Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Med Chem ; 62(15): 6913-6924, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31339316

ABSTRACT

Myeloid cell leukemia 1 (Mcl-1), an antiapoptotic member of the Bcl-2 family of proteins, whose upregulation when observed in human cancers is associated with high tumor grade, poor survival, and resistance to chemotherapy, has emerged as an attractive target for cancer therapy. Here, we report the discovery of selective small molecule inhibitors of Mcl-1 that inhibit cellular activity. Fragment screening identified thienopyrimidine amino acids as promising but nonselective hits that were optimized using nuclear magnetic resonance and X-ray-derived structural information. The introduction of hindered rotation along a biaryl axis has conferred high selectivity to the compounds, and cellular activity was brought on scale by offsetting the negative charge of the anchoring carboxylate group. The obtained compounds described here exhibit nanomolar binding affinity and mechanism-based cellular efficacy, caspase induction, and growth inhibition. These early research efforts illustrate drug discovery optimization from thienopyrimidine hits to a lead compound, the chemical series leading to the identification of our more advanced compounds S63845 and S64315.


Subject(s)
Cell Survival/physiology , Drug Discovery/methods , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Pyrimidines/chemistry , Pyrimidines/metabolism , Thiophenes/chemistry , Thiophenes/metabolism , Cell Survival/drug effects , HCT116 Cells , HeLa Cells , Humans , Protein Structure, Tertiary , Pyrimidines/pharmacology , Structure-Activity Relationship , Thiophenes/pharmacology
2.
J Comput Aided Mol Des ; 31(8): 755-775, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28712038

ABSTRACT

The knowledge of the free energy of binding of small molecules to a macromolecular target is crucial in drug design as is the ability to predict the functional consequences of binding. We highlight how a molecular dynamics (MD)-based approach can be used to predict the free energy of small molecules, and to provide priorities for the synthesis and the validation via in vitro tests. Here, we study the dynamics and energetics of the nuclear receptor REV-ERBα with its co-repressor NCoR and 35 novel agonists. Our in silico approach combines molecular docking, molecular dynamics (MD), solvent-accessible surface area (SASA) and molecular mechanics poisson boltzmann surface area (MMPBSA) calculations. While docking yielded initial hints on the binding modes, their stability was assessed by MD. The SASA calculations revealed that the presence of the ligand led to a higher exposure of hydrophobic REV-ERB residues for NCoR recruitment. MMPBSA was very successful in ranking ligands by potency in a retrospective and prospective manner. Particularly, the prospective MMPBSA ranking-based validations for four compounds, three predicted to be active and one weakly active, were confirmed experimentally.


Subject(s)
Nuclear Receptor Co-Repressor 1/agonists , Nuclear Receptor Subfamily 1, Group D, Member 1/agonists , Binding Sites , HEK293 Cells , Humans , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Nuclear Receptor Co-Repressor 1/chemistry , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/chemistry , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Protein Binding , Protein Conformation , Solvents , Structure-Activity Relationship , Surface Properties , Thermodynamics
3.
J Med Chem ; 59(15): 7167-76, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27391254

ABSTRACT

Ligand-target residence time is emerging as a key drug discovery parameter because it can reliably predict drug efficacy in vivo. Experimental approaches to binding and unbinding kinetics are nowadays available, but we still lack reliable computational tools for predicting kinetics and residence time. Most attempts have been based on brute-force molecular dynamics (MD) simulations, which are CPU-demanding and not yet particularly accurate. We recently reported a new scaled-MD-based protocol, which showed potential for residence time prediction in drug discovery. Here, we further challenged our procedure's predictive ability by applying our methodology to a series of glucokinase activators that could be useful for treating type 2 diabetes mellitus. We combined scaled MD with experimental kinetics measurements and X-ray crystallography, promptly checking the protocol's reliability by directly comparing computational predictions and experimental measures. The good agreement highlights the potential of our scaled-MD-based approach as an innovative method for computationally estimating and predicting drug residence times.


Subject(s)
Glucokinase/chemistry , Molecular Dynamics Simulation , Crystallography, X-Ray , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucokinase/antagonists & inhibitors , Glucokinase/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Kinetics , Ligands , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Time Factors
4.
J Med Chem ; 59(2): 687-706, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26685731

ABSTRACT

7-Azaindole has been identified as a novel bidentate anchor point for allosteric glucokinase activators. A systematic investigation around three principal parts of the new small molecule glucokinase activators led to a robust SAR in agreement with structural data that also helped to assess the conformational flexibility of the allosteric activation site. The increase in glucose uptake resulting from glucokinase activation in hepatocytes in vitro translated into the efficient lowering of glucose levels in vivo with the best compounds.


Subject(s)
Enzyme Activators/chemistry , Enzyme Activators/pharmacology , Glucokinase/metabolism , Indoles/chemistry , Indoles/pharmacology , Animals , Crystallography, X-Ray , Glucose/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hypoglycemic Agents/pharmacology , Models, Molecular , Molecular Conformation , Primary Cell Culture , Rats , Structure-Activity Relationship
5.
Br J Pharmacol ; 169(5): 999-1010, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23488540

ABSTRACT

BACKGROUND AND PURPOSE: Small molecule glucokinase activators (GKAs) have been associated with potent antidiabetic efficacy and hepatic steatosis in rodents. This study reports the discovery of S 50131 and S 51434, two novel GKAs with an original scaffold and an atypical pharmacological profile. EXPERIMENTAL APPROACH: Activity of the compounds was assessed in vitro by measuring activation of recombinant glucokinase, stimulation of glycogen synthesis in rat hepatocytes and increased insulin secretion from rat pancreatic islets of Langerhans. Efficacy and safety in vivo were evaluated after oral administration in db/db mice by measuring glycaemia, HbA1c and dyslipidaemia-associated events. KEY RESULTS: S 50131 and S 51434 activated GK and stimulated glycogen synthesis in hepatocytes and insulin secretion from pancreatic islets. Unexpectedly, while both compounds effectively lowered glycaemia after acute oral administration, they did not decrease HbA1c after a 4-week treatment in db/db mice. This lack of antidiabetic efficacy was associated with increased plasma free fatty acids (FFAs), contrasting with the effect of GKA50 and N00236460, two GKAs with sustained HbA1c lowering activity but neutral regarding plasma FFAs. S 50131, but not S 51434, also induced hepatic steatosis, as did GKA50 and N00236460. However, a shorter, 4-day treatment resulted in increased hepatic triglycerides without changing the plasma FFA levels, demonstrating dynamic alterations in the lipid profile over time. CONCLUSIONS AND IMPLICATIONS: In addition to confirming the occurrence of dyslipidaemia with GKAs, these findings provide new insights into understanding how such compounds may sustain or lose efficacy over time.


Subject(s)
Diabetes Mellitus/drug therapy , Enzyme Activators/therapeutic use , Glucokinase/metabolism , Hypoglycemic Agents/therapeutic use , Nicotinic Acids/therapeutic use , Polycyclic Compounds/therapeutic use , Animals , Blood Glucose/analysis , Caco-2 Cells , Cells, Cultured , Cholesterol/blood , Diabetes Mellitus/blood , Diabetes Mellitus/metabolism , Enzyme Activators/pharmacology , Fatty Acids, Nonesterified/blood , Glycated Hemoglobin/metabolism , Glycogen/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Intestinal Absorption , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Nicotinic Acids/pharmacology , Polycyclic Compounds/pharmacology , Rats, Sprague-Dawley , Rats, Wistar , Treatment Outcome , Triglycerides/metabolism
6.
Br J Pharmacol ; 168(2): 339-53, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22925001

ABSTRACT

BACKGROUND AND PURPOSE: Small-molecule glucokinase activators (GKAs) are currently being investigated as therapeutic options for the treatment of type 2 diabetes (T2D). Because liver overexpression of glucokinase is thought to be associated with altered lipid profiles, this study aimed at assessing the potential lipogenic risks linked to oral GKA administration. EXPERIMENTAL APPROACH: Nine GKA candidates were qualified for their ability to activate recombinant glucokinase and to stimulate glycogen synthesis in rat hepatocytes and insulin secretion in rat INS-1E cells. In vivo activity was monitored by plasma glucose and HbA1c measurements after oral administration in rodents. Risk-associated effects were assessed by measuring hepatic and plasma triglycerides and free fatty acids, as well as plasma aminotransferases, and alkaline phosphatase. KEY RESULTS: GKAs, while efficiently decreasing glycaemia in acute conditions and HbA1c levels after chronic administration in hyperglycemic db/db mice, were potent inducers of hepatic steatosis. This adverse outcome appeared as soon as 4 days after daily oral administration at pharmacological doses and was not transient. GKA treatment similarly increased hepatic triglycerides in diabetic and normoglycaemic rats, together with a pattern of metabolic phenotypes including different combinations of increased plasma triglycerides, free fatty acids, alanine and aspartyl aminotransferases, and alkaline phosphatase. GKAs belonging to three distinct structural families induced hepatic steatosis in db/db mice, arguing in favour of a target-mediated, rather than a chemical class-mediated, effect. CONCLUSION AND IMPLICATIONS: Given the risks associated with fatty liver disease in the general population and furthermore in patients with T2D, these findings represent a serious warning for the use of GKAs in humans. LINKED ARTICLE: This article is commented on by Rees and Gloyn, pp. 335-338 of this issue. To view this commentary visit http://dx.doi.org/10.1111/j.1476-5381.2012.02201.x.


Subject(s)
Enzyme Activators/pharmacology , Fatty Liver/chemically induced , Glucokinase/metabolism , Hypoglycemic Agents/pharmacology , Lipid Metabolism/drug effects , Animals , Blood Glucose/analysis , Caco-2 Cells , Cell Line, Tumor , Cells, Cultured , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Enzyme Activators/therapeutic use , Fatty Liver/metabolism , Glycated Hemoglobin/analysis , Hepatocytes/metabolism , Homeostasis/drug effects , Humans , Hypoglycemic Agents/therapeutic use , Intestinal Absorption , Male , Mice , Rats , Rats, Sprague-Dawley , Rats, Wistar , Rats, Zucker
7.
Anticancer Agents Med Chem ; 12(3): 194-201, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22044003

ABSTRACT

Protein kinases are important enzymes in solid tumour and leukaemia pathologies. Their structures are well understood at the atomic level and their key role in the progression of certain cancers makes them valuable targets for anti-cancer therapy. Through medicinal chemical approaches, we developed an efficient preparative stereospecific synthesis of N12, N13 pyran-bridged indolocarbazoles that opens access to functional diversity within this previously challenging series. We focused upon the indolocarbazole class of chemical inhibitors, which includes S27888, an inhibitor we previously identified. We used biochemical and cell-based assays to identify small molecule inhibitors of Checkpoint kinase 1 (Chk1), a serine/threonine protein kinase that is activated when cancer cells are treated with genotoxic agents. These compounds show a promising inhibitory profile on Chk1. Furthermore, these compounds are active against FLT3, which is a tyrosine kinase that is frequently activated in human leukaemias. These data suggest that this chemical class may provide a source of therapeutic compounds for a broad range of human cancers.


Subject(s)
Carbazoles/chemical synthesis , DNA Damage , Indoles/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Protein Kinases/metabolism , Pyran Copolymer/chemistry , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Carbazoles/chemistry , Carbazoles/therapeutic use , Checkpoint Kinase 1 , DNA Damage/drug effects , HT29 Cells , Humans , Indoles/pharmacology , Neoplasms/drug therapy , Neoplasms/enzymology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyran Copolymer/pharmacology , fms-Like Tyrosine Kinase 3/metabolism
8.
J Immunol ; 186(1): 464-70, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21098231

ABSTRACT

Monocytes and macrophages are targets of HIV-1 infection and play critical roles in multiple aspects of viral pathogenesis. During the differentiation of monocytes to macrophages, adhesion molecules such as integrins are upregulated; therefore, they provide signals that control the process and subsequently may render macrophages more susceptible to HIV-1 infection. Previous work demonstrated that blocking α(v)-containing integrins triggered a signal transduction pathway leading to the inhibition of NF-κB-dependent HIV-1 transcription. In this paper, we show the influence of the different α(v)-coupled ß integrins in HIV-1 replication in macrophages. Inhibition of ß integrins, either by specific mAbs, small arginine-glycine-aspartic acid (RGD) mimetic compounds, or RNA interference, showed that integrin ß(5) was the major contributor to the integrin-mediated blockade of HIV-1 replication. Importantly, such inhibition did not induce changes in cell adhesion to the substrate. In conclusion, our results reveal a significant role of the integrin dimer α(v)ß(5) in HIV-1 infection of macrophages.


Subject(s)
Antiviral Agents/metabolism , HIV-1/immunology , Integrin beta Chains/physiology , Protein Multimerization/immunology , Receptors, Vitronectin/physiology , Virus Replication/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Genetic Predisposition to Disease , HIV Infections/immunology , HIV Infections/metabolism , HIV-1/pathogenicity , Humans , Integrin beta Chains/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Protein Binding/genetics , Protein Binding/immunology , Protein Multimerization/genetics , Protein Subunits/biosynthesis , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Virus Replication/genetics
9.
Nat Med ; 15(4): 392-400, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19305413

ABSTRACT

Inhibitors of alpha(v)beta(3) and alpha(v)beta(5) integrin have entered clinical trials as antiangiogenic agents for cancer treatment but generally have been unsuccessful. Here we present in vivo evidence that low (nanomolar) concentrations of RGD-mimetic alpha(v)beta(3) and alpha(v)beta(5) inhibitors can paradoxically stimulate tumor growth and tumor angiogenesis. We show that low concentrations of these inhibitors promote VEGF-mediated angiogenesis by altering alpha(v)beta(3) integrin and vascular endothelial growth factor receptor-2 trafficking, thereby promoting endothelial cell migration to VEGF. The proangiogenic effects of low concentrations of RGD-mimetic integrin inhibitors could compromise their efficacy as anticancer agents and have major implications for the use of RGD-mimetic compounds in humans.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Integrin alphaVbeta3/therapeutic use , Lung Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Receptors, Vitronectin/therapeutic use , Animals , Disease Models, Animal , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Vascular Endothelial Growth Factor A/pharmacology
10.
Blood ; 113(6): 1278-86, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-18840709

ABSTRACT

Monocytes and macrophages are an important reservoir of human immunodeficiency virus (HIV) and may represent the largest reservoir of this virus in tissues. Differentiation of monocytes into macrophages leads to cell attachment and susceptibility to infection and replication of HIV. Among other cell-surface molecules, integrins are overexpressed during monocyte-macrophage differentiation and may play a role in the replication cycle of envelope viruses including HIV. Here, we show that inhibition of alphaV integrin in monocyte-derived macrophages, by RNA interference or their inhibition by a selective small heterocyclic RGD-mimetic nonpeptide compound, inhibited the replication of HIV in the absence of cytotoxicity. Interference or inhibition of alphaV integrins triggered a signal transduction pathway, leading to down-regulation of nuclear factor-kappaB-dependent HIV-1 transcription. Such inhibition was mediated by a MAP-kinase signaling cascade, probably involving ERK1/2, p38-mitogen-activated protein kinases, and HSP27. In conclusion, our results reveal a significant role of integrin alphaV-mediated adhesion in HIV-1 infection of macrophages.


Subject(s)
Cell Adhesion/physiology , HIV Infections/virology , HIV-1/physiology , Integrin alphaV/metabolism , Macrophages/virology , Blotting, Western , Cell Survival , Cells, Cultured , Enzyme Inhibitors/pharmacology , Flow Cytometry , Fluorescent Antibody Technique , HIV Infections/metabolism , HeLa Cells , Humans , Macrophages/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/metabolism , Monocytes/virology , Transcription, Genetic/drug effects , Virus Replication/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Virology ; 371(1): 155-64, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-17950396

ABSTRACT

The adenovirus penton base is a strategic protein involved in the virus internalisation pathway through interaction between its RGD sequences and integrin. In some human adenovirus serotypes, this pentameric protein features the ability of interacting together by twelve, leading to the formation of a symmetric nanoparticle called dodecahedron (Dd). This non-infectious adenovirus-like particle exhibiting sixty RGD sequences interacts with integrin but also with heparan sulfate proteoglycans (HSPGs) expressed at the cell surface. In this study, we discriminate the respective importance of HSPGs and integrin on human adenovirus serotype 3 dodecahedron attachment and entry. Using different cell lines and a specific integrin inhibitor, we have determined that HSPGs are mainly responsible for particle attachment to the cell surface, favouring a strictly required interaction with integrin that triggers internalisation. No other receptors are involved in Dd entry and integrins on their own can mediate the particle entry in HSPGs-deficient cells. Moreover, integrin recognition by Dd is highly susceptible to cations and particularly to manganese that enhances particle binding by 4- to 7-fold compared to calcium. Interestingly, investigations on Dd receptors along the cell cycle revealed an enhanced particle targeting to mitotic cells and a loss of internalisation at this stage. This phenomenon observed with both HeLa- and HSPGs-deficient cells, depends on integrin remodelling during mitosis. This provides new clues for the use of this adenovirus nanoparticle as a delivery vector and sheds light on the integrin and HSPGs relationship in both resting and dividing cells.


Subject(s)
Adenoviruses, Human/physiology , Capsid Proteins/metabolism , Cell Cycle , Heparin/analogs & derivatives , Integrins/physiology , Proteoglycans/physiology , Adenoviruses, Human/ultrastructure , Animals , CHO Cells , Cell Adhesion/physiology , Cricetinae , Cricetulus , HeLa Cells , Heparin/physiology , Humans
12.
Blood ; 108(9): 3035-44, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16835373

ABSTRACT

Alpha v integrins are thought to play an important role in tumor angiogenesis. However, discrepancies between findings with Arg-Gly-Asp (RGD) mimetics, which block angiogenesis in animal models, and knockout mice, in which loss of some alpha v integrins enhances tumor angiogenesis, raise questions concerning the function of these integrins and the precise role of alpha v substrate mimetics in antiangiogenic therapies. We have examined the effects of a novel non-peptide RGD mimetic, S 36578-2, on human endothelial cells to elucidate its antagonist activity and to identify possible agonist functions. S 36578-2 is highly selective for alpha v beta3 and alpha v beta5 integrins and induces detachment, caspase-8 activation, and apoptosis in human umbilical endothelial cells (HUVECs) plated on vitronectin. Importantly, the compound has no effect on the morphology or survival of cells plated on interstitial matrix components such as fibronectin, and it does not potentiate the apoptotic process in suspended cells. Identical results were obtained with a cyclic RGD peptide with similar target specificity. In microvascular endothelial cells, S 36578-2-induced death was also linked to its antiadhesive effect, with established lines markedly more resistant than primary cultures to the antiadhesive and proapoptotic effects. Altogether, these findings have important implications for the development of this class of antiangiogenics.


Subject(s)
Anoikis/physiology , Benzocycloheptenes/pharmacology , Endothelium, Vascular/physiology , Integrin alphaVbeta3/antagonists & inhibitors , Integrins/antagonists & inhibitors , Oligopeptides/pharmacology , Receptors, Vitronectin/antagonists & inhibitors , gamma-Aminobutyric Acid/analogs & derivatives , Anoikis/drug effects , Apoptosis/drug effects , Cell Culture Techniques , Cell Death/drug effects , Cell Death/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fetal Blood/cytology , Humans , Integrin alphaVbeta3/drug effects , Integrin alphaVbeta3/genetics , Integrins/drug effects , Integrins/genetics , Neoplasms/blood supply , Neovascularization, Pathologic , Receptors, Vitronectin/drug effects , Receptors, Vitronectin/genetics , Umbilical Veins , Vitronectin/physiology , gamma-Aminobutyric Acid/pharmacology
13.
Mol Pharmacol ; 63(6): 1281-8, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12761337

ABSTRACT

The role of adhesion molecules, such as alphav integrins, in the control of the survival of quiescent tumor cells is unclear. We used S 34961, a novel small molecule alphav integrin antagonist, to investigate the role of integrin-signaling in the survival of populations of quiescent human HT-29 and HCT 116 colon carcinoma cells. S 34961 at 1 microM induced detachment, but cells retained viability, existing as clusters. Nonligated beta-integrins may recruit and activate caspase-8 [J Cell Biol 155:459-470, 2001]. However, congruent with the absence of apoptosis, no activation of caspase-8 in these cells was detected after incubation with S 34961. A rapid (2 h) change in conformation of the N terminus of proapoptotic Bak was observed before detachment, together with a decrease in phosphorylation of focal adhesion kinase (2 h) and subsequent (8 h) decreases in phosphorylation of extracellular signal-regulated kinase-1/2 and Akt. Together, these results suggested that although treatment with S 34961 has no effect on survival per se, it may reduce the survival threshold of the tumor cells, with Bak in an activated state. Indeed, concomitant incubation of S 34961 with 10 microM U-0126 (a mitogen-activated protein kinase kinase inhibitor) was found to lead to apoptosis (at 24 h), whereas U-0126 alone had no effect. Together, these observations could guide the use of combination therapy with integrin antagonists in the clinic.


Subject(s)
Caspases/metabolism , Colonic Neoplasms/pathology , Cycloheptanes/pharmacology , Integrin alpha5/metabolism , Integrins/antagonists & inhibitors , Pyridines/pharmacology , Apoptosis , Caspase 8 , Caspase 9 , Cell Adhesion , Cell Survival/drug effects , Colonic Neoplasms/enzymology , Enzyme Activation , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , HT29 Cells , Humans , Integrin alpha5/drug effects , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , bcl-2 Homologous Antagonist-Killer Protein
14.
Bioorg Med Chem Lett ; 12(22): 3291-6, 2002 Nov 18.
Article in English | MEDLINE | ID: mdl-12392735

ABSTRACT

A novel series of potent and specific alpha(v) integrin antagonists has been obtained by aminoalkyl substitutions on benzocyloheptene acetic acids as a rigid GD bioisostere. The preferred compounds 1-2, 1-3 and 1-8, showed nano- to subnanomolar IC(50) values on alpha(v)beta(3) and alpha(v)beta(5) integrins, with favorable pharmacokinetics.


Subject(s)
Benzocycloheptenes/chemical synthesis , Benzocycloheptenes/pharmacology , Integrin alphaV/drug effects , Animals , Biological Availability , Cell Adhesion/drug effects , Cell Membrane Permeability , Humans , Inhibitory Concentration 50 , Integrin alphaVbeta3/antagonists & inhibitors , Integrins/antagonists & inhibitors , Microsomes, Liver/metabolism , Molecular Conformation , Oligopeptides , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Rats , Receptors, Vitronectin/antagonists & inhibitors , Structure-Activity Relationship , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...