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










Publication year range
1.
Int J Mol Sci ; 23(19)2022 Oct 03.
Article in English | MEDLINE | ID: mdl-36233031

ABSTRACT

CXCR4 antagonists have been claimed to reduce mortality after myocardial infarction in myocardial infarction (MI) animals, presumably due to suppressing inflammatory responses caused by myocardial ischemia-reperfusion injury, thus, subsequently facilitating tissue repair and cardiac function recovery. This study aims to determine whether a newly designed CXCR4 antagonist DBPR807 could exert better vascular-protective effects than other clinical counterparts (e.g., AMD3100) to alleviate cardiac damage further exacerbated by reperfusion. Consequently, we find that instead of traditional continuous treatment or multiple-dose treatment at different intervals of time, a single-dose treatment of DBPR807 before reperfusion in MI animals could attenuate inflammation via protecting oxidative stress damage and preserve vascular/capillary density and integrity via mobilizing endothelial progenitor cells, leading to a desirable fibrosis reduction and recovery of cardiac function, as evaluated with the LVEF (left ventricular ejection fraction) in infarcted hearts in rats and mini-pigs, respectively. Thus, it is highly suggested that CXCR4 antagonists should be given at a single high dose prior to reperfusion to provide the maximal cardiac functional improvement. Based on its favorable efficacy and safety profiles indicated in tested animals, DBPR807 has a great potential to serve as an adjunctive medicine for percutaneous coronary intervention (PCI) therapies in acute MI patients.


Subject(s)
Myocardial Infarction , Myocardial Reperfusion Injury , Percutaneous Coronary Intervention , Receptors, CXCR4 , Animals , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/etiology , Rats , Receptors, CXCR4/antagonists & inhibitors , Stroke Volume , Swine , Swine, Miniature , Ventricular Function, Left
2.
J Biomed Sci ; 28(1): 81, 2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34819065

ABSTRACT

BACKGROUND: RAD51-dependent homologous recombination (HR) is one of the most important pathways for repairing DNA double-strand breaks (DSBs), and its regulation is crucial to maintain genome integrity. Elp1 gene encodes IKAP/ELP1, a core subunit of the Elongator complex, which has been implicated in translational regulation. However, how ELP1 contributes to genome maintenance is unclear. METHODS: To investigate the function of Elp1, Elp1-deficient mouse embryonic fibroblasts (MEFs) were generated. Metaphase chromosome spreading, immunofluorescence, and comet assays were used to access chromosome abnormalities and DSB formation. Functional roles of Elp1 in MEFs were evaluated by cell viability, colony forming capacity, and apoptosis assays. HR-dependent DNA repair was assessed by reporter assay, immunofluorescence, and western blot. Polysome profiling was used to evaluate translational efficiency. Differentially expressed proteins and signaling pathways were identified using a label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) proteomics approach. RESULTS: Here, we report that Elp1 depletion enhanced genomic instability, manifested as chromosome breakage and genotoxic stress-induced genomic DNA fragmentation upon ionizing radiation (IR) exposure. Elp1-deficient cells were hypersensitive to DNA damage and exhibited impaired cell proliferation and defective HR repair. Moreover, Elp1 depletion reduced the formation of IR-induced RAD51 foci and decreased RAD51 protein levels. Polysome profiling analysis revealed that ELP1 regulated RAD51 expression by promoting its translation in response to DNA damage. Notably, the requirement for ELP1 in DSB repair could be partially rescued in Elp1-deficient cells by reintroducing RAD51, suggesting that Elp1-mediated HR-directed repair of DSBs is RAD51-dependent. Finally, using proteome analyses, we identified several proteins involved in cancer pathways and DNA damage responses as being differentially expressed upon Elp1 depletion. CONCLUSIONS: Our study uncovered a molecular mechanism underlying Elp1-mediated regulation of HR activity and provides a novel link between translational regulation and genome stability.


Subject(s)
Chromosome Breakage , DNA Damage/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Biosynthesis/genetics , Rad51 Recombinase/genetics , Recombinational DNA Repair/genetics , Animals , Fibroblasts , Genomic Instability , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Rad51 Recombinase/metabolism
3.
Invest New Drugs ; 30(1): 164-75, 2012 Feb.
Article in English | MEDLINE | ID: mdl-20890633

ABSTRACT

Designed from a high throughput screened hit compound, novel 2-amino-1-thiazolyl imidazoles were synthesized and demonstrated cytotoxicity against human cancer cells. 1-(4-Phenylthiazol-2-yl)-4-(thiophen-2-yl)-1H-imidazol-2-amine (compound 2), a 2-amino-1-thiazolyl imidazole, inhibited tubulin polymerization, interacted with the colchicine-binding sites of tubulins, and caused cell cycle arrest at the G(2)/M phase in human gastric cancer cells. Disruption of the microtubule structure in cancer cells by compound 2 was also observed. Compound 2 concentration-dependently inhibited the proliferation of cancer cells in histocultured human gastric and colorectal tumors. Given orally, compound 2 prolonged the lifespans of leukemia mice intraperitoneally inoculated with the murine P388 leukemic cells. We report 2-amino-1-thiazolyl imidazoles as a novel class of orally active microtubule-destabilizing anticancer agents.


Subject(s)
Antineoplastic Agents/administration & dosage , Imidazoles/administration & dosage , Neoplasms, Experimental/drug therapy , Thiazoles/administration & dosage , Tubulin Modulators/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Binding, Competitive , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/metabolism , Dose-Response Relationship, Drug , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Imidazoles/chemical synthesis , Imidazoles/metabolism , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred DBA , Mice, Nude , Microtubules/drug effects , Microtubules/metabolism , Molecular Structure , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/metabolism , Time Factors , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/metabolism
4.
Anticancer Res ; 30(7): 2813-22, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20683017

ABSTRACT

BACKGROUND: Antimitotic tubulin-binding BPR0L075 is structurally analogous to the vascular-disrupting combretastatin A-4. MATERIALS AND METHODS: In vitro/in vivo models of endothelial cells cultures, Matrigel plug assay, tumor-bearing nude mice, and murine leukemia cells-inoculated mice were utilized to evaluate BPR0L075 for antiangiogenic and antitumoral activity spectra. RESULTS: BPR0L075 concentration-dependently inhibited proliferation and migration of human umbilical vein endothelial cells (HUVECs), disrupted capillary tube formations of HUVECs and rat aorta endothelial cells, and suppressed in vivo VEGF-mediated angiogenesis in Matrigel plugs in mice. Besides inhibiting the colony growth of cancer cells, BPR0L075 suppressed growth of subcutaneously-xenografted human lung, colorectal, and cervical solid tumors in nude mice. Combination treatments of BPR0L075 plus cisplatin, compared to either agent alone, demonstrated a stronger growth inhibition against the tumor xenografts in nude mice and longer lifespan in the leukemia mice. CONCLUSION: BPR0L075 is an antitumoral and antiangiogenic agent and potentiates the anticancer activity of cisplatin.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Indoles/pharmacology , Neoplasms/drug therapy , Angiogenesis Inhibitors/administration & dosage , Animals , Cell Growth Processes/drug effects , Cisplatin/administration & dosage , Drug Screening Assays, Antitumor , Drug Synergism , Endothelial Cells/drug effects , Female , HCT116 Cells , Humans , Indoles/administration & dosage , Leukemia P388/drug therapy , Male , Mice , Mice, Inbred DBA , Mice, Nude , Neoplasms/pathology , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
5.
J Med Chem ; 53(6): 2409-17, 2010 Mar 25.
Article in English | MEDLINE | ID: mdl-20170097

ABSTRACT

2-Amino-1-arylidenaminoimidazoles, a novel class of orally (po) active microtubule-destabilizing anticancer agents, were synthesized. The compounds were designed from a hit compound identified in a drug discovery platform by using cancer cell-based high throughput screening assay. Selective synthesized compounds exerted cell cytotoxicity against human cancer cells. The underlying mechanisms for the anticancer activity were demonstrated as interacting with the tubulins and inhibiting microtubule assembly, leading to proliferation inhibition and apoptosis induction in the human tumor cells. Furthermore, two compounds showed in vivo anticancer activities in both po and intravenously (iv) administered routes and prolonged the life spans of murine leukemic P388 cells-inoculated mice. These new po active antimitotic anticancer agents are to be further examined in preclinical studies and developed for clinical uses.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Neoplasms/drug therapy , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Female , Humans , Imidazoles/chemistry , Inhibitory Concentration 50 , Leukemia P388/drug therapy , Leukemia P388/pathology , Mice , Mice, Inbred DBA , Mice, Nude , Microtubules/drug effects , Microtubules/metabolism , Models, Chemical , Molecular Structure , Neoplasms/pathology , Structure-Activity Relationship , Tubulin/metabolism , Xenograft Model Antitumor Assays
6.
Biochem Pharmacol ; 78(2): 203-10, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19439267

ABSTRACT

DPP-IV (EC 3.4.14.5) is a validated drug target for human type II diabetes. DPP-IV inhibitors without DPP8/9 inhibitory activity have been sought because a possible association has been reported between a "DPP8/9 inhibitor" and severe toxicity in animals. However, at present, it is not known whether the observed toxicity is associated with DPP8/9 inhibition, or an off-target effect induced by the compound. We investigated whether the inhibition of DPP8/9 is the cause of the severe toxicity in animals using a very potent and selective DPP8/9 inhibitor with different pharmacophore, 1G244. By Ki measurement, 1G244 is 15- and 8-fold more potent against DPP8 and DPP9, respectively, than the "DPP8/9 inhibitor". Strikingly, the "DPP8/9 inhibitor" does not penetrate the plasma membrane but remains outside the cells, whereas 1G244 readily enters the cells, even at low doses. By repeatedly exposing Sprague-Dawley rats to 1G244 by intravenous injection for a period of 14 days, we observed no significant toxicological symptoms associated with 1G244. Blood and serum chemistry parameters were all within the normal ranges for the treated animals. Because of the high potency, good membrane penetration and adequate tissue distribution of 1G244, the mild symptoms observed are probably associated with DPP8/9 inhibition.


Subject(s)
Dipeptidases/antagonists & inhibitors , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/toxicity , Animals , Cell Line , Dipeptidases/blood , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/blood , Female , Humans , Male , Protease Inhibitors/chemistry , Random Allocation , Rats , Rats, Sprague-Dawley
7.
Anal Biochem ; 375(1): 115-23, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18190777

ABSTRACT

Although several cell-based reporter assays have been developed for screening of viral protease inhibitors, most of these assays have a significant limitation in that numerous false positives can be generated for the compounds that are interfering with reporter gene detection due to the cellular viability. To improve, we developed a mammalian cell-based assay based on the reverse two-hybrid system to monitor the proteolytic activity of human enterovirus 71 (EV71) 3C protease and to validate the cytotoxicity of compounds at the same time. In this system, the GAL4 DNA binding domain (M3) and transactivation domain (VP16) were fused, in-frame, with 3C or 3C(mut). The 3C(mut) was an inactivated protease with mutations at the predicted catalytic triad. The reporter plasmid contains a secreted alkaline phosphatase (SEAP) gene under the control of GAL4 activating sequences. We demonstrated that M3-3C-VP16 failed to turn on the expression of SEAP due to the separation of M3 and the VP16 domains by self-cleavage of 3C. In contrast, SEAP expression was induced by the M3-3C(mut)-VP16 fusion protein or the M3-3C-VP16 in cells treated with AG7088, a potent inhibitor of human rhinoviruses (HRVs) 3C protease. Potentially, this protease detection system should greatly facilitate anti-EV71 drug discovery through a high-throughput screening.


Subject(s)
Endopeptidases/metabolism , Enterovirus A, Human/enzymology , Two-Hybrid System Techniques , Viral Proteins/metabolism , Animals , COS Cells , Cell-Free System , Chlorocebus aethiops , Genes, Reporter , Herpes Simplex Virus Protein Vmw65/metabolism , Mutant Proteins/metabolism , Protease Inhibitors/pharmacology , Recombinant Fusion Proteins/metabolism , Trans-Activators/metabolism
8.
Mol Pharmacol ; 69(3): 749-58, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16332992

ABSTRACT

The molecular mechanisms for the anti-inflammatory activity of phenanthroindolizidine alkaloids were examined in an in vitro system mimicking acute inflammation by studying the suppression of lipopolysaccharide (LPS)/interferon-gamma (IFNgamma)-induced nitric oxide production in RAW264.7 cells. Two of the phenanthroindolizidine alkaloids, NSTP0G01 (tylophorine) and NSTP0G07 (ficuseptine-A), exhibited potent suppression of nitric oxide production and did not show significant cytotoxicity to the LPS/IFNgamma-stimulated RAW264.7 cells, in contrast to their respective cytotoxic effects on cancer cells. Tylophorine was studied further to investigate the responsible mechanisms. It was found to inhibit the induced protein levels of tumor necrosis factor-alpha, inducible nitric-oxide synthase (iNOS), and cyclooxygenase (COX)-II. It also inhibited the activation of murine iNOS and COX-II promoter activity. However, of the two common responsive elements of iNOS and COX-II promoters, nuclear factor-kappaB (NF-kappaB) and adaptor protein (AP)1, only AP1 activation was inhibited by tylophorine in the LPS/IFNgamma-stimulated RAW264.7 cells. Further studies showed that the tylophorine enhanced the phosphorylation of Akt and thus decreased the expression and phosphorylation levels of c-Jun protein, thereby causing the subsequent inhibition of AP1 activity. Furthermore, the tylophorine was able to block mitogen-activated protein/extracellular signal-regulated kinase kinase 1 activity and its downstream signaling activation of NF-kappaB and AP1. Thus, NSTP0G01 exerts its anti-inflammatory effects by inhibiting expression of the proinflammatory factors and related signaling pathways.


Subject(s)
Alkaloids/pharmacology , Anti-Inflammatory Agents/pharmacology , Ficus/chemistry , Indolizines/pharmacology , Nitric Oxide/metabolism , Phenanthrenes/pharmacology , Adaptor Protein Complex 1/antagonists & inhibitors , Animals , Cells, Cultured , Chromones/pharmacology , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Morpholines/pharmacology , NF-kappa B/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Phosphorylation , Promoter Regions, Genetic/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Response Elements/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors
9.
Bioorg Med Chem Lett ; 15(8): 2027-32, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15808462

ABSTRACT

A series of benzothiazolium compounds were identified as novel classes of inhibitors of nitric oxide production in a cell culture system. They exhibited approximately 1600 folds potency with IC(50) at approximately 50nM to several microM as compared to IC(50) 88.4microM of l-NMMA, a known inhibitor of nitric oxide synthase. The mechanistic studies suggest that decreased iNOS protein synthesis and mRNA transcription, at least in part, were related to the inhibitory activity of effective benzothiazolium compounds. The correlation of in vivo and in vitro activities using mouse paw edema model was also demonstrated.


Subject(s)
Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Nitric Oxide/antagonists & inhibitors , Thiazoles/chemistry , Animals , Benzothiazoles , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Interferon-gamma/antagonists & inhibitors , Lipopolysaccharides/antagonists & inhibitors , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Thiazoles/pharmacology
10.
J Med Chem ; 47(17): 4247-57, 2004 Aug 12.
Article in English | MEDLINE | ID: mdl-15293996

ABSTRACT

The synthesis and study of the structure-activity relationships of two new classes of synthetic antitubulin compounds based on 1-aroylindole and 3-aroylindole skeletons are described. Lead compounds 3, 10, and 14 displayed potent cytotoxicities with IC50 = 0.9-26 nM against human NUGC3 stomach, MKN45 stomach, MESSA uterine, A549 lung, and MCF-7 breast carcinoma cell lines. The inhibition of proliferation correlated with in vitro polymerization inhibitory activities. Structure-activity relationships revealed that 6-methoxy substitution of 3-aroylindoles and 5-methoxy substitution of 1-aroylindoles contribute to a significant extent for maximal activity by mimicking the para substitution of the methoxy group to the carbonyl group in the case of aminobenzophenones. Addition of a methyl group at the C-2 position on the indole ring exerts an increased potency. The 3,4,5-trimethoxybenzoyl moiety was necessary for better activity but not essential and can be replaced by 3,5-dimethoxybenzoyl and 3,4,5-trimethoxybenzyl moieties. We conclude that 1- and 3-aroylindoles constitute an interesting new class of antitubulin agents with the potential to be clinically developed for cancer treatment.


Subject(s)
Antineoplastic Agents/chemical synthesis , Indoles/chemical synthesis , Stilbenes/chemical synthesis , Tubulin Modulators , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biopolymers/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , Indoles/chemistry , Indoles/pharmacology , Molecular Structure , Stilbenes/chemistry , Stilbenes/pharmacology , Structure-Activity Relationship , Tubulin/chemistry
11.
Bioorg Med Chem Lett ; 14(5): 1169-72, 2004 Mar 08.
Article in English | MEDLINE | ID: mdl-14980658

ABSTRACT

A series of 5-substituted 2-cyanoimino-4-imidazodinone and 2-cyanoimino-4-pyrimidinone derivatives were synthesized and their anticancer cytotoxicity were evaluated in in vitro assay. It was found that the bulky aryl functionality in the 5-position of the 2-cyanoimino-4-imidazolidinone compounds was essential for the cytotoxicity of these heterocyclic compounds. Some of the derivatives exhibited modest cytotoxicity against a variety of cancer cell lines. One of the derivatives, [1-[6-(4-chlorophenoxy)hexyl]-5-oxo-4-phenyl-3-(4-pyridyl)tetrahydro-1H-2-imidazolyliden]aminomethanenitrile (Compound 11), exhibited the most potent cytotoxic activity with IC(50) in the nanomolar range. The cytotoxicity of these derivatives was selection with no apparent toxic effect toward normal fibroblasts.


Subject(s)
Imidazoles/chemical synthesis , Imidazoles/toxicity , Pyrimidinones/chemical synthesis , Pyrimidinones/toxicity , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Line, Tumor , Fibroblasts/drug effects , Humans
12.
J Med Chem ; 46(9): 1706-15, 2003 Apr 24.
Article in English | MEDLINE | ID: mdl-12699388

ABSTRACT

A series of N-heterocyclic indolyl glyoxylamides were synthesized and evaluated for in vitro and in vivo anticancer activities. They exhibited a broad spectrum of anticancer activity not only in murine leukemic cancer cells but also in human gastric, breast, and uterus cancer cells as well as their multidrug resistant sublines with a wide range of IC(50) values. They also induced apoptosis and caused DNA fragmentation in human gastric cancer cells. Among the compounds studied, 7 showed the most potent activity of growth inhibition (IC(50) = 17-1711 nM) in several human cancer cells. Given orally, compounds 7 and 13 dose-dependently prolonged the survival of animals inoculated with P388 leukemic cancer cells. N-Heterocyclic indolyl glyoxylamides may be useful as orally active chemotherapeutic agents against cancer and refractory cancerous diseases of multidrug resistance phenotype.


Subject(s)
Antineoplastic Agents/chemical synthesis , Glyoxylates/chemical synthesis , Indoles/chemical synthesis , Thiazoles/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Cell Division/drug effects , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Glyoxylates/chemistry , Glyoxylates/pharmacology , Humans , Indoles/chemistry , Indoles/pharmacology , Leukemia P388/mortality , Mice , Mice, Inbred DBA , Neoplasm Transplantation , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/pharmacology , Tumor Cells, Cultured
13.
Bioorg Med Chem Lett ; 12(19): 2729-32, 2002 Oct 07.
Article in English | MEDLINE | ID: mdl-12217364

ABSTRACT

Novel vitamin K(3) analogues were synthesized and evaluated for their anticancer activity. Compound 6, 9, 10, 11, 14, and (+/-)15 demonstrated a strong inhibitory activity against the tumor cells of A-549, Hep G2, MCF7, MES-SA, MES-SA/Dx5, MKN45, SW-480, and TW-039. Compound (+/-)15 displayed potent tumor cell cytotoxicity, and compound 14 selectively affected MCF7, even though it did not influence normal cells Detroit551 and WI-38. Compound (+/-)15 inhibited MES-SA and MES-SA/Dx5, and this specific result shows that compound (+/-)15 may become a good anticancer drug candidate.


Subject(s)
Antineoplastic Agents/pharmacology , Vitamin K 3/analogs & derivatives , Vitamin K 3/pharmacology , Antineoplastic Agents/chemical synthesis , Genes, MDR , Humans , Indicators and Reagents , Tumor Cells, Cultured , Vitamin K 3/chemical synthesis
14.
J Med Chem ; 45(12): 2556-62, 2002 Jun 06.
Article in English | MEDLINE | ID: mdl-12036364

ABSTRACT

A new type of inhibitor of tubulin polymerization was discovered on the basis of the combretastatin molecular skeleton. The lead compounds in this series, compounds 6 and 7, strongly inhibited tubulin polymerization in vitro and significantly arrested cells at the G(2)/M phase. Compounds 6 and 7 yielded 50- to 100-fold lower IC(50) values than did combretastatin A-4 against Colo 205, NUGC3, and HA22T human cancer cell lines as well as similar or greater growth inhibitory activities than did combretastain A-4 against DLD-1, HR, MCF-7, DU145, HONE-1, and MES-SA/DX5 human cancer cell lines. Structure-activity relationship information revealed that introduction of an amino group at the ortho position of the benzophenone ring plays an integral role for increased growth inhibition.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzophenones/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzophenones/chemistry , Benzophenones/pharmacology , Binding, Competitive , Biopolymers , Colchicine/chemistry , Drug Screening Assays, Antitumor , Humans , Mitosis/drug effects , Solubility , Structure-Activity Relationship , Tubulin/chemistry , Tumor Cells, Cultured
15.
J Med Chem ; 45(8): 1644-55, 2002 Apr 11.
Article in English | MEDLINE | ID: mdl-11931618

ABSTRACT

When skeletons of Win compounds were used as templates, computer-assisted drug design led to the identification of a novel series of imidazolidinone derivatives with significant antiviral activity against enterovirus 71 (EV 71), the infection of which had resulted in about 80 fatalities during the 1998 epidemic outbreak in Taiwan. In addition to inhibiting all the genotypes (A, B, and C) of EV 71 in the submicromolar to low micromolar range, compounds 1 and 8 were extensively evaluated against a variety of viruses, showing potent activity against coxsackievirus A9 (IC(50) = 0.47-0.55 microM) and coxsackievirus A24 (IC(50) = 0.47-0.55 microM) as well as moderate activity against enterovirus 68 (IC(50) = 2.13 microM) and echovirus 9 (IC(50) = 2.6 microM). Our SAR studies revealed that imidazolidinone analogues with an aryl substituent at the para position of the phenoxyl ring, such as compounds 20, 21, 27, 57, 58, and 61, in general exhibited the highest activity against EV 71. Among them, compound 20 and its corresponding hydrochloride salt 57, in terms of potency and selectivity index, appear to be the most promising candidates in this series for further development of anti-EV-71 agents. Preliminary results of the study on the mode of action by a time-course experiment suggest that test compounds 1 and 8 can effectively inhibit the virus replication at the early stages, referring to virus attachment or uncoating. This indicates that the surface protein may be the target for this type of compounds.


Subject(s)
Antiviral Agents/chemical synthesis , Enterovirus/drug effects , Imidazoles/chemical synthesis , Pyridines/chemical synthesis , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Neutralization Tests , Oxadiazoles/chemistry , Oxazoles , Pyridines/chemistry , Pyridines/pharmacology , Structure-Activity Relationship , Viral Plaque Assay
SELECTION OF CITATIONS
SEARCH DETAIL