Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Breast Cancer Res ; 18(1): 37, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27009091

ABSTRACT

BACKGROUND: Inflammatory breast cancer (IBC) is an aggressive type of advanced breast cancer with a poor prognosis. We recently found that focal adhesion kinase 1 (FAK1) is upregulated and phosphorylated (active) in IBC. In this study, we investigated the effect of CEP-37440, a dual inhibitor of FAK1 and anaplastic lymphoma kinase (ALK), using human IBC cell lines and preclinical models of IBC. METHODS: Cell proliferation assays were performed in the presence of several concentrations of CEP-37440 using IBC and triple-negative breast cancer non-IBC cell lines. In vitro, we studied the expression of total FAK1, phospho-FAK1 (Tyr 397), total ALK and phospho-ALK (Tyr 1604). In vivo, we tested CEP-37440 using FC-IBC02, SUM149, and SUM190 IBC xenograft mouse models. RESULTS: CEP-37440 at low concentration decreased the proliferation of the IBC cell lines FC-IBC02, SUM190, and KPL4, while not affecting the proliferation of normal breast epithelial cells. At higher concentration, CEP-37440 was also able to inhibit the proliferation of the IBC cell line MDA-IBC03 and the triple-negative non-IBC cell lines MDA-MB-231 and MDA-MB-468; the IBC cell line SUM149 showed a slight response to the drug. CEP-37440 decreased the cell proliferation of FC-IBC02, SUM190, and KPL4 by blocking the autophosphorylation kinase activity of FAK1 (Tyr 397). None of the cells evaluated expressed ALK. In vivo, after 7 weeks of CEP-37440 treatment, the SUM190, FC-IBC02, and SUM149 breast tumor xenografts were smaller in mice treated with 55 mg/kg bid CEP-37440 compared to the controls; the tumor growth inhibition (TGI) was 79.7 %, 33 %, and 23 %, respectively. None of the FC-IBC02 breast xenografts mice treated with CEP-37440 developed brain metastasis while 20 % of the mice in the control group developed brain metastasis. Expression array analyses in FC-IBC02 cells showed that CEP-37440 affects the expression of genes related to apoptosis, interferon signaling, and cytokines. CONCLUSIONS: CEP-37440 is effective against some IBC cells that express phospho-FAK1 (Tyr 397), and its antiproliferative activity is related to its ability to decrease phospho-FAK1. Our results suggest that combinational therapies could be more effective than using CEP-37440 as a single agent.


Subject(s)
Benzamides/administration & dosage , Benzocycloheptenes/administration & dosage , Enzyme Inhibitors/administration & dosage , Focal Adhesion Protein-Tyrosine Kinases/genetics , Inflammatory Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/drug therapy , Anaplastic Lymphoma Kinase , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inflammatory Breast Neoplasms/genetics , Inflammatory Breast Neoplasms/pathology , Mice , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
2.
Bioorg Med Chem Lett ; 25(5): 1047-52, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25655723

ABSTRACT

The diastereoselective synthesis and biological activity of piperidine-3,4-diol and piperidine-3-ol-derived pyrrolotriazine inhibitors of anaplastic lymphoma kinase (ALK) are described. Although piperidine-3,4-diol and piperidine-3-ol derivatives showed comparable in vitro ALK activity, the latter subset of inhibitors demonstrated improved physiochemical and pharmacokinetic properties. Furthermore, the stereochemistry of the C3 and C4 centers had a marked impact on the in vivo inhibition of ALK autophosphorylation. Thus, trans-4-aryl-piperidine-3-ols (22) were more potent than the cis diastereomers (20).


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Lymphoma, Large-Cell, Anaplastic/drug therapy , Pyrroles/chemistry , Pyrroles/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Triazines/chemistry , Triazines/therapeutic use , Anaplastic Lymphoma Kinase , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Crystallography, X-Ray , Humans , Lymphoma, Large-Cell, Anaplastic/enzymology , Mice, SCID , Models, Molecular , Piperidines/chemistry , Piperidines/pharmacokinetics , Piperidines/therapeutic use , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Pyrroles/pharmacokinetics , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/metabolism , Triazines/pharmacokinetics
3.
J Immunol ; 187(7): 3840-53, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21880982

ABSTRACT

Accumulating evidence suggests that autoreactive plasma cells play an important role in systemic lupus erythematosus (SLE). In addition, several proinflammatory cytokines promote autoreactive B cell maturation and autoantibody production. Hence, therapeutic targeting of such cytokine pathways using a selective JAK2 inhibitor, CEP-33779 (JAK2 enzyme IC(50) = 1.3 nM; JAK3 enzyme IC(50)/JAK2 enzyme IC(50) = 65-fold), was tested in two mouse models of SLE. Age-matched, MRL/lpr or BWF1 mice with established SLE or lupus nephritis, respectively, were treated orally with CEP-33779 at 30 mg/kg (MRL/lpr), 55 mg/kg or 100 mg/kg (MRL/lpr and BWF1). Studies included reference standard, dexamethasone (1.5 mg/kg; MRL/lpr), and cyclophosphamide (50 mg/kg; MRL/lpr and BWF1). Treatment with CEP-33779 extended survival and reduced splenomegaly/lymphomegaly. Several serum cytokines were significantly decreased upon treatment including IL-12, IL-17A, IFN-α, IL-1ß, and TNF-α. Anti-nuclear Abs and frequencies of autoantigen-specific, Ab-secreting cells declined upon CEP-33779 treatment. Increased serum complement levels were associated with reduced renal JAK2 activity, histopathology, and spleen CD138(+) plasma cells. The selective JAK2 inhibitor CEP-33779 was able to mitigate several immune parameters associated with SLE advancement, including the protection and treatment of mice with lupus nephritis. These data support the possibility of using potent, orally active, small-molecule inhibitors of JAK2 to treat the debilitative disease SLE.


Subject(s)
Enzyme Inhibitors/therapeutic use , Janus Kinase 2/antagonists & inhibitors , Lupus Nephritis/drug therapy , Plasma Cells/drug effects , Pyridines/therapeutic use , Triazoles/therapeutic use , Administration, Oral , Animals , Antibodies, Antinuclear/blood , Autoimmunity/drug effects , Autoimmunity/immunology , Cell Separation , Cytokines/blood , Disease Models, Animal , Enzyme Inhibitors/pharmacokinetics , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Mice , Mice, Inbred MRL lpr , Plasma Cells/immunology , Pyridines/pharmacokinetics , Triazoles/pharmacokinetics
4.
Bioorg Med Chem Lett ; 21(1): 463-6, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21074994

ABSTRACT

The synthesis and biological evaluation of potent and selective anaplastic lymphoma kinase (ALK) inhibitors from a novel class of 2,4-diaminopyrimidines, incorporating 2,3,4,5-tetrahydro-benzo[d]azepine fragments, is described. An orally bioavailable analogue (18) that displayed antitumor efficacy in ALCL xenograft models in mice was identified and extensively profiled.


Subject(s)
Benzazepines/chemistry , Lymphoma, Large-Cell, Anaplastic/drug therapy , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/chemistry , Administration, Oral , Anaplastic Lymphoma Kinase , Animals , Benzazepines/pharmacokinetics , Benzazepines/therapeutic use , Mice , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases , Xenograft Model Antitumor Assays
5.
Proc Natl Acad Sci U S A ; 105(52): 20852-7, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19088198

ABSTRACT

The mechanisms of malignant cell transformation caused by the oncogenic, chimeric nucleophosmin (NPM)/anaplastic lymphoma kinase (ALK) remain only partially understood, with most of the previous studies focusing mainly on the impact of NPM/ALK on cell survival and proliferation. Here we report that the NPM/ALK-carrying T cell lymphoma (ALK+TCL) cells strongly express the immunosuppressive cell-surface protein CD274 (PD-L1, B7-H1), as determined on the mRNA and protein level. The CD274 expression is strictly dependent on the expression and enzymatic activity of NPM/ALK, as demonstrated by inhibition of the NPM/ALK function in ALK+TCL cells by the small molecule ALK inhibitor CEP-14083 and by documenting CD274 expression in IL-3-depleted BaF3 cells transfected with the wild-type NPM/ALK, but not the kinase-inactive NPM/ALK K210R mutant or empty vector alone. NPM/ALK induces CD274 expression by activating its key signal transmitter, transcription factor STAT3. STAT3 binds to the CD274 gene promoter in vitro and in vivo, as shown in the gel electromobility shift and chromatin immunoprecipitation assays, and is required for the PD-L1 gene expression, as demonstrated by siRNA-mediated STAT3 depletion. These findings identify an additional cell-transforming property of NPM/ALK and describe a direct link between an oncoprotein and an immunosuppressive cell-surface protein. These results also provide an additional rationale to therapeutically target NPM/ALK and STAT3 in ALK+TCL. Finally, they suggest that future immunotherapeutic protocols for this type of lymphoma may need to include the inhibition of NPM/ALK and STAT3 to achieve optimal clinical efficacy.


Subject(s)
Antigens, CD/biosynthesis , Gene Expression Regulation, Leukemic , Lymphoma, T-Cell/metabolism , Oncogene Proteins, Fusion/metabolism , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/biosynthesis , Antigens, CD/genetics , B7-H1 Antigen , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , Humans , Lymphoma, T-Cell/genetics , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , RNA, Small Interfering/genetics , STAT3 Transcription Factor/genetics
6.
Bioorg Med Chem Lett ; 20(11): 3356-60, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20430619

ABSTRACT

A novel series of 8-(2-tetrahydropyranyl)-12,13-dihydroindazolo[5,4-a]pyrrolo[3,4-c]carbazoles (THP-DHI) was synthesized and evaluated as dual TIE-2 and VEGF-R2 receptor tyrosine kinase inhibitors. Development of the structure-activity relationships (SAR) with the support of X-ray crystallography led to identification of 7f and 7g as potent, selective dual TIE-2/VEGF-R2 inhibitors with excellent cellular potency and acceptable pharmacokinetic properties. Compounds 7f and 7g were orally active in tumor models with no observed toxicity.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Receptor, TIE-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Cells, Cultured , Crystallography, X-Ray , Humans , Models, Molecular , Protein Kinase Inhibitors/pharmacokinetics , Rats , Structure-Activity Relationship
7.
Biochemistry ; 48(16): 3600-9, 2009 Apr 28.
Article in English | MEDLINE | ID: mdl-19249873

ABSTRACT

Abnormal expression of constitutively active anaplastic lymphoma kinase (ALK) chimeric proteins in the pathogenesis of anaplastic large-cell lymphoma (ALCL) is well established. Recent studies with small molecule kinase inhibitors have provided solid proof-of-concept validation that inhibition of ALK is sufficient to attenuate the growth and proliferation of ALK (+) ALCL cells. In this study, several missense mutants of ALK in the phosphate anchor and gatekeeper regions were generated and their kinase activity was measured. NPM-ALK L182M, L182V, and L256M mutants displayed kinase activity in cells comparable to or higher than that of NPM-ALK wild type (WT) and rendered BaF3 cells into IL-3-independent growth, while NPM-ALK L182R, L256R, L256V, L256P, and L256Q displayed much weaker or little kinase activity in cells. Similar kinase activities were obtained with corresponding GST-ALK mutants with in vitro kinase assays. With regard to inhibitor response, NPM-ALK L182M and L182V exhibited sensitivity to a fused pyrrolocarbazole (FP)-derived ALK inhibitor comparable to that of NPM-ALK WT but were dramatically less sensitive to a diaminopyrimidine (DAP)-derived ALK inhibitor. On the other hand, NPM-ALK L256M exhibited >30-fold lower sensitivity to both FP-derived and DAP-derived ALK inhibitors. The growth inhibition and cytotoxicity of BaF3/NPM-ALK mutant cells induced by ALK inhibitors were consistent with inhibition of cellular NPM-ALK autophosphorylation. In a mouse survival model, treatment with the orally bioavailable DAP-ALK inhibitor substantially extended the survival of the mice inoculated with BaF3/NPM-ALK WT cells but not those inoculated with BaF3/NPM-ALK L256M cells. Binding of ALK inhibitors to ALK WT and mutants was analyzed using ALK homology models. In summary, several potential active ALK mutants were identified, and our data indicate that some of these mutants are resistant to select small molecule ALK inhibitors. Further characterization of these mutants may help to identify and develop potent ALK inhibitors active against both WT and resistant mutants of ALK.


Subject(s)
Mutation, Missense , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Anaplastic Lymphoma Kinase , Animals , Cell Line , Humans , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/metabolism , Mice , Molecular Sequence Data , Molecular Structure , Neoplasm Transplantation , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Survival Rate
8.
J Clin Invest ; 116(12): 3171-82, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17111047

ABSTRACT

Anaplastic large cell lymphomas (ALCLs) represent a subset of lymphomas in which the anaplastic lymphoma kinase (ALK) gene is frequently fused to the nucleophosmin (NPM) gene. We previously demonstrated that the constitutive phosphorylation of ALK chimeric proteins is sufficient to induce cellular transformation in vitro and in vivo and that ALK activity is strictly required for the survival of ALK-positive ALCL cells. To elucidate the signaling pathways required for ALK-mediated transformation and tumor maintenance, we analyzed the transcriptomes of multiple ALK-positive ALCL cell lines, abrogating their ALK-mediated signaling by inducible ALK RNA interference (RNAi) or with potent and cell-permeable ALK inhibitors. Transcripts derived from the gene expression profiling (GEP) analysis uncovered a reproducible signature, which included a novel group of ALK-regulated genes. Functional RNAi screening on a set of these ALK transcriptional targets revealed that the transcription factor C/EBPbeta and the antiapoptotic protein BCL2A1 are absolutely necessary to induce cell transformation and/or to sustain the growth and survival of ALK-positive ALCL cells. Thus, we proved that an experimentally controlled and functionally validated GEP analysis represents a powerful tool to identify novel pathogenetic networks and validate biologically suitable target genes for therapeutic interventions.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , Anaplastic Lymphoma Kinase , Animals , Apoptosis/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Cycle/genetics , Cell Line , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Flow Cytometry , Gene Expression Profiling/methods , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/physiopathology , Mice , Minor Histocompatibility Antigens , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Oligonucleotide Array Sequence Analysis , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor Protein-Tyrosine Kinases , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
9.
Mol Carcinog ; 47(2): 148-56, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17849422

ABSTRACT

Tumors of the exocrine pancreas are a major cause of cancer death and have among the poorest prognosis of any malignancy. Following the "cancer stem cell hypothesis," where tumors are believed to originate in tissue specific stem cells, we screened primary ductal pancreatic carcinomas and cell lines for the expression of possible stem cell factors. We find 32/46 (70%) of primary tumors and 9/10 (90%) of cell lines express PAX6. PAX6 is a transcription factor expressed throughout the pancreatic bud during embryogenesis but not in the mature exocrine pancreas. PAX proteins have also been implicated in maintaining stem cells in a committed but undifferentiated state but a role for PAX proteins in putative pancreas stem cells is not known. We induced a pancreatic carcinoma cell line, Panc-1, to differentiate by transfecting wild-type p53 and treating the cells with differentiation agents gastrin or butyrate. This treatment induces cells to terminally differentiate into a growth-arrested cell with neurite-like processes, express the terminal differentiation marker somatostatin and downregulate PAX6. This phenotype can be replicated by directly inhibiting PAX6 expression. These data support a model where PAX proteins are aberrantly expressed in tumors and downregulation leads to differentiation.


Subject(s)
Adenocarcinoma/genetics , Cell Differentiation/genetics , Eye Proteins/genetics , Homeodomain Proteins/genetics , Paired Box Transcription Factors/genetics , Pancreatic Neoplasms/genetics , Repressor Proteins/genetics , 3T3 Cells , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Base Sequence , Cell Division , Cell Line, Tumor , DNA Primers , Mice , PAX6 Transcription Factor , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Rats , Tumor Suppressor Protein p53/metabolism
10.
Bioorg Med Chem Lett ; 18(6): 1916-21, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18308565

ABSTRACT

Fused dihydroindazolopyrrolocarbazole oximes have been identified as low nanomolar, potent dual TIE-2 and VEGF-R2 receptor tyrosine kinase inhibitors with excellent cellular potency. Development of the structure-activity relationships (SAR) led to identification of compounds 35 and 40 as potent, selective dual TIE-2/VEGF-R2 inhibitors with favorable pharmacokinetic properties. Compound 35 was orally active in tumor models with no observed toxicity.


Subject(s)
Drug Design , Melanoma, Experimental/drug therapy , Oximes/chemical synthesis , Oximes/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Receptor, TIE-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Administration, Oral , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Hemangiosarcoma/blood supply , Hemangiosarcoma/drug therapy , Hemangiosarcoma/enzymology , Humans , Melanoma, Experimental/blood , Melanoma, Experimental/blood supply , Melanoma, Experimental/enzymology , Molecular Structure , Neovascularization, Pathologic , Oximes/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Rats , Receptor, TIE-2/metabolism , Structure-Activity Relationship , Umbilical Veins , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
Biochem Pharmacol ; 73(1): 1-14, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-16904651

ABSTRACT

PAX proteins function as transcription factors and play an essential role in organogenesis during embryonic development in regulating cell proliferation and self-renewal, resistance to apoptosis, migration of embryonic precursor cells, and the coordination of specific differentiation programs. Recent studies have also discovered a role for PAX proteins in specific stem cell or progenitor cell populations, including melanocytes, muscle, and B-cells. The normal functions of the PAX proteins, including apoptosis resistance and repression of terminal differentiation, may be subverted during the progression of a number of specific malignancies. This is supported by the fact that expression of PAX proteins is dysregulated in several different types of tumors, although the precise roles for PAX proteins in cancer are not clearly understood. An emerging hypothesis is that PAX proteins play an essential role in maintaining tissue specific stem cells by inhibiting terminal differentiation and apoptosis and that these functional characteristics may facilitate the development and progression of specific cancers. In this review, we provide a general background to the PAX protein family and focus on specific cells and tissues and the role PAX proteins play within these tissues in terms of development, mature tissue maintenance, and expression in tumors. Understanding the normal developmental pathways regulated by PAX proteins may shed light on potentially parallel pathways shared in tumors, and ultimately result in defining new molecular targets and signaling pathways for the development of novel anti-cancer therapies.


Subject(s)
Embryonic Development/physiology , Neoplasms/physiopathology , Paired Box Transcription Factors/physiology , Disease Progression , Humans
12.
J Med Chem ; 59(16): 7478-96, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27527804

ABSTRACT

Analogues structurally related to anaplastic lymphoma kinase (ALK) inhibitor 1 were optimized for metabolic stability. The results from this endeavor not only led to improved metabolic stability, pharmacokinetic parameters, and in vitro activity against clinically derived resistance mutations but also led to the incorporation of activity for focal adhesion kinase (FAK). FAK activation, via amplification and/or overexpression, is characteristic of multiple invasive solid tumors and metastasis. The discovery of the clinical stage, dual FAK/ALK inhibitor 27b, including details surrounding SAR, in vitro/in vivo pharmacology, and pharmacokinetics, is reported herein.


Subject(s)
Benzamides/pharmacology , Benzocycloheptenes/pharmacology , Drug Discovery , Focal Adhesion Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Administration, Oral , Anaplastic Lymphoma Kinase , Animals , Benzamides/administration & dosage , Benzamides/chemistry , Benzocycloheptenes/administration & dosage , Benzocycloheptenes/chemistry , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Focal Adhesion Kinase 1/metabolism , Humans , Mice , Mice, Nude , Mice, SCID , Models, Molecular , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship
13.
Clin Cancer Res ; 8(1): 267-74, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11801568

ABSTRACT

Angiogenesis plays a pivotal role in tumor growth and represents a key target for chemopreventive intervention. On the basis of the structural features and lack of target organ specificity of the synthetic dithiolethione oltipraz, inhibition of angiogenesis was assessed as a potential mechanism for its broad-based chemopreventive activity. The effects of oltipraz on the development and maturation of a vascular network was determined in vitro using two-dimensional capillary tube formation assays with human umbilical vein endothelial cells plated on Matrigel and ex vivo using primary rat aortic ring explant cultures in three-dimensional collagen gels, respectively. The antiangiogenic and antitumor efficacy of oltipraz administration in vivo in nude mice was evaluated by determining its effects on neovascularization in s.c. Matrigel implants seeded with vascular endothelial growth factor and basic fibroblast growth factor-stimulated porcine aortic endothelial cells and on tumor growth and angiogenesis in SVR murine angiosarcoma xenografts implanted s.c. A dose-dependent reduction (0.4-100 microM) in microvessel formation was observed in both human and rodent bioassays after oltipraz exposure, with inhibition approaching 100% in the rat aortic ring assay at the highest concentration (P < 0.01). Similarly, oltipraz (40 microM) inhibited complete capillary tube formation by human umbilical vein endothelial cells by 62% (P < 0.05) relative to control cultures. p.o. administration of oltipraz (250 mg/kg/day for 6 days) to nude mice implanted with porcine aortic endothelial cell-Matrigel plugs resulted in a 42% reduction in neovascularization (P < 0.05) relative to vehicle-treated control mice. Administration of the same dose of oltipraz to athymic mice bearing established s.c. SVR angiosarcoma xenografts for 10 days resulted in a significant inhibition of tumor growth as early as day 4 of dosing (P < 0.005), with a maximum inhibition of tumor growth (81%, P < 0.001) relative vehicle-treated mice by day 10. The observed efficacy of oltipraz in this model is comparable with that of SU 5416 and TNP-470, known antiangiogenic agents currently under clinical development. Plasma levels of oltipraz at the termination of in vivo efficacy studies were 66.4 +/- 7 microM as determined by reversed phase high-performance liquid chromatography, a concentration range associated with significant antiangiogenic activity of oltipraz in vitro and ex vivo. These data suggest that the chemopreventive agent oltipraz may be effective in the treatment of advanced stage cancers and metastases, in part, because of its antiangiogenic activity in vivo.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Anticarcinogenic Agents/therapeutic use , Hemangiosarcoma/blood supply , Neovascularization, Pathologic/drug therapy , Pyrazines/therapeutic use , Animals , Cell Division/drug effects , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Hemangiosarcoma/pathology , Hemangiosarcoma/prevention & control , Humans , Insulin-Like Growth Factor I/metabolism , Liver/metabolism , Male , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis/methods , Rats , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 4 , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Ribonuclease, Pancreatic/metabolism , Thiones , Thiophenes , Transplantation, Heterologous , Tumor Cells, Cultured
14.
Clin Cancer Res ; 8(6): 1924-31, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12060637

ABSTRACT

PURPOSE: Aberrant expression of trk receptor kinases and enhanced expression of various neurotrophins (NTs) have been implicated in the development and progression of human prostatic carcinoma and pancreatic ductal adenocarcinoma. We examined the antitumor efficacy of administration of NT neutralizing antibodies on the growth of established human prostatic carcinoma and pancreatic ductal adenocarcinoma xenografts in nude mice. EXPERIMENTAL DESIGN: In initial studies, tumor-bearing nude mice were treated with a mixture of NT antibodies [100 microg each of anti-nerve growth factor (NGF), anti-brain-derived neurotrophic factor, anti-NT-3, and anti-NT-4/5] or normal rabbit IgG (400 microg) intratumorally and peritumorally three times/week over a 15-day dosing period. In subsequent studies, tumor-bearing nude mice were treated with individual NT antibodies (100 microg), affinity-purified anti-NGF (0.1, 1.0, or 10.0 microg), or normal rabbit IgG (100 microg) using the same dosing schedule. RESULTS: Treatment with the antibody mixture inhibited significantly the growth of TSU-Pr1 and AsPC-1 xenografts as compared with IgG-treated controls (maximal inhibition of 53 and 53%, respectively), whereas this treatment caused significant regression in PC-3 xenografts. Treatment of TSU-Pr1 xenografts with either anti-NGF or anti-NT-3 resulted in maximal tumor growth inhibition of 67 and 64%, respectively, whereas anti-brain-derived neurotrophic factor and anti-NT-4/5 did not inhibit tumor growth in this tumor model. Administration of various concentrations (0.1, 1.0, or 10.0 microg) of affinity-purified anti-NGF resulted in maximal TSU-Pr1 tumor growth inhibition of 49, 62, and 66%, respectively. CONCLUSIONS: These data add further support for the therapeutic potential of disrupting trk-signaling events in select types of nonneuronal human cancers, specifically prostatic and pancreatic carcinomas.


Subject(s)
Carcinoma, Pancreatic Ductal/prevention & control , Nerve Growth Factors/physiology , Pancreatic Neoplasms/prevention & control , Prostatic Neoplasms/prevention & control , Receptor, trkA/physiology , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Division , Disease Progression , Female , Humans , Immunoglobulin G/therapeutic use , Male , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transplantation, Heterologous
15.
J Med Chem ; 58(3): 1140-58, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25581261

ABSTRACT

The spread of intra-abdominal cancers is a vexing clinical problem for which there is no widely effective treatment. We discovered previously that (2E)-3-[(4-tert-butylphenyl)sulfonyl]acrylonitrile (1) induced cancer cell apoptosis during adhesion to normal mesothelial cells which line the peritoneum. We recently demonstrated that the sulfonylacrylonitrile portion of 1 and hydrophobic aryl substitution were essential for pro-apoptotic activity in cancer cells. Here we synthesized a diverse series of analogues of 1 in order to improve the efficacy and pharmaceutical properties. Analogues and 1 were compared in their ability to cause cancer cell death during adhesion to normal mesothelial cell monolayers. Potent analogues identified in the in vitro assay were validated and found to exhibit improved inhibition of intra-abdominal cancer in two clinically relevant murine models of ovarian and pancreatic cancer spread and metastasis, highlighting their potential clinical use as an adjunct to surgical resection of cancers.


Subject(s)
Acrylonitrile/pharmacology , Antineoplastic Agents/pharmacology , Drug Design , Ovarian Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Sulfones/pharmacology , Acrylonitrile/chemical synthesis , Acrylonitrile/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , HT29 Cells , Humans , Mice , Molecular Structure , Ovarian Neoplasms/pathology , Ovarian Neoplasms/secondary , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/secondary , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/chemistry
16.
J Med Chem ; 46(25): 5375-88, 2003 Dec 04.
Article in English | MEDLINE | ID: mdl-14640546

ABSTRACT

A series of potent vascular endothelial growth factor R2 (VEGF-R2) tyrosine kinase inhibitors from a new indenopyrrolocarbazole template is reported. The structure-activity relationships for a series of 9-alkoxymethyl-12-(3-hydroxypropyl)indeno[2,1-a]pyrrolo[3,4-c]carbazole-5-ones revealed an optimal R9 substitution with ethoxymethyl 19 (VEGF-R2 IC(50) = 4 nM) and isopropoxymethyl 21 (VEGF-R2 IC(50) = 8 nM) being the most potent inhibitors in the series. The VEGF-R2 activity was reduced appreciably by increasing the size of the R9 alkoxy group or by alpha-methyl branching adjacent to the ring. The combined R9 alkoxymethyl and N12 hydroxypropyl substitutions were required for potent VEGF-R2 activity, and the corresponding thioether analogues were weaker than their ether counterparts. Compound 21 (R9 isopropoxymethyl, CEP-5214) was identified as a potent, low-nanomolar pan inhibitor of human VEGF-R tyrosine kinases, displaying IC(50) values of 16, 8, and 4 nM for VEGF-R1/FLT-1, VEGF-R2/KDR, and VEGF-R3/FLT-4, respectively, with cellular activity equivalent to the isolated enzyme activity. Compound 21 exhibited good selectivity against numerous tyrosine and serine/threonine kinases including PKC, Tie2, TrkA, CDK1, p38, JNK, and IRK. To increase water solubility and oral bioavailability, the N,N-dimethylglycine ester 40 was prepared. In pharmacokinetic studies in mice and rats, increased plasma levels of 21 were observed after oral administration of 40. Compound 21 demonstrated significant in vivo antitumor activity in numerous tumor models and was advanced into phase I clinical trials as the water-soluble N,N-dimethylglycine ester prodrug 40 (CEP-7055).


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Carbazoles/chemical synthesis , Prodrugs/chemical synthesis , Sarcosine/analogs & derivatives , Sarcosine/chemistry , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Carbazoles/chemistry , Carbazoles/pharmacology , Cells, Cultured , Drug Screening Assays, Antitumor , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Enzyme-Linked Immunosorbent Assay , Female , Hemangiosarcoma/drug therapy , Humans , In Vitro Techniques , Indenes/chemical synthesis , Indenes/chemistry , Indenes/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Nude , Models, Molecular , Phosphorylation , Prodrugs/chemistry , Prodrugs/pharmacology , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Solubility , Structure-Activity Relationship
17.
Anticancer Res ; 22(6A): 3127-34, 2002.
Article in English | MEDLINE | ID: mdl-12530055

ABSTRACT

Perillyl alcohol, farnesol and geraniol have chemotherapeutic activity toward pancreatic and other cancers. Perillyl alcohol induces apoptosis and increases expression of the proapoptotic protein Bak in cultured pancreatic tumor cells. We tested the hypothesis that farnesol and geraniol would have similar effects. After 48 hours of treatment with farnesol geraniol or perillyl alcohol, human BxPC3 pancreatic cancer cells exhibited a 3 to 10-fold increase in apoptosis and higher Bak expression than the controls. We then tested the hypotheses that perillyl alcohol and farnesol would have chemopreventive activity toward pancreatic cancer and would increase Bak expression and apoptosis in vivo. Hamster pancreatic cancer was initiated at time 0 with N-nitrosobis(2-oxopropyl)amine. Animals were fed control, 2% (w/w) perillyl alcohol, or 1% (w/w) farnesol diets from weeks 5-42. Pancreatic carcinoma incidence was decreased by perillyl alcohol and farnesol. Hyperplastic pancreatic ductal neoplasms from perillyl alcohol and farnesol-treated animals had higher Bak protein expression (p < 0.05), and somewhat higher apoptotic rates, diminished expression of the antiapoptotic protein BCL-XL, and lower rates of DNA synthesis than the controls.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/biosynthesis , Carcinoma, Pancreatic Ductal/prevention & control , Farnesol/pharmacology , Membrane Proteins/biosynthesis , Monoterpenes/pharmacology , Pancreatic Neoplasms/prevention & control , Animals , Carcinogens , Carcinoma, Pancreatic Ductal/chemically induced , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cricetinae , Humans , Male , Mesocricetus , Nitrosamines , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured , bcl-2 Homologous Antagonist-Killer Protein
18.
Biochem Pharmacol ; 87(1): 150-61, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23817077

ABSTRACT

Preclinical models of human cancers are indispensable in the drug discovery and development process for new cancer drugs, small molecules and biologics. They are however imperfect facsimiles of human cancers given the genetic and epigenetic heterogeneity of the latter and the multiplicity of dysregulated survival and growth-regulatory pathways that characterize this spectrum of diseases. This review discusses pre-clinical tumor models - traditional ectopic xenografts, orthotopic xenografts, genetically engineered tumor models, primary human tumorgrafts, and various multi-stage carcinogen-induced tumor models - their advantages, limitations, physiological and pathological relevance. Collectively, these animal models represent a portfolio of test systems that should be utilized at specific stages in the drug discovery process in a pragmatic and hierarchical manner of increasing complexity, physiological relevance, and clinical predictability of the human response. Additionally, evaluating the efficacy of novel therapeutic agents emerging from drug discovery programs in a variety of pre-clinical models can better mimic the heterogeneity of human cancers and also aid in establishing dose levels, dose regimens and drug combinations for use in clinical trials. Nonetheless, despite the sophistication and physiological relevance of these human cancer models (e.g., genetically engineered tumor models and primary human tumografts), the ultimate proof of concept for efficacy and safety of novel oncology therapeutics lies in humans. The judicious interpretation and extrapolation of data derived from these models to humans, and a correspondingly greater emphasis placed on translational medical research in early stage clinical trials, are essential to improve on the current clinical attrition rates for novel oncology therapeutic agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Disease Models, Animal , Drug Discovery/methods , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Evaluation, Preclinical/methods , Humans , Neoplasms/pathology , Translational Research, Biomedical , Xenograft Model Antitumor Assays/methods
19.
Int Immunopharmacol ; 12(1): 257-70, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22178195

ABSTRACT

Current therapies for late-stage systemic lupus erythematosus (SLE) are limited to cytotoxic agents. Delanzomib (CEP-18770) is an orally active, reversible P2 threonine boronic acid inhibitor of the 26S mammalian proteasome. Delanzomib was tested in a head-to-head comparison against bortezomib to protect and treat mice with fatal lupus nephritis (LN). Age matched MRL/lpr or NZBWF1 mice with established SLE or LN, respectively, were treated with delanzomib either 3 mg/kg once or twice weekly intravenously or orally at 10 mg/kg. Mice were also treated with reference agent bortezomib at 0.5 mg/kg, intraperitoneally, once a week or 0.3 mg/kg once or twice a week. Reductions in the frequencies of specific anti-chromatin, smith and dsDNA antibody secreting cells and levels of the corresponding circulating antinuclear antibodies, were observed following delanzomib treatment. Reductions in several serum pro-inflammatory cytokines were observed in delanzomib-treated animals. Delanzomib treatment suppressed the development and progression of renal tissue damage and extended the survival of ill mice. Proteinuria was significantly decreased and severity of various renal histopathologies reduced relative to vehicle-treated nephritic mice. Treatment of lupus in these models demonstrated that delanzomib treatment lead to greater tolerability and rate of response resulting in improved stabilization of disease.


Subject(s)
Boronic Acids/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Lupus Nephritis/drug therapy , Proteasome Inhibitors , Threonine/analogs & derivatives , Animals , Antibodies, Antinuclear/blood , Boronic Acids/administration & dosage , Cytokines/blood , Disease Models, Animal , Drug Administration Routes , Female , Kidney/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Lupus Nephritis/blood , Lupus Nephritis/metabolism , Mice , Proteasome Endopeptidase Complex/metabolism , Spleen/metabolism , Threonine/administration & dosage , Threonine/therapeutic use
20.
Mol Cancer Ther ; 11(4): 984-93, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22334590

ABSTRACT

Constitutively activated STAT3 and STAT5 are expressed in a wide variety of human malignancies including solid and hematopoietic cancers and often correlate with a poor prognosis and resistance to multiple therapies. Given the well established role of STAT3 in tumorigenesis, inhibition of Janus-activated kinase 2 (JAK2) activity might represent an attractive therapeutic approach. Using a mouse model of colitis-induced colorectal cancer, we show that a novel, orally active, selective JAK2 inhibitor, CEP-33779, induced regression of established colorectal tumors, reduced angiogenesis, and reduced proliferation of tumor cells. Histopathologic analysis confirmed reduced incidence of histologic-grade neoplasia by CEP-33779. Tumor regression correlated with inhibition of STAT3 and NF-κB (RelA/p65) activation in a CEP-33779 dose-dependent manner. In addition, the expression of proinflammatory, tumor-promoting cytokines interleukin (IL)-6 and IL-1ß was strongly reduced upon JAK2 inhibition. The ability of CEP-33779 to suppress growth of colorectal tumors by inhibiting the IL-6/JAK2/STAT3 signaling suggests a potential therapeutic utility of JAK2 inhibitors in multiple tumors types, particularly those with a strong inflammatory component.


Subject(s)
Colitis/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Triazoles/pharmacology , Animals , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Disease Models, Animal , Female , Humans , Janus Kinase 2/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation , Protein Kinase Inhibitors/pharmacokinetics , Pyridines/pharmacokinetics , Triazoles/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL