Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Inflamm Bowel Dis ; 24(6): 1251-1265, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29669006

ABSTRACT

Background: Anti-tumor necrosis factor alpha (anti-TNFα) therapy has become the mainstay of therapy for Crohn's disease (CD). However, post-therapy, the recurrence rate is still high. The aim of this study was to dissect the molecular mechanism for recurrence of CD treated with anti-TNFα therapy and investigate novel therapeutic options that could induce complete remission. Methods: We re-analyzed publicly available mucosal gene expression data from CD patients pre- and post-infliximab therapy to extract the transcriptional differences between responders and healthy controls. We used a systematic computational approach based on identified differences to discover novel therapies and validated this prediction through in vitro and in vivo experimentation. Results: We identified a set of 3545 anti-TNFα therapy-untreatable genes (TUGs) that are significantly regulated in intestinal epithelial cells, which remain altered during remission. Pathway enrichment analysis of these genes clearly showed excessive growth state and suppressed terminal differentiation, whereas immune components were clearly resolved. Through in silico screening strategy, we observed that MEK inhibitors were predicted to revert expression of genes dysregulated in infliximab responders. In vitro transcriptome analysis demonstrated that selective MEK1/2 inhibitor significantly normalized reference genes from TUGs. In addition, in vitro functional study proved that MEK1/2 inhibitor facilitated intestinal epithelial differentiation. Finally, using murine colitis model, administration of MEK1/2 inhibitor significantly improved diarrhea and histological score. Conclusions: Our data revealed the abnormalities in anti-TNFα responders' CD colons that would be cause of recurrence of CD. Also, we provided evidence regarding MEK1/2 inhibitor as a potential treatment against CD to achieve sustainable remission.


Subject(s)
Crohn Disease/drug therapy , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adalimumab , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Caco-2 Cells , Colon/pathology , Crohn Disease/metabolism , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Humans , Infliximab , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recurrence , Remission Induction
2.
PLoS One ; 12(10): e0186587, 2017.
Article in English | MEDLINE | ID: mdl-29065190

ABSTRACT

Scleroderma has clinical characteristics including skin and other tissue fibrosis, but there is an unmet need for anti-fibrotic therapy. Halofuginone (HF) is a well-known anti-fibrosis agent in preclinical and clinical studies which exerts its effect via inhibition of TGF-ß/Smad3 signaling pathway. Recently, prolyl-tRNA synthetase (PRS) was elucidated as a target protein for HF that binds to the proline binding site of the catalytic domain of PRS. Here, we characterized a new class of PRS inhibitor (T-3833261) that is carefully designed in a way that binds to the ATP site of the catalytic domain and does not disrupt binding of proline. The anti-fibrotic activity and the mechanism of action for T-3833261 on TGF-ß-induced fibrotic assay were compared with those of HF in primary human skin fibroblast. We evaluated in vivo effect of topical application of T-3833261 and HF on TGF-ß-induced fibrotic genes expression in mice. We found that T-3833261 suppressed TGF-ß-induced α-smooth muscle actin (α-SMA) and type I collagen α1 (COL1A1) expression through the Smad3 axis in a similar fashion to HF. In vivo topical application of T-3833261 reduced the increase of fibrotic genes expression such as α-Sma, Col1a1 and Col1a2 by TGF-ß intradermal injection to the ear of a mouse. We revealed that T-3833261 is more effective than HF under the conditions of high proline concentration, as reported in fibrotic tissues. These results suggest the potential of ATP competitive PRS inhibitors for the treatment of fibrotic diseases such as scleroderma.


Subject(s)
Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Scleroderma, Systemic/drug therapy , Animals , Cells, Cultured , Drug Discovery , Enzyme Inhibitors/therapeutic use , Humans , Male , Mice , Mice, Inbred C3H
3.
Mol Cancer Ther ; 16(2): 273-284, 2017 02.
Article in English | MEDLINE | ID: mdl-27903753

ABSTRACT

Dysregulation of lysine (K)-specific demethylase 1A (LSD1), also known as KDM1A, has been implicated in the development of various cancers, including leukemia. Here, we describe the antileukemic activity and mechanism of action of T-3775440, a novel irreversible LSD1 inhibitor. Cell growth analysis of leukemia cell lines revealed that acute erythroid leukemia (AEL) and acute megakaryoblastic leukemia cells (AMKL) were highly sensitive to this compound. T-3775440 treatment enforced transdifferentiation of erythroid/megakaryocytic lineages into granulomonocytic-like lineage cells. Mechanistically, T-3775440 disrupted the interaction between LSD1 and growth factor-independent 1B (GFI1B), a transcription factor critical for the differentiation processes of erythroid and megakaryocytic lineage cells. Knockdown of LSD1 and GFI1B recapitulated T-3775440-induced transdifferentiation and cell growth suppression, highlighting the significance of LSD1-GFI1B axis inhibition with regard to the anti-AML effects of T-3775440. Moreover, T-3775440 exhibited significant antitumor efficacy in AEL and AMKL xenograft models. Our findings provide a rationale for evaluating LSD1 inhibitors as potential treatments and indicate a novel mechanism of action against AML, particularly AEL and AMKL. Mol Cancer Ther; 16(2); 273-84. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transdifferentiation/drug effects , Histone Demethylases/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cluster Analysis , Computational Biology/methods , Disease Models, Animal , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Gene Knockdown Techniques , Hematopoiesis/genetics , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mice , Molecular Targeted Therapy , Protein Binding , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Xenograft Model Antitumor Assays
4.
Biochem Biophys Res Commun ; 480(3): 380-386, 2016 Nov 18.
Article in English | MEDLINE | ID: mdl-27771247

ABSTRACT

Inhibitors of apoptosis proteins (IAPs) are antiapoptotic regulators that block cell death, and are frequently overexpressed in several human cancers, where they facilitate evasion of apoptosis and promote cell survival. IAP antagonists are also known as second mitochondria-derived activator of caspase (SMAC)-mimetics, and have recently been considered as novel therapeutic agents for inducing apoptosis, alone and in combination with other anticancer drugs. In this study, we showed that T-3256336, the orally available IAP antagonist has synergistically enhances the antiproliferative effects of the NEDD8-activating enzyme (NAE) inhibitor pevonedistat (TAK-924/MLN4924), and these effects were attenuated by a TNFα-neutralizing antibody. In the present mechanistic analyses, pevonedistat induced TNFα mRNA and triggered IAP antagonist-dependent extrinsic apoptotic cell death in cancer cell lines. Furthermore, synergistic effects of the combination of T-3256336 and pevonedistat were demonstrated in a HL-60 mouse xenograft model. Our findings provide mechanistic evidence of the effects of IAP antagonists in combination with NAE inhibitors, and demonstrate the potential of a new combination therapy for cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cyclopentanes/administration & dosage , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Oligopeptides/administration & dosage , Pyrazines/administration & dosage , Pyrimidines/administration & dosage , Ubiquitins/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Humans , Mice , NEDD8 Protein , Neoplasms, Experimental/pathology , Treatment Outcome
5.
Biochem Biophys Res Commun ; 479(2): 179-185, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27608596

ABSTRACT

Inhibitors of apoptosis proteins (IAPs) are a family of antiapoptotic regulators that have attracted attention as potential targets for cancer therapeutics. Although recent studies have revealed that small-molecule IAP antagonists induce tumor selective cell death in an autocrine tumor necrosis factor (TNF)α-dependent manner, the single-agent efficacy of IAP antagonists is restricted to a small subset of cancer cells. In this study, we showed that the single-agent activity of T-3256336 was limited to a few cancer cell lines in vitro, and these cell lines were defined by relatively high levels of TNFα mRNA expression. However, some other cancer cells, including PANC-1 cells, become drastically sensitive to T-3256336 when costimulated with exogenous TNFα. In PANC-1 mouse xenograft models, the administration of T-3256336 increased levels of several cytokines including TNFα and lead to tumor regression as a single agent, which was attenuated by the neutralization of circulating mouse TNFα with an antibody. These results suggest dual roles of IAP antagonists, increase systemic cytokines including TNFα, and sensitization of tumors to IAP antagonist-induced death.


Subject(s)
Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Oligopeptides/pharmacology , Pyrazines/pharmacology , Tumor Necrosis Factor-alpha/genetics , Administration, Oral , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic/drug effects , HL-60 Cells , Humans , Inhibitor of Apoptosis Proteins/metabolism , MCF-7 Cells , Mice , Neoplasms/genetics , Neoplasms/metabolism , Oligopeptides/administration & dosage , Pyrazines/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/drug effects , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/pharmacology , Xenograft Model Antitumor Assays
6.
Bioorg Med Chem ; 21(24): 7938-54, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24169315

ABSTRACT

We previously reported octahydropyrrolo[1,2-a]pyrazine derivative 2 (T-3256336) as a potent antagonist for inhibitors of apoptosis (IAP) proteins. Because compound 2 was susceptible to MDR1 mediated efflux, we developed another scaffold, hexahydropyrazino[1,2-a]indole, using structure-based drug design. The fused benzene ring of this scaffold was aimed at increasing the lipophilicity and decreasing the basicity of the scaffold to improve the membrane permeability across MDR1 expressing cells. We established a chiral pool synthetic route to yield the desired tricyclic chiral isomers. Chemical modification of the core scaffold led to a representative compound 50, which showed strong inhibition of IAP binding (X chromosome-linked IAP [XIAP]: IC50 23 nM and cellular IAP [cIAP]: IC50 1.1 nM) and cell growth inhibition (MDA-MB-231 cells: GI50 2.8 nM) with high permeability and low potential of MDR1 substrate.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Membrane Permeability/drug effects , Drug Design , Indoles/pharmacology , Inhibitor of Apoptosis Proteins/pharmacology , Pyrazines/pharmacology , ATP Binding Cassette Transporter, Subfamily B , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Indoles/chemical synthesis , Indoles/chemistry , Inhibitor of Apoptosis Proteins/chemical synthesis , Inhibitor of Apoptosis Proteins/chemistry , Models, Molecular , Molecular Structure , Pyrazines/chemical synthesis , Pyrazines/chemistry , Structure-Activity Relationship
7.
Bioorg Med Chem ; 21(18): 5725-37, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23928071

ABSTRACT

We recently reported the discovery of octahydropyrrolo[1,2-a]pyrazine A as a lead compound for an inhibitor of apoptosis proteins (IAP) antagonist. To develop IAP antagonists with favorable PK profiles, we designed novel tri-cyclic compounds, octahydro-1H-cyclopropa[4,5]pyrrolo[1,2-a]pyrazines 1 and 2 based on co-crystal structural analysis of A with cellular IAP-1 (cIAP-1). The additional cyclopropane moiety was used to block the predicted metabolic site of compound A without detriment to the binding affinity for cIAP. Compounds 1 and 2 were stereoselectively synthesized via intermediates 4a and 5b', which were obtained by Simmons-Smith cyclopropanation of ethylester 3a and silyl ether 3b'. Compounds 1 and 2 showed strong growth inhibition in MDA-MB-231 breast cancer cells and improved metabolic stability in comparison to A. Compound 2 exhibited significant in vivo PD effects to increase tumor necrosis factor-alpha mRNA in a dose dependent manner.


Subject(s)
Drug Design , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Pyrazines/chemistry , Pyrroles/chemical synthesis , Animals , Benzopyrans/chemical synthesis , Benzopyrans/pharmacokinetics , Benzopyrans/therapeutic use , Binding Sites , Breast Neoplasms/drug therapy , Cell Line, Tumor , Crystallography, X-Ray , Female , Half-Life , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mice , Molecular Dynamics Simulation , Protein Structure, Tertiary , Pyrazines/chemical synthesis , Pyrazines/pharmacokinetics , Pyrazines/therapeutic use , Pyrroles/chemistry , Pyrroles/pharmacokinetics , Pyrroles/therapeutic use , RNA, Messenger/metabolism , Stereoisomerism , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
J Med Chem ; 56(16): 6478-94, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23906342

ABSTRACT

With the aim of discovering a selective kinase inhibitor targeting pan-RAF kinase inhibition, we designed novel 1,3-benzothiazole derivatives based on our thiazolo[5,4-b]pyridine class RAF/VEGFR2 inhibitor 1 and developed a regioselective cyclization methodology for the C-7-substituted 1,3-benzothiazole scaffold utilizing meta-substituted anilines. Eventually, we selected 7-cyano derivative 8B (TAK-632) as a development candidate and confirmed its binding mode by cocrystal structure with BRAF. Accommodation of the 7-cyano group into the BRAF-selectivity pocket and the 3-(trifluoromethyl)phenyl acetamide moiety into the hydrophobic back pocket of BRAF in the DFG-out conformation contributed to enhanced RAF potency and selectivity vs VEGFR2. Reflecting its potent pan-RAF inhibition and slow off-rate profile, 8B demonstrated significant cellular activity against mutated BRAF or mutated NRAS cancer cell lines. Furthermore, in both A375 (BRAF(V600E)) and HMVII (NRAS(Q61K)) xenograft models in rats, 8B demonstrated regressive antitumor efficacy by twice daily, 14-day repetitive administration without significant body weight loss.


Subject(s)
Benzothiazoles/chemical synthesis , Benzothiazoles/pharmacology , Drug Discovery , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Benzothiazoles/chemistry , Blood-Brain Barrier , Cell Line, Tumor , Crystallography, X-Ray , Drug Evaluation, Preclinical , Humans , Models, Molecular , Protein Kinase Inhibitors/chemistry , Surface Plasmon Resonance
9.
J Med Chem ; 56(3): 1228-46, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23298277

ABSTRACT

To develop novel inhibitor of apoptosis (IAP) proteins antagonists, we designed a bicyclic octahydropyrrolo[1,2-a]pyrazine scaffold as a novel proline bioisostere. This design was based on the X-ray co-crystal structure of four N-terminal amino acid residues (AVPI) of the second mitochondria-derived activator of caspase (Smac) with the X-chromosome-linked IAP (XIAP) protein. Lead optimization of this scaffold to improve oral absorption yielded compound 45, which showed potent cellular IAP1 (cIAP1 IC(50): 1.3 nM) and XIAP (IC(50): 200 nM) inhibitory activity, in addition to potent tumor growth inhibitory activity (GI(50): 1.8 nM) in MDA-MB-231 breast cancer cells. X-ray crystallographic analysis of compound 45 bound to XIAP and to cIAP1 was achieved, revealing the various key interactions that contribute to the higher cIAPI affinity of compound 45 over XIAP. Because of its potent IAP inhibitory activities, compound 45 (T-3256336) caused tumor regression in a MDA-MB-231 tumor xenograft model (T/C: -53% at 30 mg/kg).


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Oligopeptides/chemistry , Oligopeptides/pharmacology , Peptidomimetics , Proline/chemistry , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Crystallography, X-Ray , Drug Design , Magnetic Resonance Spectroscopy , Models, Molecular , Oligopeptides/chemical synthesis
10.
Mol Cancer Ther ; 12(2): 230-40, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23243058

ABSTRACT

Inhibitor of apoptosis proteins (IAP), which are key regulators of apoptosis, are inhibited by second mitochondria-derived activator of caspase (SMAC). Small-molecule IAP antagonists have recently been reported as novel therapeutic treatments for cancer. In this study, we showed that the octahydro-pyrrolo[1,2-a]pyrazine derivative, T-3256336, is a novel and orally available small-molecule IAP antagonist. T-3256336 selectively binds to and antagonizes protein interactions involving cellular IAP-1 (cIAP-1), cIAP-2, and X-linked IAP (XIAP). T-3256336 induced the rapid proteasomal degradation of cIAP-1 and activated TNF-α-dependent extrinsic apoptosis signaling in cultured cells. In a MDA-MB-231-Luc breast cancer xenograft model, T-3256336 induced cIAP-1 degradation, TNF-α production, and caspase activation in tumors, which resulted in strong antitumor activities. T-3256336 induced increases in the plasma levels of TNF-α and fragmented cytokeratin-18, which correlated with the antitumor potency in MDA-MB-231-Luc xenograft models. This study provided further insights into biomarkers of IAP antagonists. Furthermore, our data provided evidence that T-3256336 is a promising new anticancer drug worthy of further evaluation and development.


Subject(s)
Breast Neoplasms/drug therapy , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Oligopeptides/pharmacology , Pyrazines/pharmacology , Animals , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Caspases/metabolism , Cell Line , Cell Survival/drug effects , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factors/metabolism
11.
Bioorg Med Chem ; 20(18): 5600-15, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22883026

ABSTRACT

Our aim was to discover RAF/vascular endothelial growth factor receptor 2 (VEGFR2) inhibitors that possess strong activity and sufficient oral absorption, and thus, we selected a 5-amino-linked thiazolo[5,4-d]pyrimidine derivative as the lead compound because of its potential kinase inhibitory activities and its desired solubility. The novel tertiary 1-cyano-1-methylethoxy substituent was designed to occupy the hydrophobic region of 'back pocket' of BRAF on the basis of the X-ray co-crystal structure data of BRAF. In addition, we found that N-methylation of the amine linker could control the twisted molecular conformation leading to improved solubility. These approaches produced N-methyl thiazolo[5,4-b]pyridine-5-amine derivative 5. To maximize the in vivo efficacy, we attempted salt formation of 5. Our result indicated that the besylate monohydrate salt form (5c) showed significant improvement of both solubility and oral absorption. Owing to the improved physicochemical properties, compound 5c demonstrated regressive antitumor efficacy in a HT-29 xenograft model.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Neoplasms, Experimental/drug therapy , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Thiazoles/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , HT29 Cells , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Mice , Microsomes/drug effects , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemistry , Rats , Rats, Inbred F344 , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
12.
Bioorg Med Chem ; 20(15): 4680-92, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22763369

ABSTRACT

As an alternative to the previously reported solid dispersion formulation for enhancing the oral absorption of thiazolo[5,4-b]pyridine 1, we investigated novel N-acyl imide prodrugs of 1 as RAF/vascular endothelial growth factor receptor 2 (VEGFR2) inhibitors. Introducing N-acyl promoieties at the benzanilide position gave chemically stable imides. N-tert-Butoxycarbonyl (Boc) introduced imide 6 was a promising prodrug, which was converted to the active compound 1 after its oral administration in mice. Cocrystals of 6 with AcOH (6b) possessed good physicochemical properties with moderate thermodynamic solubility (19µg/mL). This crystalline prodrug 6b was rapidly and enzymatically converted into 1 after its oral absorption in mice, rats, dogs, and monkeys. Prodrug 6b showed in vivo antitumor regressive efficacy (T/C=-6.4%) in an A375 melanoma xenograft model in rats. Hence, we selected 6b as a promising candidate and are performing further studies. Herein, we report the design, synthesis, and characterization of novel imide-type prodrugs.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Imides/pharmacology , Prodrugs/pharmacology , Protein Kinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Crystallography, X-Ray , Dogs , Dose-Response Relationship, Drug , Female , Haplorhini , Humans , Imides/administration & dosage , Imides/chemical synthesis , Mice , Models, Molecular , Molecular Structure , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemical synthesis , Rats , Rats, Nude , Solubility , Structure-Activity Relationship , Thermodynamics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
13.
J Med Chem ; 55(7): 3452-78, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22376051

ABSTRACT

To develop RAF/VEGFR2 inhibitors that bind to the inactive DFG-out conformation, we conducted structure-based drug design using the X-ray cocrystal structures of BRAF, starting from an imidazo[1,2-b]pyridazine derivative. We designed various [5,6]-fused bicyclic scaffolds (ring A, 1-6) possessing an anilide group that forms two hydrogen bond interactions with Cys532. Stabilizing the planarity of this anilide and the nitrogen atom on the six-membered ring of the scaffold was critical for enhancing BRAF inhibition. The selected [1,3]thiazolo[5,4-b]pyridine derivative 6d showed potent inhibitory activity in both BRAF and VEGFR2. Solid dispersion formulation of 6d (6d-SD) maximized its oral absorption in rats and showed significant suppression of ERK1/2 phosphorylation in an A375 melanoma xenograft model in rats by single administration. Tumor regression (T/C = -7.0%) in twice-daily repetitive studies at a dose of 50 mg/kg in rats confirmed that 6d is a promising RAF/VEGFR2 inhibitor showing potent anticancer activity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzamides/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Thiazoles/chemical synthesis , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Benzamides/pharmacokinetics , Benzamides/pharmacology , Benzoates/chemical synthesis , Benzoates/pharmacokinetics , Benzoates/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Humans , Imidazoles/chemical synthesis , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Molecular , Molecular Structure , Phosphorylation , Pyridazines/chemical synthesis , Pyridazines/pharmacokinetics , Pyridazines/pharmacology , Rats , Structure-Activity Relationship , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Xenograft Model Antitumor Assays
14.
Genes Cells ; 12(7): 853-61, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17584297

ABSTRACT

The tumor suppressor gene p53 plays a central role in determining cell fate in response to DNA damage; cells may undergo either senescence or apoptosis, depending on cell type. Phosphorylation of Serine 46 (Ser(46)) of p53 is considered to be a primary determinant for the induction of apoptosis, by selectively inducing transactivation of p53 target genes that have proapoptotic function. However, the generality of this mechanism of regulation of p53 remains a matter of debate. We investigated the role of p53 phosphorylation in adriamycin (ADR)-induced apoptosis. We found that Ser(46) was phosphorylated in four different cell lines undergoing ADR-induced senescence, as well as in two different cell lines undergoing ADR-induced apoptosis. Using alanine and glutamic acid substitution mutants of p53 Ser(46), we showed that Ser(46 )phosphorylation is not a prerequisite for induction of the proapoptotic gene AIP1. These results indicate that Ser(46) phosphorylation of p53 is not required for ADR-induced apoptosis.


Subject(s)
Apoptosis , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/physiology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Cellular Senescence , DNA-Binding Proteins/physiology , Doxorubicin/pharmacology , HCT116 Cells , Humans , Nuclear Proteins/physiology , Phosphorylation , Serine/physiology , Transcriptional Activation , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...