effects of this compound were measured in cell lines bearing the JAK2V617F or JAK1V658F mutation, and its antiproliferative activity against primary polycythemiavera patient cells was determined using clonogenic assays. Antineoplastic activity in vivo was determined using a JAK2V617F-driven xenograft model, and effects of the compound on survival, organomegaly, body weight, and disease-associated inflammatory markers were measured. RESULTS: INCB16562 potently inhibited proliferation of cell lines and primary cells from PV patients carrying the JAK2V617F or JAK1V658F mutation by blocking JAK-STAT signaling and inducing apoptosis. In vivo, INCB16562 reduced malignant cell burden, reversed splenomegaly and normalized splenic architecture, improved body weight gains, and extended survival in a model of JAK2V617F-driven hematologic malignancy. Moreover, these mice suffered from markedly elevated levels of inflammatory cytokines, similar to advanced myeloproliferative disease patients, which was reversed upon treatment. CONCLUSIONS: These data showed that administration of the dual JAK1/2 inhibitor INCB16562 reduces malignant cell burden, normalizes spleen size and architecture, suppresses inflammatory cytokines, improves weight gain, and extends survival in a rodent model of JAK2V617F-driven hematologic malignancy. Thus, selective inhibitors of JAK1 and JAK2 represent a novel therapy for the patients with myeloproliferative diseases and other neoplasms associated with JAK dysregulation.


Subject(s)
Enzyme Inhibitors/pharmacology , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mutation , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Inhibitory Concentration 50 , Kinetics , Mice , Neoplasm Transplantation , Polycythemia Vera/drug therapy
8.
Cancer Res ; 68(17): 7083-9, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18757423

ABSTRACT

The ErbB family of receptors is overexpressed in numerous human tumors. Overexpression correlates with poor prognosis and resistance to therapy. Use of ErbB-specific antibodies to the receptors (Herceptin or Erbitux) or ErbB-specific small-molecule inhibitors of the receptor tyrosine kinase activity (Iressa or Tarceva) has shown clinical efficacy in several solid tumors. An alternative method of affecting ErbB-initiated tumor growth and survival is to block sheddase activity. Sheddase activity is responsible for cleavage of multiple ErbB ligands and receptors, a necessary step in availability of the soluble, active form of the ligand and a constitutively activated ligand-independent receptor. This sheddase activity is attributed to the ADAM (a disintegrin and metalloprotease) family of proteins. ADAM 10 is the main sheddase of epidermal growth factor (EGF) and HER-2/neu cleavage, whereas ADAM17 is required for cleavage of additional EGF receptor (EGFR) ligands (transforming growth factor-alpha, amphiregulin, heregulin, heparin binding EGF-like ligand). This study has shown that addition of INCB3619, a potent inhibitor of ADAM10 and ADAM17, reduces in vitro HER-2/neu and amphiregulin shedding, confirming that it interferes with both HER-2/neu and EGFR ligand cleavage. Combining INCB3619 with a lapatinib-like dual inhibitor of EGFR and HER-2/neu kinases resulted in synergistic growth inhibition in MCF-7 and HER-2/neu-transfected MCF-7 human breast cancer cells. Combining the INCB7839 second-generation sheddase inhibitor with lapatinib prevented the growth of HER-2/neu-positive BT474-SC1 human breast cancer xenografts in vivo. These results suggest that there may be an additional clinical benefit of combining agents that target the ErbB pathways at multiple points.


Subject(s)
Disintegrins/antagonists & inhibitors , Metalloproteases/antagonists & inhibitors , Protease Inhibitors/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Line, Tumor , Female , Humans , Mice , Piperidines/pharmacology , Signal Transduction , Spiro Compounds/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL