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










Publication year range
1.
PLoS One ; 14(3): e0212670, 2019.
Article in English | MEDLINE | ID: mdl-30913212

ABSTRACT

Immunotherapy has fundamentally changed the landscape of cancer treatment. Despite the encouraging results with the checkpoint modulators, response rates vary widely across tumor types, with a majority of patients exhibiting either primary resistance without a significant initial response to treatment or acquired resistance with subsequent disease progression. Hematopoietic progenitor kinase 1 (HPK1) is predominantly expressed in hematopoietic cell linages and serves as a negative regulator in T cells and dendritic cells (DC). While HPK1 gene knockout (KO) studies suggest its role in anti-tumor immune responses, the involvement of kinase activity and thereof its therapeutic potential remain unknown. To investigate the potential of pharmacological intervention using inhibitors of HPK1, we generated HPK1 kinase dead (KD) mice which carry a single loss-of-function point mutation in the kinase domain and interrogated the role of kinase activity in immune cells in the context of suppressive factors or the tumor microenvironment (TME). Our data provide novel findings that HKP1 kinase activity is critical in conferring suppressive functions of HPK1 in a wide range of immune cells including CD4+, CD8+, DC, NK to Tregs, and inactivation of kinase domain was sufficient to elicit robust anti-tumor immune responses. These data support the concept that an HPK1 small molecule kinase inhibitor could serve as a novel agent to provide additional benefit in combination with existing immunotherapies, particularly to overcome resistance to current treatment regimens.


Subject(s)
Immunity, Cellular , Immunologic Surveillance , Lymphocytes/immunology , Neoplasms, Experimental/immunology , Protein Serine-Threonine Kinases/immunology , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Lymphocytes/pathology , Mice , Mice, Mutant Strains , Neoplasms, Experimental/genetics , Point Mutation , Protein Serine-Threonine Kinases/genetics , Tumor Microenvironment/genetics
2.
Clin Cancer Res ; 17(12): 4031-41, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21531814

ABSTRACT

PURPOSE: The extensive involvement of the HER kinases in epithelial cancer suggests that kinase inhibitors targeting this receptor family have the potential for broad spectrum antitumor activity. BMS-690514 potently inhibits all three HER kinases, and the VEGF receptor kinases. This report summarizes data from biochemical and cellular pharmacology studies, as well as antitumor activity of BMS-690514. EXPERIMENTAL DESIGN: The potency and selectivity of BMS-690514 was evaluated by using an extensive array of enzymatic and binding assays, as well as cellular assays that measure proliferation and receptor signaling. Antitumor activity was evaluated by using multiple xenograft models that depend on HER kinase signaling. The antiangiogenic properties of BMS-690514 were assessed in a matrigel plug assay, and effect on tumor blood flow was measured by dynamic contrast-enhanced MRI. RESULTS: BMS-690514 is a potent and selective inhibitor of epidermal growth factor receptor (EGFR), HER2, and HER4, as well as the VEGF receptor kinases. It inhibits proliferation of tumor cells with potency that correlates with inhibition of receptor signaling, and induces apoptosis in lung tumor cells that have an activating mutation in EGFR. Antitumor activity was observed with BMS-690514 at multiple doses that are well tolerated in mice. There was evidence of suppression of tumor angiogenesis and endothelial function by BMS-690514, which may contribute to its efficacy. CONCLUSIONS: By combining inhibition of two receptor kinase families, BMS-690524 is a novel targeted agent that disrupts signaling in the tumor and its vasculature.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Neoplasms/enzymology , Piperidines/pharmacology , Pyrroles/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Triazines/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/blood supply , Neovascularization, Pathologic/enzymology , Receptor, ErbB-2/metabolism , Regional Blood Flow/drug effects , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem Lett ; 21(2): 781-5, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21177105
4.
J Med Chem ; 52(21): 6527-30, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19821562

ABSTRACT

Structure-activity relationships in a series of 4-[1H-indazol-5-ylamino]pyrrolo[2,1-f][1,2,4]triazine-6-carbamates identified dual human epidermal growth factor receptor (HER)1/HER2 kinase inhibitors with excellent biochemical potency and kinase selectivity. On the basis of its favorable pharmacokinetic profile and robust in vivo activity in HER1 and HER2 driven tumor models, 13 (BMS-599626) was selected as a clinical candidate for treatment of solid tumors.


Subject(s)
Antineoplastic Agents/chemical synthesis , Carbamates/chemical synthesis , ErbB Receptors/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Triazines/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biological Availability , Carbamates/pharmacokinetics , Carbamates/pharmacology , Cell Line, Tumor , Dogs , Drug Screening Assays, Antitumor , Humans , Macaca fascicularis , Mice , Neoplasm Transplantation , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous , Triazines/pharmacokinetics , Triazines/pharmacology
5.
J Med Chem ; 52(5): 1251-4, 2009 Mar 12.
Article in English | MEDLINE | ID: mdl-19260711

ABSTRACT

Substituted N-(4-(2-aminopyridin-4-yloxy)-3-fluoro-phenyl)-1-(4-fluorophenyl)-2-oxo-1,2-dihydropyridine-3-carboxamides were identified as potent and selective Met kinase inhibitors. Substitution of the pyridine 3-position gave improved enzyme potency, while substitution of the pyridone 4-position led to improved aqueous solubility and kinase selectivity. Analogue 10 demonstrated complete tumor stasis in a Met-dependent GTL-16 human gastric carcinoma xenograft model following oral administration. Because of its excellent in vivo efficacy and favorable pharmacokinetic and preclinical safety profiles, 10 has been advanced into phase I clinical trials.


Subject(s)
Aminopyridines/chemical synthesis , Antineoplastic Agents/chemical synthesis , Dihydropyridines/chemical synthesis , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridones/chemical synthesis , Administration, Oral , Aminopyridines/pharmacokinetics , Aminopyridines/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Dihydropyridines/pharmacokinetics , Dihydropyridines/pharmacology , Dogs , Humans , Mice , Mice, Nude , Models, Molecular , Pyridones/pharmacokinetics , Pyridones/pharmacology , Rats , Solubility , Structure-Activity Relationship , Xenograft Model Antitumor Assays
6.
Bioorg Med Chem Lett ; 17(17): 4947-54, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17606372

ABSTRACT

Pyrrolotriazine dual EGFR/HER2 kinase inhibitors with a 5-((4-aminopiperidin-1-yl)methyl) solubilizing group were found to be superior to analogs with previously reported C-5 solubilizing groups. New synthetic methodology was developed for the parallel synthesis of C-4 analogs with the new solubilizing group. Interesting new leads were evaluated in tumor xenograft models and the C-4 aminofluorobenzylindazole, 1c, was found to exhibit the best antitumor activity. It is hypothesized that this solubilizing group extends into the ribose-phosphate portion of the ATP binding pocket and enhances the binding affinity of the inhibitor.


Subject(s)
Chemistry, Pharmaceutical/methods , ErbB Receptors/chemistry , Neoplasms/drug therapy , Piperidines/chemical synthesis , Pyrroles/chemical synthesis , Receptor, ErbB-2/chemistry , Triazines/chemical synthesis , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , Humans , Inhibitory Concentration 50 , Insecta , Models, Chemical , Neoplasm Transplantation , Piperidines/chemistry , Piperidines/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Triazines/chemistry , Triazines/pharmacology
7.
Bioorg Med Chem Lett ; 17(10): 2828-33, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17368025

ABSTRACT

Novel C-5 aminomethyl pyrrolotriazines were prepared and optimized for dual EGFR and HER2 protein tyrosine kinase inhibition. The homopiperazine, 1p, emerged as a key lead and it showed promising oral efficacy in EGFR and dual EGFR/HER2 driven human tumor xenograft models. It is hypothesized that the C-5 homopiperazine side chain binds in the ribose-phosphate portion of the ATP binding pocket.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Methylamines/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrroles/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Triazines/pharmacology , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Screening Assays, Antitumor , Humans , Methylamines/chemistry , Mice , Models, Molecular , Molecular Structure , Neoplasm Transplantation , Pyrroles/chemistry , Structure-Activity Relationship , Triazines/chemistry , Xenograft Model Antitumor Assays
9.
Clin Cancer Res ; 12(20 Pt 1): 6186-93, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17062696

ABSTRACT

PURPOSE: The studies described here are intended to characterize the ability of BMS-599626, a small-molecule inhibitor of the human epidermal growth factor receptor (HER) kinase family, to modulate signaling and growth of tumor cells that depend on HER1 and/or HER2. EXPERIMENTAL DESIGN: The potency and selectivity of BMS-599626 were assessed in biochemical assays using recombinant protein kinases, as well as in cell proliferation assays using tumor cell lines with varying degrees of dependence on HER1 or HER2 signaling. Modulation of receptor signaling was determined in cell assays by Western blot analyses of receptor autophosphorylation and downstream signaling. The ability of BMS-599626 to inhibit receptor heterodimer signaling in tumor cells was studied by receptor coimmunoprecipitation. Antitumor activity of BMS-599626 was evaluated using a number of different xenograft models that represent a spectrum of human tumors with HER1 or HER2 overexpression. RESULTS: BMS-599626 inhibited HER1 and HER2 with IC50 of 20 and 30 nmol/L, respectively, and was highly selective when tested against a broad panel of diverse protein kinases. Biochemical studies suggested that BMS-599626 inhibited HER1 and HER2 through distinct mechanisms. BMS-599626 abrogated HER1 and HER2 signaling and inhibited the proliferation of tumor cell lines that are dependent on these receptors, with IC50 in the range of 0.24 to 1 micromol/L. BMS-599626 was highly selective for tumor cells that depend on HER1/HER2 and had no effect on the proliferation of cell lines that do not express these receptors. In tumor cells that are capable of forming HER1/HER2 heterodimers, BMS-599626 inhibited heterodimerization and downstream signaling. BMS-599626 had antitumor activity in models that overexpress HER1 (GEO), as well as in models that have HER2 gene amplification (KPL4) or overexpression (Sal2), and there was good correlation between the inhibition of receptor signaling and antitumor activity. CONCLUSIONS: BMS-599626 is a highly selective and potent inhibitor of HER1 and HER2 kinases and inhibits tumor cell proliferation through modulation of receptor signaling. BMS-599626 inhibits HER1/HER2 receptor heterodimerization and provides an additional mechanism of inhibiting tumors in which receptor coexpression and heterodimerization play a major role in driving tumor growth. The preclinical data support the advancement of BMS-599626 into clinical development for the treatment of cancer.


Subject(s)
Antineoplastic Agents/toxicity , Enzyme Inhibitors/toxicity , ErbB Receptors/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , CD8 Antigens/immunology , Cell Division/drug effects , Cell Line , Cell Line, Tumor , Dimerization , Humans
10.
Mol Cancer Ther ; 5(6): 1571-6, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16818516

ABSTRACT

Receptor tyrosine kinases (RTK) remain an area of therapeutic interest because of their role in epithelial tumors, and experimental models specific to these targets are highly desirable. Chimeric receptors were prepared by in-frame fusion of the CD8 extracellular sequence with the cytoplasmic sequences of RTKs. A CD8HER2 fusion protein was shown to form disulfide-mediated homodimers and to transform fibroblasts and epithelial cells. CD8RTK fusion proteins transform rat kidney epithelial cells and impart phenotypes that may reflect signaling specificity inherent in the native receptors. Transgenic expression of CD8HER2 and CD8Met in mice resulted in the formation of salivary and mammary gland tumors. The transgenic tumors allow the derivation of allograft tumors and cell lines that are sensitive to inhibition by small molecule kinase inhibitors. This approach provides excellent cell and tumor models for the characterization of signaling properties of diverse RTKs and for the evaluation of rationally designed antagonists targeting these kinases.


Subject(s)
CD8 Antigens/metabolism , Gene Expression Regulation, Neoplastic/physiology , Mammary Neoplasms, Animal/genetics , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/genetics , Salivary Gland Neoplasms/genetics , Animals , Blotting, Western , Cell Transformation, Neoplastic/genetics , Dimerization , Disease Models, Animal , Disulfides/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/etiology , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Peptide Fragments/immunology , Plasmids , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Rats , Receptor Protein-Tyrosine Kinases/metabolism , Salivary Gland Neoplasms/drug therapy , Salivary Gland Neoplasms/etiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...