Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
ACS Med Chem Lett ; 12(2): 288-294, 2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33603977

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) is a heme-containing dioxygenase enzyme implicated in cancer immune response. This account details the discovery of BMS-986242, a novel IDO1 inhibitor designed for the treatment of a variety of cancers including metastatic melanoma and renal cell carcinoma. Given the substantial interest around this target for cancer immunotherapy, we sought to identify a structurally differentiated clinical candidate that performs comparably to linrodostat (BMS-986205) in terms of both in vitro potency and in vivo pharmacodynamic effect in a mouse xenograft model. On the basis of its preclinical profile, BMS-986242 was selected as a candidate for clinical development.

2.
ACS Med Chem Lett ; 11(2): 172-178, 2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32071685

ABSTRACT

Novel imidazole-based TGFßR1 inhibitors were identified and optimized for potency, selectivity, and pharmacokinetic and physicochemical characteristics. Herein, we report the discovery, optimization, and evaluation of a potent, selective, and orally bioavailable TGFßR1 inhibitor, 10 (BMS-986260). This compound demonstrated functional activity in multiple TGFß-dependent cellular assays, excellent kinome selectivity, favorable pharmacokinetic properties, and curative in vivo efficacy in combination with anti-PD-1 antibody in murine colorectal cancer (CRC) models. Since daily dosing of TGFßR1 inhibitors is known to cause class-based cardiovascular (CV) toxicities in preclinical species, a dosing holiday schedule in the anti-PD-1 combination efficacy studies was explored. An intermittent dosing regimen of 3 days on and 4 days off allowed mitigation of CV toxicities in one month dog and rat toxicology studies and also provided similar efficacy as once daily dosing.

3.
ACS Med Chem Lett ; 9(11): 1117-1122, 2018 Nov 08.
Article in English | MEDLINE | ID: mdl-30429955

ABSTRACT

The multifunctional cytokine TGFß plays a central role in regulating antitumor immunity. It has been postulated that inhibition of TGFß signaling in concert with checkpoint blockade will provide improved and durable immune response against tumors. Herein, we describe a novel series of 4-azaindole TGFß receptor kinase inhibitors with excellent selectivity for TGFß receptor 1 kinase. The combination of compound 3f and an antimouse-PD-1 antibody demonstrated significantly improved antitumor efficacy compared to either treatment alone in a murine tumor model.

4.
Bioorg Med Chem ; 26(5): 1026-1034, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29422332

ABSTRACT

The TGFß-TGFßR signaling pathway has been reported to play a protective role in the later stages of tumorigenesis via increasing immunosuppressive Treg cells and facilitating the epithelial to mesenchymal transition (EMT). Therefore, inhibition of TGFßR has the potential to enhance antitumor immunity. Herein we disclose the identification and optimization of novel heterobicyclic inhibitors of TGFßRI that demonstrate potent inhibition of SMAD phosphorylation. Application of structure-based drug design to the novel pyrrolotriazine chemotype resulted in improved binding affinity (Ki apparent = 0.14 nM), long residence time (T1/2 > 120 min) and significantly improved potency in the PSMAD cellular assay (IC50 = 24 nM). Several analogs inhibited phosphorylation of SMAD both in vitro and in vivo. Additionally, inhibition of TGFß-stimulated phospho-SMAD was observed in primary human T cells.


Subject(s)
Bridged Bicyclo Compounds/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Binding Sites , Bridged Bicyclo Compounds/chemical synthesis , Bridged Bicyclo Compounds/pharmacology , Cells, Cultured , Crystallography, X-Ray , Drug Design , Epithelial-Mesenchymal Transition/drug effects , Humans , Molecular Dynamics Simulation , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrroles/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Smad Proteins/metabolism , Structure-Activity Relationship , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thiazines/chemical synthesis , Thiazines/chemistry , Thiazines/metabolism
5.
ACS Cent Sci ; 2(1): 27-31, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-27163023

ABSTRACT

The fungal-derived Taiwanese natural product antroquinonol A has attracted both academic and commercial interest due to its reported exciting biological properties. This reduced quinone is currently in phase II trials (USA and Taiwan) for the treatment of non-small-cell lung carcinoma (NSCLC) and was recently granted orphan drug status by the FDA for the treatment of pancreatic cancer and acute myeloid leukemia. Pending successful completion of human clinical trials, antroquinonol is expected to be commercialized under the trade name Hocena. A synthesis-enabled biological re-examination of this promising natural product, however, reveals minimal in vitro and in vivo antitumor activity in preclinical models.

6.
Exp Cell Res ; 338(2): 251-60, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26302264

ABSTRACT

Cellular levels of inhibitor of apoptosis (IAP) proteins are elevated in multiple human cancers and their activities often play a part in promoting cancer cell survival by blocking apoptotic pathways, controlling signal transduction pathways and contributing to resistance. These proteins function through interactions of their BIR (baculoviral IAP repeat) protein domains with pathway components and these interactions are endogenously antagonized by Smac/Diablo (second mitochondrial activator of caspases/direct IAP binding protein with low isoelectric point). This report describes development of synthetic smac mimetics (SM) and compares their binding, antiproliferative and anti-tumor activities. All dimeric antagonists inhibit in vitro smac tetrapeptide binding to recombinant IAP proteins, rescue IAP-bound caspase-3 activity and show anti-proliferative activity against human A875 melanoma cells. One heterodimeric SM, SM3, binds tightly to IAP proteins in vitro and slowly dissociates (greater than two hours) from these protein complexes compared to the other antagonists. In addition, in vitro SM anti-proliferation potency is influenced by ABCB1 transporter (ATP-binding cassette, sub-family B; MDR1, P-gp) activities and one antagonist, SM5, does not appear to be an ABCB1 efflux pump substrate. All dimeric smac mimetics inhibit the growth of human melanoma A875 tumors implanted in athymic mice at well-tolerated doses. One antagonist, SM4, shows broad spectrum in vivo anti-tumor activity and modulates known pharmacodynamic markers of IAP antagonism. These data taken together demonstrate the range of diverse dimeric IAP antagonist activities and supports their potential as anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Caspase 3/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Mitochondrial Proteins/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Biomimetics/methods , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , HCT116 Cells , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Melanoma/drug therapy , Melanoma/metabolism , Mice, Inbred BALB C , Mice, Nude , Protein Binding/drug effects , Protein Structure, Tertiary/drug effects
7.
ACS Med Chem Lett ; 6(7): 770-5, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26191364

ABSTRACT

A series of dimeric macrocyclic compounds were prepared and evaluated as antagonists for inhibitor of apoptosis proteins. The most potent analogue 11, which binds to XIAP and c-IAP proteins with high affinity and induces caspase-3 activation and ultimately cell apoptosis, inhibits growth of human melanoma and colorectal cell lines at low nanomolar concentrations. Furthermore, compound 11 demonstrated significant antitumor activity in the A875 human melanoma xenograft model at doses as low as 2 mg/kg on a q3d schedule.

8.
J Med Chem ; 58(6): 2855-61, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25695766

ABSTRACT

Affinity selection screening of macrocycle libraries derived from DNA-programmed chemistry identified XIAP BIR2 and BIR3 domain inhibitors that displace bound pro-apoptotic caspases. X-ray cocrystal structures of key compounds with XIAP BIR2 suggested potency-enhancing structural modifications. Optimization of dimeric macrocycles with similar affinity for both domains were potent pro-apoptotic agents in cancer cell lines and efficacious in shrinking tumors in a mouse xenograft model.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/therapeutic use , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 3/metabolism , Cell Line, Tumor , Crystallography, X-Ray , Drug Discovery , Female , Gene Library , Humans , Macrocyclic Compounds/pharmacokinetics , Mice , Models, Molecular , X-Linked Inhibitor of Apoptosis Protein/metabolism
9.
J Med Chem ; 58(3): 1556-62, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25584393

ABSTRACT

The prominent role of IAPs in controlling cell death and their overexpression in a variety of cancers has prompted the development of IAP antagonists as potential antitumor therapies. We describe the identification of a series of heterodimeric antagonists with highly potent antiproliferative activities in cIAP- and XIAP-dependent cell lines. Compounds 15 and 17 further demonstrate curative efficacy in human melanoma and lung cancer xenograft models and are promising candidates for advanced studies.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Proline/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mice , Molecular Structure , Neoplasms, Experimental/pathology , Proline/chemical synthesis , Proline/chemistry , Structure-Activity Relationship
10.
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
11.
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
12.
Cancer Res ; 70(18): 7221-31, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20807811

ABSTRACT

Agents targeting insulin-like growth factor-I receptor (IGF-IR), including antibodies and small-molecule inhibitors, are currently in clinical development for the treatment of cancers including sarcoma. However, development of resistance is a common phenomenon resulting in failures of anticancer therapies. In light of this problem, we developed two resistant models from the rhabdomyosarcoma cell line Rh41: Rh41-807R, with acquired resistance to BMS-754807, a small-molecule dual-kinase inhibitor targeting IGF-IR and insulin receptor (IR), and Rh41-MAB391R, with resistance to MAB391, an IGF-IR-blocking antibody. In addition, tumor xenograft models were established from Rh41 and Rh41-807R cell lines. Gene expression and DNA copy number analyses of these models revealed shared as well as unique acquired resistance mechanisms for the two types of IGF-IR inhibitors. Each resistant model used different signaling pathways as a mechanism for proliferation. Platelet-derived growth factor receptor α (PDGFRα) was amplified, overexpressed, and constitutively activated in Rh41-807R cells and tumors. Knockdown of PDGFRα by small interfering RNA in Rh41-807R resensitized the cells to BMS-754807. Synergistic activities were observed when BMS-754807 was combined with PDGFRα inhibitors in the Rh41-807R model in vitro. In contrast, AXL expression was highly elevated in Rh41-MAB391R but downregulated in Rh41-807R. Notably, BMS-754807 was active in Rh41-MAB391R cells and able to overcome resistance to MAB391, but MAB391 was not active in Rh41-807R cells, suggesting potentially broader clinical activity of BMS-754807. This is the first study to define and compare acquired resistance mechanisms for IGF-IR-targeted therapies. It provides insights into the differential acquired resistance mechanisms for IGF-IR/IR small-molecule inhibitor versus anti-IGF-IR antibody.


Subject(s)
Antibodies, Monoclonal/pharmacology , Pyrazoles/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Rhabdomyosarcoma/therapy , Triazines/pharmacology , Animals , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Dosage , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Receptor, IGF Type 1/biosynthesis , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/immunology , Receptor, Platelet-Derived Growth Factor alpha/agonists , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , Receptor, Platelet-Derived Growth Factor alpha/deficiency , Receptor, Platelet-Derived Growth Factor alpha/genetics , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/immunology , Transfection , Xenograft Model Antitumor Assays
13.
Bioorg Med Chem Lett ; 20(9): 2998-3002, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20382527

ABSTRACT

Biarylamine-based inhibitors of Met kinase have been identified. Lead compounds demonstrate nanomolar potency in Met kinase biochemical assays and significant activity in the Met-driven GTL-16 human gastric carcinoma cell line. X-ray crystallography revealed that these compounds adopt a bioactive conformation, in the kinase domain, consistent with that previously seen with 2-pyridone-based Met kinase inhibitors. Compound 9b demonstrated potent in vivo antitumor activity in the GTL-16 human tumor xenograft model.


Subject(s)
Amines/chemistry , Aminopyridines/chemical synthesis , Antineoplastic Agents/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Aminopyridines/chemistry , Aminopyridines/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Humans , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
14.
Eur J Cancer ; 46(9): 1537-53, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20303261

ABSTRACT

PURPOSE: Tamoxifen, a selective oestrogen receptor modulator (SERM), and brivanib alaninate, a vascular endothelial growth factor receptor 2 (VEGFR-2) inhibitor, are two target specific agents that result in a substantial decrease in tumour growth when given alone. Tamoxifen activates SERM stimulated breast and endometrial tumour growth. Tamoxifen and brivanib alaninate have side-effects that can affect therapeutic outcomes. The primary goal of the current study was to evaluate the therapeutic effects of lower doses of both agents when given in combination to mice with SERM sensitive, oestrogen stimulated tumour xenografts (MCF-7 E2 tumours). Experiments were conducted to evaluate the response of SERM stimulated breast (MCF-7 Tam, MCF-7 Ral) and endometrial tumours (EnCa 101) to demonstrate the activity of brivanib alaninate in SERM resistant models. EXPERIMENTAL DESIGN: In the current study, tumour xenografts were minced and bi-transplanted into the mammary fat pads of athymic, ovariectomised mice. Preliminary experiments were conducted to determine an effective oral dose of tamoxifen and brivanib alaninate that had minimal effect on tumour growth. Doses of 125 microg of tamoxifen and 0.05 mg/g of brivanib alaninate were evaluated. An experiment was designed to evaluate the effect of the two agents together when started at the time of tumour implantation. An additional experiment was done in which tumours were already established and then treated, to obtain enough tumour tissue for molecular analysis. RESULTS: Brivanib alaninate was effective at inhibiting tumour growth in SERM sensitive (MCF-7 E2) and SERM stimulated (EnCa 101, MCF-7 Ral, MCF-7 Tam) models. The effect of the low dose drug combination as an anti-tumour strategy for SERM sensitive (MCF-7 E2) in early treatment was as effective as higher doses of either drug used alone. In established tumours, the combination is successful at decreasing tumour growth, while neither agent alone is effective. Molecular analysis revealed a decreased phosphorylation of VEGFR-2 in tumours that were treated with brivanib alaninate and an increase in VEGFA transcription to compensate for the blockade of VEGFR-2 by increasing the transcription of VEGFA. Tamoxifen increases the phosphorylation of VEGFR-2 and this effect is abrogated by brivanib alaninate. There was also increased necrosis in tumours treated with brivanib alaninate. CONCLUSION: Historically, tamoxifen has a role in blocking angiogenesis as well as the blockade of the ER. Tamoxifen and a low dose of an angiogenesis inhibitor, brivanib alaninate, can potentially be combined not only to maximise therapeutic efficacy but also to retard SERM resistant tumour growth.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Alanine/analogs & derivatives , Animals , Dose-Response Relationship, Drug , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Transplantation , Pyrroles/administration & dosage , Random Allocation , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/administration & dosage , Transplantation, Heterologous , Triazines/administration & dosage , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Mol Cancer Ther ; 9(2): 369-78, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20103604

ABSTRACT

Tumor angiogenesis is a complex and tightly regulated network mediated by various proangiogenic factors. The fibroblast growth factor (FGF) and vascular endothelial growth factor (VEGF) family of growth factors, and associated tyrosine kinase receptors have a major influence in tumor growth and dissemination and may work synergistically to promote angiogenesis. Brivanib alaninate is the orally active prodrug of brivanib, a selective dual inhibitor of FGF and VEGF signaling. Here, we show that brivanib demonstrates antitumor activity in a broad range of xenograft models over multiple dose levels and that brivanib alaninate shows dose-dependent efficacy equivalent to brivanib in L2987 human tumor xenografts. Brivanib alaninate (107 mg/kg) reduced tumor cell proliferation as determined by a 76% reduction in Ki-67 staining and reduced tumor vascular density as determined by a 76% reduction in anti-CD34 endothelial cell staining. Furthermore, Matrigel plug assays in athymic mice showed that brivanib alaninate inhibited angiogenesis driven by VEGF or basic FGF alone, or combined. Dynamic contrast-enhanced magnetic resonance imaging, used to assess the effects of brivanib alaninate on tumor microcirculation, showed a marked decrease in gadopentetate dimeglumine contrast agent uptake at 107 mg/kg dose, with a reduction in area under the plasma concentration-time curve from time 0 to 60 minutes at 24 and 48 hours of 54% and 64%, respectively. These results show that brivanib alaninate is an effective antitumor agent in preclinical models across a range of doses, and that efficacy is accompanied by changes in cellular and vascular activities.


Subject(s)
Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Triazines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Alanine/analogs & derivatives , Animals , Antigens, CD34/biosynthesis , Cell Line, Tumor , Collagen/chemistry , Dose-Response Relationship, Drug , Drug Combinations , Female , Humans , Laminin/chemistry , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Proteoglycans/chemistry , Signal Transduction , Time Factors
16.
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
17.
Cancer Chemother Pharmacol ; 65(1): 55-66, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19396600

ABSTRACT

PURPOSE: Brivanib alaninate is a prodrug of brivanib (BMS-540215), a potent oral VEGFR-2 inhibitor and is currently in development for the treatment of hepatocellular and colon carcinomas. In vitro and in vivo studies were conducted to characterize the preclinical pharmacokinetics and disposition of brivanib and brivanib alaninate, and antitumor efficacy in mice bearing human xenografts. METHODS: In vitro studies were conducted in liver and intestinal fractions, plasma and Caco-2 cells to assess the metabolic stability. Pharmacokinetics of brivanib were determined in preclinical species after administration of single intravenous or oral doses of both brivanib and brivanib alaninate. The antitumor efficacy was assessed at equimolar doses in nude mice bearing human tumor xenografts. Human efficacious dose was predicted based on projected human pharmacokinetic parameters and exposure at efficacious doses in the mouse efficacy models. RESULTS: In vitro and in vivo studies indicated that brivanib alaninate was efficiently converted to brivanib. Brivanib showed good brain penetration in rats consistent with its high intrinsic permeability and lack of active efflux in Caco-2 cells. The oral bioavailability of brivanib varied among species (22-88%) and showed dissolution rate-limited absorption even when combined with organic co-solvents. Administration of brivanib as brivanib alaninate allowed completely aqueous vehicles, and an improvement in the oral bioavailability (55-97%) was observed. The clearance of brivanib in humans is anticipated to be low to intermediate (hepatic extraction ratio < 0.7), while its volume of distribution is expected to be high. The minimum efficacious dose of brivanib alaninate was determined to be 60 mg/kg per day. CONCLUSIONS: Brivanib alaninate is rapidly and efficiently converted to the parent, brivanib, as demonstrated both in vitro and in vivo and offers an excellent mode to deliver brivanib orally.


Subject(s)
Alanine/analogs & derivatives , Antineoplastic Agents/pharmacokinetics , Pyrroles/pharmacokinetics , Triazines/pharmacokinetics , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Administration, Oral , Alanine/pharmacokinetics , Alanine/pharmacology , Animals , Antineoplastic Agents/pharmacology , Biological Availability , Brain/metabolism , Caco-2 Cells , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Injections, Intravenous , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prodrugs , Pyrroles/pharmacology , Rats , Solubility , Tissue Distribution , Triazines/pharmacology , Xenograft Model Antitumor Assays
18.
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
19.
Clin Cancer Res ; 14(24): 8123-31, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19088027

ABSTRACT

PURPOSE: Angiogenesis is a critical step in the establishment, growth, and metastasis of solid tumors, and combination of antiangiogenic agents with chemotherapy is an attractive therapeutic option. We investigated the potential of ixabepilone, the first in a new class of antineoplastic agents known as epothilones, to synergize with antiangiogenic agents to inhibit tumor growth. EXPERIMENTAL DESIGN: In vitro and in vivo cytotoxicity of ixabepilone as single agent and in combination with two targeted antiangiogenic agents, bevacizumab or sunitinib, were examined in preclinical tumor models. Direct effects of the agents against endothelial cells was also examined and compared with the effects of paclitaxel as single agent and in combination with bevacizumab. RESULTS: Ixabepilone showed robust synergistic antitumor activity in combination with bevacizumab and sunitinib in preclinical in vivo models derived from breast, colon, lung, and kidney cancers. The synergistic antitumor effect was greater with ixabepilone compared with paclitaxel. Furthermore, ixabepilone was more effective than paclitaxel at killing endothelial cells expressing P-glycoprotein in vitro and inhibiting endothelial cell proliferation and tumor angiogenesis in vivo. CONCLUSIONS: Ixabepilone may enhance the antitumor effects of antiangiogenic therapy by direct cytotoxicity and also indirectly via the killing of tumor-associated endothelial cells. Given that ixabepilone has reduced susceptibility to drug efflux pumps compared with taxanes, these data may explain the increased antiangiogenic and antitumor activity of ixabepilone in combination with antiangiogenic agents. Phase II studies to assess the efficacy and safety of ixabepilone plus bevacizumab in locally recurrent or metastatic breast cancer are planned.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Epothilones/administration & dosage , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Bevacizumab , Drug Synergism , Endothelial Cells/drug effects , Epothilones/pharmacology , Female , Humans , Indoles/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Pyrroles/administration & dosage , Sunitinib , Xenograft Model Antitumor Assays
20.
Clin Cancer Res ; 14(19): 6146-53, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829493

ABSTRACT

PURPOSE: Hepatocellular carcinoma (HCC) is the fifth most common primary neoplasm; surgery is the only curative option but 5-year survival rates are only 25% to 50%. Vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) are known to be involved in growth and neovascularization of HCC. Therefore, agents that target these pathways may be effective in the treatment of HCC. The aim of this study was to determine the antineoplastic activity of brivanib alaninate, a dual inhibitor of VEGF receptor (VEGFR) and FGF receptor (FGFR) signaling pathways. EXPERIMENTAL DESIGN: Six different s.c. patient-derived HCC xenografts were implanted into mice. Tumor growth was evaluated in mice treated with brivanib compared with control. The effects of brivanib on apoptosis and cell proliferation were evaluated by immunohistochemistry. The SK-HEP1 and HepG2 cells were used to investigate the effects of brivanib on the VEGFR-2 and FGFR-1 signaling pathways in vitro. Western blotting was used to determine changes in proteins in these xenografts and cell lines. RESULTS: Brivanib significantly suppressed tumor growth in five of six xenograft lines. Furthermore, brivanib-induced growth inhibition was associated with a decrease in phosphorylated VEGFR-2 at Tyr(1054/1059), increased apoptosis, reduced microvessel density, inhibition of cell proliferation, and down-regulation of cell cycle regulators. The levels of FGFR-1 and FGFR-2 expression in these xenograft lines were positively correlated with its sensitivity to brivanib-induced growth inhibition. In VEGF-stimulated and basic FGF stimulated SK-HEP1 cells, brivanib significantly inhibited VEGFR-2, FGFR-1, extracellular signal-regulated kinase 1/2, and Akt phosphorylation. CONCLUSION: This study provides a strong rationale for clinical investigation of brivanib in patients with HCC.


Subject(s)
Alanine/analogs & derivatives , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Triazines/pharmacology , Alanine/pharmacology , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Male , Mice , Models, Chemical , Neoplasm Transplantation , Neovascularization, Pathologic , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...