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










Publication year range
1.
Cancer Res ; 83(11): 1883-1904, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37074042

ABSTRACT

The EGFR and TGFß signaling pathways are important mediators of tumorigenesis, and cross-talk between them contributes to cancer progression and drug resistance. Therapies capable of simultaneously targeting EGFR and TGFß could help improve patient outcomes across various cancer types. Here, we developed BCA101, an anti-EGFR IgG1 mAb linked to an extracellular domain of human TGFßRII. The TGFß "trap" fused to the light chain in BCA101 did not sterically interfere with its ability to bind EGFR, inhibit cell proliferation, or mediate antibody-dependent cellular cytotoxicity. Functional neutralization of TGFß by BCA101 was demonstrated by several in vitro assays. BCA101 increased production of proinflammatory cytokines and key markers associated with T-cell and natural killer-cell activation, while suppressing VEGF secretion. In addition, BCA101 inhibited differentiation of naïve CD4+ T cells to inducible regulatory T cells (iTreg) more strongly than the anti-EGFR antibody cetuximab. BCA101 localized to tumor tissues in xenograft mouse models with comparable kinetics to cetuximab, both having better tumor tissue retention over TGFß "trap." TGFß in tumors was neutralized by approximately 90% in animals dosed with 10 mg/kg of BCA101 compared with 54% in animals dosed with equimolar TGFßRII-Fc. In patient-derived xenograft mouse models of head and neck squamous cell carcinoma, BCA101 showed durable response after dose cessation. The combination of BCA101 and anti-PD1 antibody improved tumor inhibition in both B16-hEGFR-expressing syngeneic mouse models and in humanized HuNOG-EXL mice bearing human PC-3 xenografts. Together, these results support the clinical development of BCA101 as a monotherapy and in combination with immune checkpoint therapy. SIGNIFICANCE: The bifunctional mAb fusion design of BCA101 targets it to the tumor microenvironment where it inhibits EGFR and neutralizes TGFß to induce immune activation and to suppress tumor growth.


Subject(s)
Antibodies, Monoclonal, Humanized , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Neoplasms , Animals , Humans , Mice , Antibodies, Monoclonal, Humanized/therapeutic use , Carcinoma, Squamous Cell/therapy , Cell Line, Tumor , Cetuximab/pharmacology , Cetuximab/therapeutic use , ErbB Receptors/metabolism , Head and Neck Neoplasms/therapy , Transforming Growth Factor beta , Tumor Microenvironment , Xenograft Model Antitumor Assays , Neoplasms/therapy
2.
Mol Cancer Ther ; 14(5): 1095-106, 2015 May.
Article in English | MEDLINE | ID: mdl-25700704

ABSTRACT

The mTOR pathway is often upregulated in cancer and thus intensively pursued as a target to design novel anticancer therapies. Approved and emerging drugs targeting the mTOR pathway have positively affected the clinical landscape. Recently, activin receptor-like kinase 1 (ALK1), belonging to the TGFß receptor family, has been reported as an emerging target for antiangiogenic cancer therapy. Here, we describe a novel orally efficacious compound, P7170, that inhibits mTORC1/mTORC2/ALK1 activity with a potent cell growth inhibition. In cell-based assays, P7170 strongly inhibited (IC50 < 10 nmol/L) the phosphorylation of p70S6K (T389) and pAKT (S473). In many cancer cell lines, such as prostate, ovarian, colon, and renal, P7170 treatment resulted in marked cell growth inhibition. Furthermore, it induced G1-S cell-cycle arrest and autophagy. In vitro HUVEC tube formation, in vivo Matrigel plug, and rat aorta ring assays demonstrated that P7170 exhibited significant antiangiogenic activity. In addition, ALK1 knockdown studies in HUVEC confirmed that the antiangiogenic activity of P7170 was primarily due to ALK1 inhibition. Strong inhibition of ALK1 in addition to mTORC1/mTORC2 differentiates P7170 in its mechanism of action in comparison with existing inhibitors. In vivo mouse xenograft studies revealed P7170 to exhibit a significant dose-dependent tumor growth inhibition in a broad range of human tumor types when administered orally at 10 to 20 mg/kg doses. The distinctive pharmacological profile with favorable pharmacokinetic parameters and in vivo efficacy makes P7170 an attractive candidate for clinical development. It is currently being tested in phase I clinical studies.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents/administration & dosage , Imidazoles/administration & dosage , Prostatic Neoplasms/drug therapy , Quinolines/administration & dosage , Activin Receptors, Type II/antagonists & inhibitors , Administration, Oral , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Hep G2 Cells , Human Umbilical Vein Endothelial Cells , Humans , Imidazoles/pharmacology , Male , Mice , Prostatic Neoplasms/metabolism , Quinolines/pharmacology , Rats , TOR Serine-Threonine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
3.
Expert Opin Biol Ther ; 14(8): 1145-59, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24762115

ABSTRACT

INTRODUCTION: Cancer cells acclimatize to the harsh tumor microenvironment by altering cellular metabolism in favor of aerobic glycolysis. This process provides a source of energy and also generates essential components for macromolecular biosynthesis, which enables cellular survival. As the dependence of cancer cells on glycolysis affects tumorigenesis, it has become an attractive target for therapeutic intervention. Several preclinical studies have shown the effectiveness of using biological targets from the glycolytic pathway for anticancer therapy. AREAS COVERED: This review provides an insight into the glycolytic pathway, highlighting potential targets for glycolytic inhibition. We then discuss recent advancement in delivery strategies that have the potential to circumvent some of the problems posed by current glycolytic inhibitors, enabling resurrection of abandoned therapeutic agents. EXPERT OPINION: Targeting the glycolysis pathway is a tactical approach for cancer therapy. However, the current nonspecific therapeutic strategies have several drawbacks such as poor bioavailability, unfavorable pharmacokinetic profile and associated nonspecific toxicity, thereby limiting preclinical investigation. In recent years, nanoparticle systems have received recognition for the delivery of therapeutic agents directly to the tumor tissue. Thus, it is envisaged that this strategy can be expanded for the delivery of current glycolytic inhibitors specifically to tumor tissues providing improved anticancer activity.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Glycolysis , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/metabolism , Aerobiosis , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Respiration/drug effects , Cell Transformation, Neoplastic/metabolism , Drug Delivery Systems/trends , Glycolysis/drug effects , Glycolysis/physiology , Humans , Metabolic Networks and Pathways/drug effects , Molecular Targeted Therapy/trends
4.
Cancer Lett ; 334(1): 86-94, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23111102

ABSTRACT

Pancreatic cancer is fourth leading cause of cancer-related deaths in the United States of America. In spite of recent advances in the current therapeutic modalities such as surgery, radiation and chemotherapy patients, the average five year survival rate remains still less than 5%. Recently, compounds from natural sources receive ample of attention as anti-cancer agents. Many epidemiological studies published over the past few decades provide a strong correlation between consumption of vegetables, fruits or plant derived products and reduced incidence of cancer. The present review focuses on the potential antitumor effects of various natural products.


Subject(s)
Anticarcinogenic Agents/pharmacology , Pancreatic Neoplasms/prevention & control , Phytochemicals/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Capsaicin/chemistry , Capsaicin/pharmacology , Curcumin/chemistry , Curcumin/pharmacology , Humans , Isothiocyanates/chemistry , Isothiocyanates/pharmacology , Pancreatic Neoplasms/drug therapy , Resveratrol , Stilbenes/chemistry , Stilbenes/pharmacology , Tea
5.
PLoS One ; 6(10): e25799, 2011.
Article in English | MEDLINE | ID: mdl-22016776

ABSTRACT

Our previous studies have shown that benzyl isothiocyanate (BITC) suppresses pancreatic tumor growth by inhibiting STAT-3; however, the exact mechanism of tumor growth suppression was not clear. Here we evaluated the effects and mechanism of BITC on pancreatic tumor angiogenesis. Our results reveal that BITC significantly inhibits neovasularization on rat aorta and Chicken-Chorioallantoic membrane. Furthermore, BITC blocks the migration and invasion of BxPC-3 and PanC-1 pancreatic cancer cells in a dose dependant manner. Moreover, secretion of VEGF and MMP-2 in normoxic and hypoxic BxPC-3 and PanC-1 cells was significantly suppressed by BITC. Both VEGF and MMP-2 play a critical role in angiogenesis and metastasis. Our results reveal that BITC significantly suppresses the phosphorylation of VEGFR-2 (Tyr-1175), and expression of HIF-α. Rho-GTPases, which are regulated by VEGF play a crucial role in pancreatic cancer progression. BITC treatment reduced the expression of RhoC whereas up-regulated the expression of tumor suppressor RhoB. STAT-3 over-expression or IL-6 treatment significantly induced HIF-1α and VEGF expression; however, BITC substantially suppressed STAT-3 as well as STAT-3-induced HIF-1α and VEGF expression. Finally, in vivo tumor growth and matrigel-plug assay show reduced tumor growth and substantial reduction of hemoglobin content in the matrigel plugs and tumors of mice treated orally with 12 µmol BITC, indicating reduced tumor angiogenesis. Immunoblotting of BITC treated tumors show reduced expression of STAT-3 phosphorylation (Tyr-705), HIF-α, VEGFR-2, VEGF, MMP-2, CD31 and RhoC. Taken together, our results suggest that BITC suppresses pancreatic tumor growth by inhibiting tumor angiogenesis through STAT-3-dependant pathway.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Isothiocyanates/pharmacology , Neovascularization, Pathologic/drug therapy , Pancreatic Neoplasms/blood supply , STAT3 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , rho GTP-Binding Proteins/antagonists & inhibitors , Animals , Cell Hypoxia/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Female , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Vitro Techniques , Isothiocyanates/therapeutic use , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Rats , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
6.
PLoS One ; 6(5): e20151, 2011.
Article in English | MEDLINE | ID: mdl-21647434

ABSTRACT

We evaluated the mechanism of capsaicin-mediated ROS generation in pancreatic cancer cells. The generation of ROS was about 4-6 fold more as compared to control and as early as 1 h after capsaicin treatment in BxPC-3 and AsPC-1 cells but not in normal HPDE-6 cells. The generation of ROS was inhibited by catalase and EUK-134. To delineate the mechanism of ROS generation, enzymatic activities of mitochondrial complex-I and complex-III were determined in the pure mitochondria. Our results shows that capsaicin inhibits about 2.5-9% and 5-20% of complex-I activity and 8-75% of complex-III activity in BxPC-3 and AsPC-1 cells respectively, which was attenuable by SOD, catalase and EUK-134. On the other hand, capsaicin treatment failed to inhibit complex-I or complex-III activities in normal HPDE-6 cells. The ATP levels were drastically suppressed by capsaicin treatment in both BxPC-3 and AsPC-1 cells and attenuated by catalase or EUK-134. Oxidation of mitochondria-specific cardiolipin was substantially higher in capsaicin treated cells. BxPC-3 derived ρ(0) cells, which lack mitochondrial DNA, were completely resistant to capsaicin mediated ROS generation and apoptosis. Our results reveal that the release of cytochrome c and cleavage of both caspase-9 and caspase-3 due to disruption of mitochondrial membrane potential were significantly blocked by catalase and EUK-134 in BxPC-3 cells. Our results further demonstrate that capsaicin treatment not only inhibit the enzymatic activity and expression of SOD, catalase and glutathione peroxidase but also reduce glutathione level. Over-expression of catalase by transient transfection protected the cells from capsaicin-mediated ROS generation and apoptosis. Furthermore, tumors from mice orally fed with 2.5 mg/kg capsaicin show decreased SOD activity and an increase in GSSG/GSH levels as compared to controls. Taken together, our results suggest the involvement of mitochondrial complex-I and III in capsaicin-mediated ROS generation and decrease in antioxidant levels resulting in severe mitochondrial damage leading to apoptosis in pancreatic cancer cells.


Subject(s)
Apoptosis/drug effects , Capsaicin/pharmacology , Electron Transport Complex III/metabolism , Electron Transport Complex I/metabolism , Oxidative Stress/drug effects , Pancreatic Neoplasms/pathology , Adenosine Triphosphate/metabolism , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Capsaicin/antagonists & inhibitors , Catalase/metabolism , Cell Line, Tumor , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex III/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Glutathione Disulfide/metabolism , Glutathione Peroxidase/metabolism , Homeostasis/drug effects , Humans , Lipid Metabolism/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
7.
Clin Cancer Res ; 17(7): 1784-95, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21350002

ABSTRACT

PURPOSE: Our previous studies have shown that benzyl isothiocyanate (BITC) suppress pancreatic cancer growth by inducing apoptosis but the molecular mechanism was unclear. In this study we hypothesized the involvement of PI3K/AKT/FOXO pathway in BITC-induced apoptosis. EXPERIMENTAL DESIGN: Mice were implanted BxPC-3 tumor xenografts and orally gavaged with 12 µmol BITC. Plasma and tumor BITC concentration was estimated by liquid chromatography/tandem mass spectrometry. BxPC-3 and PanC-1 cells were used to elucidate PI3K/AKT/FOXO pathway. Electrophoretic mobility shift assay (EMSA), DNA binding activity, immunofluorescence, and gene transfection were used to delineate the mechanism. RESULTS: BITC-treated mice showed 43% less tumor growth as compared with control mice and correlated well with the therapeutic concentrations of 6.5 µmol/L BITC achieved in plasma and 7.5 µmol/g BITC in tumor tissue. Western blot analyses and immunohistochemistry revealed that tumors from BITC-treated mice showed reduced phosphorylation of PI3K, AKT, PDK1, mTOR, FOXO1, and FOXO3a and increased apoptosis. Complementing our in vivo results, we made similar observations in a dose- and time-dependent manner in BITC-treated BxPC-3 and Panc-1 cells. Binding of FOXO1 with 14-3-3 proteins was also reduced drastically by BITC treatment indicating nuclear retention of FOXO1 and this observation was further confirmed with EMSA, immunofluorescence, DNA binding, and upregulation of FOXO-responsive proteins Bim, p27, and p21 in BxPC-3 cells. Overexpression of AKT by transient transfection significantly blocked the modulation of FOXO proteins and protected the cells from BITC-mediated apoptosis and growth suppression. CONCLUSIONS: Our results provide convincing evidence on the involvement of PI3K/AKT/FOXO pathway in BITC-mediated pancreatic tumor growth suppression.


Subject(s)
Antineoplastic Agents/pharmacology , Forkhead Transcription Factors/metabolism , Isothiocyanates/pharmacology , Pancreatic Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Acetylation , Animals , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , CREB-Binding Protein/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Down-Regulation , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Humans , I-kappa B Kinase/metabolism , Membrane Proteins/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Processing, Post-Translational , Protein Transport/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , Sirtuins/metabolism , TOR Serine-Threonine Kinases/metabolism , Transplantation, Heterologous , Tumor Burden/drug effects , p300-CBP Transcription Factors/metabolism
8.
J Enzyme Inhib Med Chem ; 25(1): 80-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20030512

ABSTRACT

Glycosidases play an important role in a wide range of physiological and pathological conditions, and have become potential targets for the discovery and development of agents useful for the treatment of diseases such as diabetes, cancer, influenza, and even AIDS. In this study, several benzimidazole derivatives were prepared from o-phenylenediamine and aromatic and heteroaromatic carboxaldehydes in very good yields, using PdCl2(CH3CN)2 as the most efficient catalyst. Synthesized compounds were assayed for their activity on yeast and rat intestinal alpha-glucosidase inhibition and cytotoxic activity against colon carcinoma cell line HT-29. Compound 3e exhibited 95.6% and 75.3% inhibition of yeast and rat intestinal alpha-glucosidase enzyme, while showing 74.8% cytotoxic activity against the HT-29 cell line at primary screening concentrations of 2.1 mM for yeast and rat intestinal alpha-glucosidase enzyme and 0.2 mM for cytotoxic activity against the HT-29 cell line, respectively. Compound 3c displayed 76% and 34.4% inhibition of yeast and rat intestinal alpha-glucosidase enzyme, and 80.4% cytotoxic activity against the HT-29 cell line at similar primary screening concentrations. The IC50 value for the most potent intestinal alpha-glucosidase inhibitor compound 3e was found to be 99.4 microM. The IC50 values for the most active cytotoxic compounds 3c and 3e were 82 microM and 98.8 microM, respectively. Both compounds displayed significant antihyperglycemic activity in starch-induced postprandial hyperglycemia in rats. This is the first report assigning yeast and rat intestinal alpha-glucosidase enzyme inhibition, cytotoxic activity against the HT-29 cell line, and antihyperglycemic activity to benzimidazole compounds 3c and 3e.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Enzyme Inhibitors/pharmacology , Glycoside Hydrolase Inhibitors , Hypoglycemic Agents/pharmacology , Animals , Cell Line, Tumor , Humans , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Rats , Rats, Wistar , Saccharomyces cerevisiae/enzymology , Spectrophotometry, Infrared
9.
Food Chem Toxicol ; 46(11): 3429-42, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18812207

ABSTRACT

There is increasing evidence that oxidative stress is implicated in pathogenesis of various diseases, including alcoholic liver injury. In the present study, we investigated the comparative protective effects of leaf, bark, root and root bark extracts of Soymida febrifuga (Roxb.) A. Juss. (Meliaceae) against ethanol induced oxidative damage in HepG2 cells. Comparatively, methanolic and aqueous extracts of bark and leaf significantly attenuated the cytotoxicity of the ethanol, as determined by cytotoxicity, lipid peroxidation, lactate dehydrogenase, alanine aminotransferases and asparatate aminotransferases, than the root and root bark extracts. Ethanol induces liver toxicity through free radical generation so initially in vitro antioxidant activity of the extracts was evaluated. Methanolic and aqueous extracts of bark and leaf have shown higher total phenolic content, reducing power, metal chelating, superoxide, hydroxyl radical, hydrogen peroxide and nitric oxide (murine macrophage cells) scavenging activity than the root and root bark extracts.


Subject(s)
Liver/drug effects , Meliaceae/chemistry , Oxidative Stress/drug effects , Phenols/pharmacology , Plant Extracts/pharmacology , Alanine Transaminase/metabolism , Aspartate Aminotransferases/metabolism , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/prevention & control , Cell Line, Tumor , Ethanol/chemistry , Humans , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Liver/enzymology , Liver Neoplasms/chemically induced , Liver Neoplasms/prevention & control , Methanol/chemistry , Phenols/analysis , Plant Bark/chemistry , Plant Extracts/analysis , Plant Leaves/chemistry , Plant Roots/chemistry , Thiobarbituric Acid Reactive Substances , Water/chemistry
10.
Phytother Res ; 22(7): 943-7, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18389470

ABSTRACT

The present study was designed to evaluate the antioxidant and antimicrobial properties of hexane (LH), methanol (LM) and aqueous (LA) extracts of Soymida febrifuga (Maliaceae) leaves, which is a traditional folk medicine in India. No pharmacological evaluation of this plant (except antiplasmodial activity) has been reported to date. Antioxidant activity of different extracts was evaluated by DPPH free radical scavenging activity, taking total phenolic content (TPC) as an index. Antimicrobial activity was tested against six bacterial and five fungal strains using the agar hole diffusion method and the minimum inhibitory concentrations (MIC) and minimum microbicidal concentration (MMC) were determined for all the test organisms against the extracts. The results showed that the methanol and aqueous extracts of leaf had a higher antioxidant activity and total phenolic content than the hexane extract. The antioxidant activity and TPC of the extracts were highly correlated. Extracts also showed several degrees of antimicrobial activity against different microbes. The methanol extract was more potent against Aspergillus fumigatus and Candida tropicana. The lowest MIC values obtained for LM, LA and LH were 78, 156, 625 microg/mL against A. fumigatus, C. tropicana and C. albicans, respectively. Hence, this study confirms that Soymida febrifuga leaves possess potent antioxidant and antimicrobial activity.


Subject(s)
Anti-Infective Agents/pharmacology , Antioxidants/pharmacology , Medicine, Traditional , Plant Extracts/pharmacology , Plant Leaves/chemistry , Plants, Medicinal/chemistry , Bacteria/drug effects , Dose-Response Relationship, Drug , Free Radical Scavengers/metabolism , Fungi/drug effects , India , Microbial Sensitivity Tests , Phenols/analysis , Plant Extracts/chemistry , Solvents
SELECTION OF CITATIONS
SEARCH DETAIL
...