Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Mol Cancer Ther ; 14(2): 620-30, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25527633

ABSTRACT

Insulin-like growth factor receptor 1 (IGF-1R)-targeting therapies are currently at an important crossroad given the low clinical response rates seen in unselected patients. Predictive biomarkers for patient selection are critical for improving clinical benefit. Coupling in vitro sensitivity testing of BMS-754807, a dual IGF-1R/IR inhibitor, with genomic interrogations in 60 human colorectal cancer cell lines, we identified biomarkers correlated with response to BMS-754807. The results showed that cell lines with BRAF(V600E) or KRAS(G13D) mutation were resistant, whereas cell lines with wild-type of both KRAS and BRAF were particularly sensitive to BMS-754807 if they have either higher RNA expression levels of IR-A or lower levels of IGFBP6. In addition, the cell lines with KRAS mutations, those with either insulin receptor substrate 2 (IRS2) copy number gain (CNG) or higher IGF-1R expression levels, were more sensitive to the drug. Furthermore, cell lines with IRS2 CNG had higher levels of ligand-stimulated activation of IGF-1R and AKT, suggesting that these cell lines with IGF-IR signaling pathways more actively coupled to AKT signaling are more responsive to IGF-1R/IR inhibition. IRS2 siRNA knockdown reduced IRS2 protein expression levels and decreased sensitivity to BMS-754807, providing evidence for the functional involvement of IRS2 in mediating the drug response. The prevalence of IRS2 CNG in colorectal cancer tumors as measured by qPCR-CNV is approximately 35%. In summary, we identified IRS2 CNG, IGF-1R, IR-A, and IGFBP6 RNA expression levels, and KRAS and BRAF mutational status as candidate predictive biomarkers for response to BMS-754807. This work proposed clinical development opportunities for BMS-754807 in colorectal cancer with patient selection to improve clinical benefit.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/genetics , Gene Dosage , Insulin Receptor Substrate Proteins/genetics , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins/genetics , Pyrazoles/pharmacology , Triazines/pharmacology , ras Proteins/genetics , Blotting, Western , Cell Line, Tumor , DNA Copy Number Variations/genetics , DNA Mutational Analysis , Gene Amplification/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 6/metabolism , Ligands , Models, Biological , Phosphorylation/drug effects , Proto-Oncogene Proteins p21(ras) , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, Insulin/antagonists & inhibitors
2.
Mol Cancer Ther ; 12(12): 2909-16, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24130056

ABSTRACT

Ovarian cancer mortality ranks highest among all gynecologic cancers with growth factor pathways playing an integral role in tumorigenesis, metastatic dissemination, and therapeutic resistance. The HER and VEGF receptor (VEGFR) are both overexpressed and/or aberrantly activated in subsets of ovarian tumors. While agents targeting either the HER or VEGF pathways alone have been investigated, the impact of these agents have not led to overall survival benefit in ovarian cancer. We tested the hypothesis that cotargeting HER and VEGFR would maximize antitumor efficacy at tolerable doses. To this end, ovarian cancer xenografts grown intraperitoneally in athymic nude mice were tested in response to AC480 (pan-HER inhibitor, "HERi"), cediranib (pan-VEGFR inhibitor "VEGFRi"), or BMS-690514 (combined HER/VEGFR inhibitor "EVRi"). EVRi was superior to both HERi and VEGFRi in terms of tumor growth, final tumor weight, and progression-free survival. Correlative tumor studies employing phosphoproteomic antibody arrays revealed distinct agent-specific alterations, with EVRi inducing the greatest overall effect on growth factor signaling. These data suggest that simultaneous inhibition of HER and VEGFR may benefit select subsets of ovarian cancer tumors. To this end, we derived a novel HER/VEGF signature that correlated with poor overall survival in high-grade, late stage, serous ovarian cancer patient tumors.


Subject(s)
Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Quinazolines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cluster Analysis , Disease Models, Animal , Disease Progression , Female , Humans , Mice , Neoplasm Grading , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Piperidines/administration & dosage , Piperidines/pharmacology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Proteomics , Pyrroles/administration & dosage , Pyrroles/pharmacology , Quinazolines/administration & dosage , Signal Transduction , Triazines/administration & dosage , Triazines/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Mol Cancer Res ; 9(11): 1551-61, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21954435

ABSTRACT

Emerging literature suggests that metabolic pathways play an important role in the maintenance and progression of human cancers. In particular, recent studies have implicated lipid biosynthesis and desaturation as a requirement for tumor cell survival. In the studies reported here, we aimed to understand whether tumor cells require the activity of either human isoform of stearoyl-CoA-desaturase (SCD1 or SCD5) for survival. Inhibition of SCD1 by siRNA or a small molecule antagonist results in strong induction of apoptosis and growth inhibition, when tumor cells are cultured in reduced (2%) serum conditions, but has little impact on cells cultured in 10% serum. Depletion of SCD5 had minimal effects on cell growth or apoptosis. Consistent with the observed dependence on SCD1, but not SCD5, levels of SCD1 protein increased in response to decreasing serum levels. Both induction of SCD1 protein and sensitivity to growth inhibition by SCD1 inhibition could be reversed by supplementing growth media with unsaturated fatty acids, the product of the enzymatic reaction catalyzed by SCD1. Transcription profiling of cells treated with an SCD inhibitor revealed strong induction of markers of endoplasmic reticulum stress. Underscoring its importance in cancer, SCD1 protein was found to be highly expressed in a large percentage of human cancer specimens. SCD inhibition resulted in tumor growth delay in a human gastric cancer xenograft model. Altogether, these results suggest that desaturated fatty acids are required for tumor cell survival and that SCD may represent a viable target for the development of novel agents for cancer therapy.


Subject(s)
Fatty Acids, Unsaturated/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Stearoyl-CoA Desaturase/antagonists & inhibitors , Amino Acid Sequence , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Survival/physiology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/pathology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Stearoyl-CoA Desaturase/biosynthesis , Stearoyl-CoA Desaturase/deficiency , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Transfection
5.
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
6.
Bioorg Med Chem Lett ; 21(2): 781-5, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21177105
7.
MAbs ; 3(1): 38-48, 2011.
Article in English | MEDLINE | ID: mdl-21099371

ABSTRACT

Engineered domains of human fibronectin (Adnectins™) were used to generate a bispecific Adnectin targeting epidermal growth factor receptor (EGFR) and insulin-like growth factor-I receptor (IGF-IR), two transmembrane receptors that mediate proliferative and survival cell signaling in cancer. Single-domain Adnectins that specifically bind EGFR or IGF-IR were generated using mRNA display with a library containing as many as 10 ( 13) Adnectin variants. mRNA display was also used to optimize lead Adnectin affinities, resulting in clones that inhibited EGFR phosphorylation at 7 to 38 nM compared to 2.6 µM for the parental clone. Individual, optimized, Adnectins specific for blocking either EGFR or IGF-IR signaling were engineered into a single protein (EI-Tandem Adnectin). The EI-Tandems inhibited phosphorylation of EGFR and IGF-IR, induced receptor degradation, and inhibited down-stream cell signaling and proliferation of human cancer cell lines (A431, H292, BxPC3 and RH41) with IC 50 values ranging from 0.1 to 113 nM. Although Adnectins bound to EGFR at a site distinct from those of anti-EGFR antibodies cetuximab, panitumumab and nimotuzumab, like the antibodies, the anti-EGFR Adnectins blocked the binding of EGF to EGFR. PEGylated EI-Tandem inhibited the growth of both EGFR and IGF-IR driven human tumor xenografts, induced degradation of EGFR, and reduced EGFR phosphorylation in tumors. These results demonstrate efficient engineering of bispecific Adnectins with high potency and desired specificity. The bispecificity may improve biological activity compared to monospecific biologics as tumor growth is driven by multiple growth factors. Our results illustrate a technological advancement for constructing multi-specific biologics in cancer therapy.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Fibronectins/chemistry , Peptide Fragments/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , ErbB Receptors/metabolism , Female , Humans , Immunoblotting , Kinetics , Mice , Mice, Nude , Molecular Sequence Data , Panitumumab , Peptide Fragments/metabolism , Phosphorylation/drug effects , Protein Binding , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
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
9.
Mol Cancer Ther ; 7(9): 2589-98, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18765823

ABSTRACT

We have reported previously the activity of the insulin-like growth factor-I (IGF-IR)/insulin receptor (InsR) inhibitor, BMS-554417, in breast and ovarian cancer cell lines. Further studies indicated treatment of OV202 ovarian cancer cells with BMS-554417 increased phosphorylation of HER-2. In addition, treatment with the pan-HER inhibitor, BMS-599626, resulted in increased phosphorylation of IGF-IR, suggesting a reciprocal cross-talk mechanism. In a panel of five ovarian cancer cell lines, simultaneous treatment with the IGF-IR/InsR inhibitor, BMS-536924 and BMS-599626, resulted in a synergistic antiproliferative effect. Furthermore, combination therapy decreased AKT and extracellular signal-regulated kinase activation and increased biochemical and nuclear morphologic changes consistent with apoptosis compared with either agent alone. In response to treatment with BMS-536924, increased expression and activation of various members of the HER family of receptors were seen in all five ovarian cancer cell lines, suggesting that inhibition of IGF-IR/InsR results in adaptive up-regulation of the HER pathway. Using MCF-7 breast cancer cell variants that overexpressed HER-1 or HER-2, we then tested the hypothesis that HER receptor expression is sufficient to confer resistance to IGF-IR-targeted therapy. In the presence of activating ligands epidermal growth factor or heregulin, respectively, MCF-7 cells expressing HER-1 or HER-2 were resistant to BMS-536924 as determined in a proliferation and clonogenic assay. These data suggested that simultaneous treatment with inhibitors of the IGF-I and HER family of receptors may be an effective strategy for clinical investigations of IGF-IR inhibitors in breast and ovarian cancer and that targeting HER-1 and HER-2 may overcome clinical resistance to IGF-IR inhibitors.


Subject(s)
Benzimidazoles/pharmacology , Drug Resistance, Neoplasm/drug effects , Pyridones/pharmacology , Receptor, ErbB-2/metabolism , Receptor, IGF Type 1/antagonists & inhibitors , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor Cross-Talk/drug effects , Receptor, ErbB-2/antagonists & inhibitors , Receptor, Insulin/antagonists & inhibitors
10.
Bioorg Med Chem Lett ; 18(6): 1945-51, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18289854

ABSTRACT

An amide library derived from the pyrrolo[2,1-f][1,2,4]triazine scaffold led to the identification of modest inhibitors of Met kinase activity. Introduction of polar side chains at C-6 of the pyrrolotriazine core provided significant improvements in in vitro potency. The amide moiety could be replaced with acylurea and malonamide substituents to give compounds with improved potency in the Met-driven GTL-16 human gastric carcinoma cell line. Acylurea pyrrolotriazines with substitution at C-5 demonstrated single digit nanomolar kinase activity. X-ray crystallography revealed that the C-5 substituted pyrrolotriazines bind to the Met kinase domain in an ATP-competitive manner.


Subject(s)
Enzyme Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrroles/chemistry , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Triazines/chemistry , Animals , Caco-2 Cells/drug effects , Cell Proliferation/drug effects , Cells, Cultured/drug effects , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Glutathione Transferase/antagonists & inhibitors , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Structure , Protein Conformation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-met , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Stomach Neoplasms/blood , Stomach Neoplasms/enzymology , Structure-Activity Relationship
11.
J Clin Oncol ; 25(22): 3230-7, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17664471

ABSTRACT

PURPOSE: The antiepidermal growth factor receptor (EGFR) antibody cetuximab shows activity in multiple epithelial tumor types; however, responses are seen in only a subset of patients. This study was conducted to identify markers that are associated with disease control in patients treated with cetuximab. PATIENTS AND METHODS: One hundred ten patients with metastatic colorectal cancer were enrolled onto a cetuximab monotherapy trial. Transcriptional profiling was conducted on RNA from mandatory pretreatment metastatic biopsies to identify genes whose expression correlates with best clinical responses. EGFR and K-ras mutation analyses and EGFR gene copy number analyses were performed on DNA from pretreatment biopsies. RESULTS: Gene expression profiles showed that patients with tumors that express high levels of the EGFR ligands epiregulin and amphiregulin are more likely to have disease control with cetuximab (EREG, P = .000015; AREG, P = .000025). Additionally, patients whose tumors do not have K-ras mutations have a significantly higher disease control rate than patients with K-ras mutations (P = .0003). Furthermore, patients with tumors that have high expression of EREG or AREG also have significantly longer progression-free survival (PFS) than patients with low expression (EREG: P = .0002, hazard ratio [HR] = 0.47, and median PFS, 103.5 v 57 days, respectively; AREG: P < .0001, HR = 0.44, and median PFS, 115.5 v 57 days, respectively). CONCLUSION: Patients with tumors that have high gene expression levels of epiregulin and amphiregulin and patients with wild-type K-ras are more likely to have disease control on cetuximab treatment. The identified markers could be developed further to select patients for cetuximab therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Epidermal Growth Factor/genetics , Genes, ras , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mutation , Adult , Aged , Aged, 80 and over , Amphiregulin , Antibodies, Monoclonal, Humanized , Biomarkers, Tumor/analysis , Cetuximab , EGF Family of Proteins , Enzyme-Linked Immunosorbent Assay , Epidermal Growth Factor/metabolism , Epiregulin , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Profiling , Glycoproteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Neoplasm Metastasis , Predictive Value of Tests , Proportional Hazards Models , Survival Rate , Treatment Outcome
12.
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
13.
Cancer Res ; 67(5): 2226-38, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17332353

ABSTRACT

Dasatinib is a multitargeted kinase inhibitor that was recently approved for the treatment of chronic myelogenous leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia with resistance or intolerance to prior therapy. It is also in clinical trials for treating patients with solid tumors. The identification of molecular markers predictive of response to dasatinib could assist in clinical development by selecting patients most likely to derive clinical benefit. Using baseline gene expression profiling of a panel of 23 breast cancer cell lines, we identified genomic signatures highly correlated with in vitro sensitivity to dasatinib. The ability of these signatures to predict dasatinib sensitivity was further confirmed and validated in independent test cell lines. A six-gene model was used to correctly predict dasatinib sensitivity in 11 out of 12 (92%) additional breast and 19 out of 23 (83%) lung cancer cell lines. Quantitative real-time PCR and immunohistochemical assays further confirmed the differential expression pattern of selected markers. Finally, these gene signatures were observed in a subset of primary breast, lung, and ovarian tumors suggesting potential utility in patient selection. The subset of breast cancer patients expressing the dasatinib-sensitive signature includes a distinct clinical and molecular subgroup: the so-called "triple negative" (i.e., estrogen receptor-negative, progesterone receptor-negative, and HER2-negative) or "basal" breast cancer subtype. This patient population has a poor prognosis and currently has few effective treatment options. Our results implicate that dasatinib may represent a valuable treatment option in this difficult-to-treat population. To test this hypothesis, clinical studies are now under way to determine the activity of dasatinib in these patients.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Patient Selection , Pyrimidines/therapeutic use , Thiazoles/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Cell Line, Tumor , Dasatinib , Gene Expression Profiling , Humans , Models, Genetic , Prognosis
14.
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
15.
Mol Cell Biol ; 27(8): 3165-75, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17296734

ABSTRACT

Type I insulin-like growth factor receptor (IGF-IR) can transform mouse fibroblasts; however, little is known about the transforming potential of IGF-IR in human fibroblasts or epithelial cells. We found that overexpression of a constitutively activated IGF-IR (CD8-IGF-IR) was sufficient to cause transformation of immortalized human mammary epithelial cells and growth in immunocompromised mice. Furthermore, CD8-IGF-IR caused cells to undergo an epithelial-to-mesenchymal transition (EMT) which was associated with dramatically increased migration and invasion. The EMT was mediated by the induction of the transcriptional repressor Snail and downregulation of E-cadherin. NF-kappaB was highly active in CD8-IGF-IR-MCF10A cells, and both increased levels of Snail and the EMT were partially reversed by blocking NF-kappaB or IGF-IR activity. This study places IGF-IR among a small group of oncogenes that, when overexpressed alone, can confer in vivo tumorigenic growth of MCF10A cells and indicates the hierarchy in the mechanism of IGF-IR-induced EMT.


Subject(s)
Cell Transformation, Neoplastic , Epithelial Cells/cytology , Mammary Glands, Human/cytology , Mesoderm/cytology , NF-kappa B/metabolism , Receptor, IGF Type 1/metabolism , Transcription Factors/metabolism , Animals , Benzimidazoles/pharmacology , CD8 Antigens/metabolism , Cadherins/genetics , Cell Transformation, Neoplastic/drug effects , Collagen/drug effects , Down-Regulation/drug effects , Drug Combinations , Epithelial Cells/drug effects , Genes, Regulator , Humans , Laminin/drug effects , Mammary Glands, Human/drug effects , Mammary Glands, Human/growth & development , Mice , Models, Biological , Morphogenesis/drug effects , Proteoglycans/drug effects , Pyridones/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Snail Family Transcription Factors , Transplantation, Heterologous
17.
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
18.
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
19.
Clin Cancer Res ; 11(15): 5558-65, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16061873

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFR), a protein tyrosine kinase expressed in many types of human cancers, has been strongly associated with tumor progression. Cetuximab is an IgG(1) anti-EGFR chimeric mouse/human monoclonal antibody that has been approved for the treatment of advanced colon cancer. Using human tumor xenografts grown in nude mice, we have determined the in vivo pharmacodynamic response of cetuximab at efficacious doses. Three pharmacodynamic end points were evaluated: tumoral phospho-EGFR, tumoral mitogen-activated protein kinase (MAPK) phosphorylation, and Ki67 expression. EXPERIMENTAL DESIGN: The pharmacodynamic study was conducted in nude mice bearing Geo tumors following a single i.p. administration of 0.25 and 0.04 mg. The tumors were analyzed by immunohistochemistry. The levels of phospho-EGFR were quantitated by an ELISA assay. RESULTS: At 0.25 mg, phospho-EGFR was maximally inhibited by 91% at 24 hours, whereas the level of inhibition decreased to 72% by 72 hours. At 0.04 mg, the maximum inhibition of phospho-EGFR was 53% at 24 hours, whereas the level of inhibition decreased to 37% by 72 hours. The time course of phospho-EGFR inhibition and recovery seemed to correlate with the pharmacokinetics of cetuximab. Immunohistochemical analysis showed that phospho-MAPK and Ki67 expression were inhibited between 24 and 72 hours at 0.25 and 0.04 mg. A pharmacokinetic/pharmacodynamic model was established and predicted that the plasma concentration of cetuximab required to inhibit 90% of phospho-EGFR was 67.5 mug/mL. CONCLUSIONS: Phospho-EGFR/phospho-MAPK could be useful clinical biomarkers to assess EGFR inhibition by cetuximab.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Biomarkers, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Xenograft Model Antitumor Assays/methods , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cetuximab , Enzyme-Linked Immunosorbent Assay , ErbB Receptors/metabolism , Female , Humans , Immunoglobulin G/chemistry , Immunohistochemistry , Ki-67 Antigen/biosynthesis , MAP Kinase Signaling System , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation , Time Factors
20.
Bioorg Med Chem Lett ; 15(21): 4774-9, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16111887

ABSTRACT

A novel series of dual EGFR and HER2 inhibitors based on the pyrrolo[2,1-f][1,2,4]triazine nucleus is described. A general route toward their synthesis, which enables functionalization at multiple sites, has been developed. Biological evaluation in enzymatic and cell-based assays has identified a series of C-6 carbamates with potent biochemical and cellular activities.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Binding Sites , Carbamates/chemical synthesis , Carbamates/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Models, Molecular , Structure-Activity Relationship , Triazines/chemical synthesis , Triazines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...