Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Antimicrob Chemother ; 76(11): 2867-2874, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34383913

ABSTRACT

OBJECTIVES: FtsZ is an essential bacterial protein and an unexplored target for the development of antibacterial drugs. The development of a novel inhibitor targeting FtsZ offers a potential opportunity to combat drug resistance. DS01750413, a new derivative of PC190723, is a novel FtsZ inhibitor with improved in vitro and in vivo activity. The objective of this study was to investigate the efficacy of DS01750413 against Staphylococcus spp., including MRSA, in in vitro and in vivo models. METHODS: In vitro activities of DS01750413 and standard-of-care antibiotics were evaluated against clinical isolates of Gram-positive pathogens. The in vivo efficacy was evaluated in a murine systemic infection model caused by MRSA. RESULTS: DS01750413 showed potent in vitro activity against MRSA clinical isolates with MIC ranges of 0.5-1 mg/L and also demonstrated concentration-dependent bactericidal killing. In the murine bacteraemia infection model of MRSA, treatment with DS01750413 resulted in prolonged survival of animals compared with placebo-treated animals and exhibited a significant reduction in the bacterial load in liver, spleen, lungs and kidneys. CONCLUSIONS: DS01750413 showed encouraging in vitro and in vivo activity against MRSA. As a novel chemical class, DS01750413 has the potential to become clinically viable antibiotics to address the drug resistance problem by its unique novel targeting mechanism of action.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cytoskeletal Proteins , Mice , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy , Staphylococcus aureus
2.
Curr Drug Deliv ; 17(4): 343-347, 2020.
Article in English | MEDLINE | ID: mdl-32077827

ABSTRACT

BACKGROUND: Rifampicin is known to degrade at the acidic pH of the stomach, especially in the presence of isoniazid. Although isoniazid also degrades partially, its degradation is reversible. OBJECTIVE: Presently, we provide a proof of the fact that the simultaneous oral administration of rifampicin (RIF), upon incorporation into solid lipid nanoparticles (RIF-SLNs), with isoniazid (INH) overcomes its INH-induced degradation and improves its oral bioavailability in rats. METHODS: Solid lipid nanoparticles of RIF (RIF-SLNs) were prepared using a novel and patented method. The effect of INH was investigated on in vivo bioavailability of RIF both in its free and encapsulated (RIF-SLNs) form, after oral administration to rats. RESULTS: Cmax and AUC0-∞ of RIF increased 158 % and 125 %, respectively, upon incorporation into SLNs versus free RIF when combined with INH. The Tmax decreased from 5.67 h to 3.3 h, and the plasma concentration of RIF remained above its MIC (8 µg/ml) at all the tested time points starting with 15 min, when administered as RIF-SLNs in combination with INH. CONCLUSION: The results confirm the scope of combining RIF-SLNs with INH to overcome the bioavailability of free RIF when combined with INH, especially in fixed dose combinations.


Subject(s)
Isoniazid/pharmacokinetics , Lipids/pharmacokinetics , Nanoparticles/chemistry , Rifampin/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Capsules/administration & dosage , Capsules/chemistry , Capsules/pharmacokinetics , Isoniazid/administration & dosage , Isoniazid/blood , Lipids/administration & dosage , Lipids/blood , Male , Nanoparticles/administration & dosage , Rats , Rats, Wistar , Rifampin/administration & dosage , Rifampin/blood
3.
Article in English | MEDLINE | ID: mdl-30670430

ABSTRACT

DS86760016 is a new leucyl-tRNA-synthetase inhibitor at the preclinical development stage. DS86760016 showed potent activity against extended-spectrum multidrug-resistant Pseudomonas aeruginosa isolated from clinical samples and in vitro biofilms. In a murine catheter-associated urinary tract infection model, DS86760016 treatment resulted in significant eradication of P. aeruginosa from the kidney, bladder, and catheter without developing drug resistance. Our data suggest that DS86760016 has the potential to act as a new drug for the treatment of Pseudomonas infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Boron Compounds/pharmacology , Catheter-Related Infections/drug therapy , Dioxoles/pharmacology , Leucine-tRNA Ligase/antagonists & inhibitors , Methylamines/pharmacology , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Urinary Tract Infections/drug therapy , Animals , Anti-Bacterial Agents/pharmacokinetics , Biofilms/growth & development , Boron Compounds/pharmacokinetics , Catheter-Related Infections/microbiology , Dioxoles/pharmacokinetics , Disease Models, Animal , Drug Resistance, Multiple, Bacterial , Female , Humans , Methylamines/pharmacokinetics , Mice , Microbial Sensitivity Tests , Pseudomonas Infections/microbiology , Urinary Tract Infections/microbiology
4.
Bioorg Med Chem Lett ; 28(17): 2993-2997, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30017316

ABSTRACT

FimH is a type I fimbrial lectin located at the tip of type-1 pili of Gram-negative uropathogenic Escherichia coli (UPEC) guiding its ability to adhere and infect urothelial cells. Accordingly, blocking FimH with small molecule inhibitor is considered as a promising new therapeutic alternative to treat urinary tract infections caused by UPEC. Herein, we report that compounds having the S-glycosidic bond (thiomannosides) had improved metabolic stability and plasma exposures when dosed orally. Especially compound 5h showed the potential to inhibit biofilm formation and also to disrupt the preformed biofilm. And compound 5h showed prophylactic effect in UTI model in mice.


Subject(s)
Fimbriae Proteins/antagonists & inhibitors , Mannosides/pharmacology , Urinary Tract Infections/drug therapy , Adhesins, Escherichia coli/metabolism , Administration, Oral , Animals , Biofilms/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Fimbriae Proteins/metabolism , Mannosides/administration & dosage , Mannosides/chemistry , Mice , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Urinary Tract Infections/urine
5.
Article in English | MEDLINE | ID: mdl-29437618

ABSTRACT

The emergence of multidrug-resistant (MDR) Gram-negative bacilli is a major concern in the treatment of nosocomial infections. Antibacterial agents with novel modes of action can be useful, as these pathogens have become resistant to almost all existing standard-of-care agents. GSK2251052, a leucyl-tRNA synthetase inhibitor, has a novel mode of action against Gram-negative bacteria. However, the phase 2 studies with this drug were terminated due to microbiological failures based on the rapid emergence of drug resistance during the treatment of complicated urinary tract infections. DS86760016 is a novel leucyl-tRNA synthetase inhibitor active against MDR Gram-negative bacteria, such as Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa, with an improved pharmacokinetic profile. DS86760016 showed lower plasma clearance, longer plasma half-life, and higher renal excretion than GSK2251052 did in mice, rats, monkeys and dogs. DS86760016 also showed lower mutant prevention concentrations against P. aeruginosa than did GSK2251052. No resistant bacteria were observed in murine urinary tract infection models at a dose that maintained urinary concentrations above the mutant prevention concentration. DS86760016 also showed a lower risk of resistance development than did GSK2251052 in comparative in vivo studies with murine urinary tract infection models. These results suggest that DS86760016 has potential as a new drug for the treatment of MDR Gram-negative bacterial infections, with a lower risk of drug resistance development than that of GSK2251052.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/pathogenicity , Gram-Negative Bacterial Infections/drug therapy , Leucine-tRNA Ligase/antagonists & inhibitors , Animals , Boron Compounds/therapeutic use , Drug Resistance, Multiple, Bacterial/genetics , Female , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/enzymology , Klebsiella pneumoniae/pathogenicity , Leucine-tRNA Ligase/metabolism , Macaca fascicularis , Male , Mice , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/pathogenicity , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology
6.
Chem Biol Interact ; 240: 146-52, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26282489

ABSTRACT

There has been a search for new anticancer agents to treat cancer resistance throughout the globe. Salinomycin (SAL), a broad spectrum antibiotic and a coccidiostat has been found to counter tumour resistance and kill cancer stem cells with better efficacy than the existing chemotherapeutic agents; paclitaxel and doxorubicin. This refocused its importance for treatment of human cancers. In this study, we studied the in vitro drug metabolism and pharmacokinetic parameters of SAL. SAL undergoes rapid metabolism in liver microsomes and has a high intrinsic clearance. SAL metabolism is mainly mediated by CYP enzymes; CYP3A4 the major enzyme metabolising SAL. The percent plasma protein binding of SAL in human was significantly lower as compared to mouse and rat plasma. CYP inhibition was carried out by chemical inhibition and recombinant enzyme studies. SAL was found to be a moderate inhibitor of CYP2D6 as well as CYP3A4. As CYP3A4 was the major enzyme responsible for metabolism of SAL, in vivo pharmacokinetic study in rats was done to check the effect of concomitant administration of Ketoconazole (KTC) on SAL pharmacokinetics. KTC, being a selective CYP3A4 inhibitor increased the systemic exposure of SAL significantly to 7-fold in AUC0-α and 3-fold increase in Cmax of SAL in rats with concomitant KTC administration.


Subject(s)
Drug Delivery Systems , Neoplastic Stem Cells/drug effects , Pyrans/pharmacology , Pyrans/pharmacokinetics , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Male , Mice , Microsomes, Liver/drug effects , Pyrans/chemistry , Rats , Rats, Sprague-Dawley
7.
Analyst ; 138(20): 6106-16, 2013 Oct 21.
Article in English | MEDLINE | ID: mdl-23964358

ABSTRACT

The aim of the present study is to develop and demonstrate the correlation between in vitro and in vivo Plasma Protein Binding (PPB) using the ultracentrifugation method for its validation by using marketed compounds like atenolol, theophylline and phenytoin. In this study, in vitro PPB is carried out using ultracentrifugation, by spiking the selected marketed compounds at concentrations of 5 and 15 µM in plasma. In an in vivo study, rats (n = 3) were given a single oral dose (10 mg kg(-1)) and post-dose samples were subjected to ultracentrifugation to obtain the protein-free fraction. A rapid and highly sensitive method was developed and validated for determining the free fraction of marketed compounds in rat plasma using protein precipitation and analysis using an ultra performance liquid chromatography electrospray ionization (ESI) tandem mass spectrometer system (UPLC-MS/MS). The in vitro free fraction (fup) values were 0.93 ± 0.07 for atenolol, 0.31 ± 0.03 for theophylline and 0.09 ± 0.02 for phenytoin which correlated well with the corresponding in vivo values of 0.91 ± 0.03 for atenolol, 0.25 ± 0.02 for theophylline and 0.09 ± 0.01 for phenytoin with a coefficient of variation less than 11.06%, 11.45% and 13.67%, respectively. Therefore the validated high-throughput in vitro PPB study is expected to have a powerful impact on reducing the cost as well as time in the drug discovery process.


Subject(s)
Blood Proteins/analysis , Blood Proteins/metabolism , Tandem Mass Spectrometry/methods , Animals , Chromatography, High Pressure Liquid/methods , Chromatography, Liquid/methods , Male , Protein Binding/physiology , Rats , Rats, Wistar , Ultracentrifugation/methods
8.
Bioorg Med Chem Lett ; 20(15): 4587-92, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20594842
9.
Bioorg Med Chem Lett ; 19(15): 4350-3, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19515564

ABSTRACT

The synthesis and optimisation of HCV NS5B polymerase inhibitors with improved potency versus the existing compound 1 is described. Substitution in the benzothiadiazine portion of the molecule, furnishing improvement in potency in the high protein Replicon assay, is highlighted, culminating in the discovery of 12h, a highly potent oxyacetamide derivative.


Subject(s)
Antiviral Agents/chemical synthesis , Benzothiadiazines/chemistry , Chemistry, Pharmaceutical/methods , Hepacivirus/enzymology , Viral Nonstructural Proteins/antagonists & inhibitors , Administration, Oral , Animals , Antiviral Agents/pharmacology , Benzothiadiazines/pharmacology , Drug Design , Humans , Inhibitory Concentration 50 , Models, Chemical , Molecular Conformation , Molecular Structure , Rats , Structure-Activity Relationship
10.
J Med Chem ; 52(7): 1828-44, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-19284751

ABSTRACT

On the basis of a mechanistic understanding of the toxicity of the 4-aminoquinoline amodiaquine (1b), three series of amodiaquine analogues have been prepared where the 4-aminophenol "metabolic alert" has been modified by replacement of the 4'-hydroxy group with a hydrogen, fluorine, or chlorine atom. Following antimalarial assessment and studies on mechanism of action, two candidates were selected for detailed ADME studies and in vitro and in vivo toxicological assessment. 4'-Fluoro-N-tert-butylamodiaquine (2k) was subsequently identified as a candidate for further development studies based on potent activity versus chloroquine-sensitive and resistant parasites, moderate to excellent oral bioavailability, low toxicity in in vitro studies, and an acceptable safety profile.


Subject(s)
Aminoquinolines/chemical synthesis , Amodiaquine/analogs & derivatives , Amodiaquine/chemical synthesis , Antimalarials/chemical synthesis , Aminoquinolines/pharmacokinetics , Aminoquinolines/pharmacology , Amodiaquine/chemistry , Amodiaquine/pharmacokinetics , Amodiaquine/pharmacology , Animals , Antimalarials/pharmacokinetics , Antimalarials/pharmacology , Cell Survival , Chloroquine/pharmacology , Dogs , Drug Resistance , Female , Haplorhini , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , In Vitro Techniques , Malaria/drug therapy , Malaria/parasitology , Male , Mice , Parasitic Sensitivity Tests , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Plasmodium yoelii/drug effects , Rats , Rats, Wistar , Structure-Activity Relationship
11.
12.
J Med Chem ; 52(5): 1408-15, 2009 Mar 12.
Article in English | MEDLINE | ID: mdl-19222165

ABSTRACT

N-tert-Butyl isoquine (4) (GSK369796) is a 4-aminoquinoline drug candidate selected and developed as part of a public-private partnership between academics at Liverpool, MMV, and GSK pharmaceuticals. This molecule was rationally designed based on chemical, toxicological, pharmacokinetic, and pharmacodynamic considerations and was selected based on excellent activity against Plasmodium falciparum in vitro and rodent malaria parasites in vivo. The optimized chemistry delivered this novel synthetic quinoline in a two-step procedure from cheap and readily available starting materials. The molecule has a full industry standard preclinical development program allowing first into humans to proceed. Employing chloroquine (1) and amodiaquine (2) as comparator molecules in the preclinical plan, the first preclinical dossier of pharmacokinetic, toxicity, and safety pharmacology has also been established for the 4-aminoquinoline antimalarial class. These studies have revealed preclinical liabilities that have never translated into the human experience. This has resulted in the availability of critical information to other drug development teams interested in developing antimalarials within this class.


Subject(s)
Aminoquinolines/pharmacology , Antimalarials/pharmacology , Benzylamines/pharmacology , Aminoquinolines/chemical synthesis , Aminoquinolines/chemistry , Aminoquinolines/pharmacokinetics , Aminoquinolines/toxicity , Amodiaquine/analogs & derivatives , Animals , Antimalarials/chemical synthesis , Antimalarials/pharmacokinetics , Antimalarials/toxicity , Benzylamines/chemical synthesis , Benzylamines/chemistry , Benzylamines/toxicity , Cytochrome P-450 Enzyme Inhibitors , Dogs , Drug Evaluation, Preclinical , Drug Resistance , Female , Haplorhini , Heme/chemistry , Humans , Malaria/drug therapy , Mice , Models, Molecular , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Plasmodium yoelii , Rats , Structure-Activity Relationship
13.
J Pharm Sci ; 98(1): 362-77, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18563832

ABSTRACT

The disposition of three 4-aminoquinoline leads, namely isoquine (ISO), des-ethyl isoquine (DEI) and N-tert-butyl isoquine (NTBI), were studied in a range of in vivo and in vitro assays to assist in selecting an appropriate candidate for further development. Analogous to amodiaquine (ADQ), ISO undergoes oxidative N-dealkylation to form DEI in vivo. Blood clearance of DEI was as much as 10-fold lower than that of ISO in animals and after oral administration, metabolite exposure exceeded that of parent by as much as 14-fold. Replacement of the N-ethyl with an N-tert-butyl substituent substantially reduced N-dealkylation as blood clearance of NTBI was approximately 2 to 3-fold lower than DEI in mouse, rat, dog and monkey. Mean NTBI oral bioavailability was generally higher than the other leads (>/=68%). Blood cell association was substantial for NTBI, particularly in dog and monkey, where blood to plasma concentration ratios >4 were observed. Human plasma protein binding was similar for NTBI, DEI, and des-ethyl amodiaquine (DEA). Allometric scaling predicted human blood clearance (CL) for NTBI to be low ( approximately 12% liver blood flow). All the 4-aminoquinolines inhibited recombinant human cytochrome P450 2D6 with similar potency; DEI also inhibited 1A2. On balance, NTBI appeared the most promising lead to progress towards full development.


Subject(s)
Aminoquinolines/pharmacokinetics , Antimalarials/pharmacokinetics , Aminoquinolines/blood , Aminoquinolines/metabolism , Animals , Antimalarials/blood , Antimalarials/metabolism , Biological Availability , Dogs , Drug Evaluation, Preclinical/methods , Female , Humans , Macaca fascicularis , Male , Mice , Rats , Rats, Sprague-Dawley , Species Specificity
14.
Drug Metab Dispos ; 36(9): 1846-52, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18556439

ABSTRACT

The pharmacokinetics and brain penetration of the novel neurokinin (NK)-1 receptor antagonist casopitant [1-piperidinecarboxamide, 4-(4-acetyl-1-piperazinyl)-N-((1R)-1-(3,5-bis(trifluoromethyl)phenyl)ethyl)-2-(4-fluoro-2-methylphenyl)-N-methyl-, (2R,4S)-; GW679769] were examined in ferrets. The ferret is known to respond to the full spectrum of agents recognized to induce emesis in humans, and the cisplatin-induced emesis models in the ferret have been used to establish the antiemetic potential of casopitant. Following single i.p. dosing to the ferret, casopitant was rapidly absorbed, with plasma and brain concentrations being approximately equal at 2 h postdose. The predominant radioactive component present in the ferret brain after a single dose of [(14)C]casopitant was parent compound, accounting for approximately 76% of the radioactivity. The major metabolites present in brain tissue following administration of [(14)C]casopitant were hydroxylated casopitant (M1) and the corresponding ketone product of the M1 metabolite (M2), which accounted for approximately 19 and 3% of the radioactivity in the brain extracts, respectively. All three molecules had relatively similar potency against ferret brain cortical NK-1, suggesting that the pharmacologic activity of casopitant in the ferret is largely attributable to parent compound and, to a lesser extent, to its oxidative metabolites. Because casopitant is intended to be administered in combination with ondansetron and because therapeutic synergy has been observed with this combination in the ferret, a drug interaction study was conducted. The additional pharmacodynamic benefit of the combination dose was not because of an alteration in the pharmacokinetics of either agent but is likely the result of the complementary mechanisms of pharmacologic action of the two drugs.


Subject(s)
Brain/metabolism , Neurokinin-1 Receptor Antagonists , Piperazines/pharmacokinetics , Piperidines/pharmacokinetics , Animals , Area Under Curve , Biotransformation , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Ferrets , Male , Piperazines/metabolism , Piperazines/pharmacology , Piperidines/metabolism , Piperidines/pharmacology , Tandem Mass Spectrometry
15.
J Pharm Sci ; 95(12): 2657-72, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16892205

ABSTRACT

GW844520 is a potent and selective inhibitor of the cytochrome bc1 complex of mitochondrial electron transport in P. falciparum, the parasite primarily responsible for the mortality associated with malaria worldwide. GW844520 is fully active against the parasite including resistance isolates, showing no cross resistance with agents in use. To evaluate full potential of this development candidate, we conducted drug metabolism and pharmacokinetic studies of this novel anti-malarial. GW844520 had low blood clearance of about 0.5-4% of hepatic blood flow and a steady-state volume of distribution of 2-4 times total body water in mouse, rat, dog, and monkey. Oral bioavailability was high (51-100%). Consistent with the in vivo data, GW844520 had low intrinsic clearance in liver microsomes and hepatocytes of animal and human origin, high passive cellular permeability and was not a P-glycoprotein substrate. GW844520 did not associate appreciably with blood cells but was highly bound to plasma proteins (>99%) in all species. GW844520 was a substrate and inhibitor of human CYP2D6 but not of CYP1A2, 2C9, 2C19, and 3A4. This conjunctive analysis supports continued evaluation of this compound in definitive pre-IND studies and exemplifies our strategy supporting the discovery of novel agents to treat diseases of the developing world.


Subject(s)
Antimalarials/pharmacokinetics , Pyridones/pharmacokinetics , Animals , Antimalarials/blood , Biological Transport , Biotransformation , Cell Line , Cell Membrane Permeability , Cytochrome P-450 Enzyme Inhibitors , Dogs , Drug Evaluation, Preclinical , Electron Transport Complex III/antagonists & inhibitors , Female , Hepatocytes/metabolism , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred Strains , Microsomes, Liver/metabolism , Protein Binding , Pyridones/blood , Rats , Rats, Sprague-Dawley
16.
J Med Chem ; 49(3): 971-83, 2006 Feb 09.
Article in English | MEDLINE | ID: mdl-16451063

ABSTRACT

Recently, we disclosed a new class of HCV polymerase inhibitors discovered through high-throughput screening (HTS) of the GlaxoSmithKline proprietary compound collection. This interesting class of 3-(1,1-dioxo-2H-1,2,4-benzothiadiazin-3-yl)-4-hydroxy-2(1H)-quinolinones potently inhibits HCV polymerase enzymatic activity and inhibits the ability of the subgenomic HCV replicon to replicate in Huh-7 cells. This report will focus on the structure-activity relationships (SAR) of substituents on the quinolinone ring, culminating in the discovery of 1-(2-cyclopropylethyl)-3-(1,1-dioxo-2H-1,2,4-benzothiadiazin-3-yl)-6-fluoro-4-hydroxy-2(1H)-quinolinone (130), an inhibitor with excellent potency in biochemical and cellular assays possessing attractive molecular properties for advancement as a clinical candidate. The potential for development and safety assessment profile of compound 130 will also be discussed.


Subject(s)
Antiviral Agents/chemical synthesis , Benzothiadiazines/chemical synthesis , Hepacivirus/enzymology , Quinolones/chemical synthesis , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Thiadiazines/chemical synthesis , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Benzothiadiazines/chemistry , Benzothiadiazines/pharmacology , Biological Availability , Blood Proteins/metabolism , Cell Line , Crystallography, X-Ray , Dogs , Genotype , Half-Life , Hepacivirus/genetics , Macaca fascicularis , Models, Molecular , Molecular Structure , Mutation , Protein Binding , Quinolones/chemistry , Quinolones/pharmacology , RNA-Dependent RNA Polymerase/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Thiadiazines/chemistry , Thiadiazines/pharmacology
17.
J Med Chem ; 48(23): 7333-42, 2005 Nov 17.
Article in English | MEDLINE | ID: mdl-16279793

ABSTRACT

The cholecystokinin-8 (CCK-8)-inactivating peptidase is a serine peptidase that has been shown to be a membrane-bound isoform of tripeptidyl peptidase II (EC 3.4.14.10). It cleaves the neurotransmitter CCK-8 sulfate at the Met-Gly bond to give Asp-Tyr(SO3H)-Met-OH + Gly-Trp-Met-Asp-Phe-NH2. Starting from Val-Pro-NHBu, a dipeptide of submicromolar affinity that had previously been generated to serve as a lead, successive optimization at P3, P1, and then P2 gave Abu-Pro-NHBu (18, Ki = 80 nM). Further transformation (by making a benzologue) gave the indoline analogue, butabindide (33) as a reversible inhibitor having nanomolar affinity (Ki = 7 nM). Retrospective analysis suggested the possibility of a general approach to designing exopeptidase inhibitors starting from the structure of the first hydrolysis product. Application of this approach to CCK-8 led to Abu-Phe-NHBu (37), but this only had Ki = 9.4 microM. Molecular modeling, to determine the minimum energy conformations and explain the 1000-fold better affinity of butabindide, indicated that 37 cannot access the likely active conformation of butabindide.


Subject(s)
Indoles/chemical synthesis , Serine Endopeptidases/chemistry , Serine Proteinase Inhibitors/chemical synthesis , Aminopeptidases , Animals , Cerebral Cortex/enzymology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases , In Vitro Techniques , Indoles/chemistry , Indoles/pharmacology , Isoenzymes/chemistry , Isoenzymes/metabolism , Models, Molecular , Molecular Conformation , Rats , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Sincalide/metabolism , Structure-Activity Relationship , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...