Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Phys Chem Chem Phys ; 23(22): 12549-12558, 2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34008647

ABSTRACT

The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) enters the host cell after the receptor binding domain (RBD) of the virus spike (S) glycoprotein binds to the human angiotensin-converting enzyme 2 (hACE2). This binding requires the RBD to undergo a conformational change from a closed to an open state. In the present study, a key pair of salt bridges formed by the side chains of K537 and E619, residues at the interfaces of SD1 and SD2, respectively, was identified to promote the opening of the RBD. Mutations of K537Q and E619D reduced their side chain lengths and eliminated this pair of salt bridges; as a result, the opening of the RBD was not observed in the MD simulations. Thus, blocking the formation of this pair of salt bridges is a promising approach for treating novel coronavirus disease 2019 (COVID-19). FDA approved drug molecules were screened by their capabilities of blocking the formation of the key pair of salt bridges, achieved by their positional stabilities in the cavity containing the side chains of K537 and E619 formed in the interface between SD1 and SD2. Simeprevir, imatinib, and naldemedine were identified to possess the desired capability with the most favorable interaction energies.


Subject(s)
Antiviral Agents/pharmacology , Drug Design , SARS-CoV-2/drug effects , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Antiviral Agents/chemistry , Drug Evaluation, Preclinical , Humans , Imatinib Mesylate/chemistry , Imatinib Mesylate/pharmacology , Molecular Docking Simulation , Naltrexone/analogs & derivatives , Naltrexone/chemistry , Naltrexone/pharmacology , Protein Domains/drug effects , SARS-CoV-2/chemistry , Simeprevir/chemistry , Simeprevir/pharmacology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism
2.
ACS Chem Neurosci ; 6(11): 1813-24, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26325040

ABSTRACT

3-Iodobenzoyl naltrexamine (IBNtxA) is a potent analgesic belonging to the pharmacologically diverse 6ß-amidoepoxymorphinan group of opioids. We present the synthesis and pharmacological evaluation of five analogs of IBNtxA. The scaffold of IBNtxA was modified by removing the 14-hydroxy group, incorporating a 7,8 double bond and various N-17 alkyl substituents. The structural modifications resulted in analogs with picomolar affinities for opioid receptors. The lead compound (MP1104) was found to exhibit approximately 15-fold greater antinociceptive potency (ED50 = 0.33 mg/kg) compared with morphine, mediated through the activation of kappa- and delta-opioid receptors. Despite its kappa agonism, this lead derivative did not cause place aversion or preference in mice in a place-conditioning assay, even at doses 3 times the analgesic ED50. However, pretreatment with the lead compound prevented the reward behavior associated with cocaine in a conditioned place preference assay. Together, these results suggest the promise of dual acting kappa- and delta-opioid receptor agonists as analgesics and treatments for cocaine addiction.


Subject(s)
Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/pharmacology , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Morphinans/chemical synthesis , Morphinans/pharmacology , Analgesics, Opioid/chemistry , Animals , Cocaine-Related Disorders/drug therapy , Cocaine-Related Disorders/metabolism , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Drug Evaluation, Preclinical , Male , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Morphinans/chemistry , Naltrexone/analogs & derivatives , Naltrexone/chemistry , Nociception/drug effects , Nociception/physiology , Nuclear Receptor Subfamily 1, Group F, Member 1/pharmacology , Random Allocation , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Reward , Spatial Behavior/drug effects , Spatial Behavior/physiology
3.
Bioorg Med Chem ; 23(14): 3948-56, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25593096

ABSTRACT

Optimization of the sulfonamide-based kappa opioid receptor (KOR) antagonist probe molecule ML140 through constraint of the sulfonamide nitrogen within a tetrahydroisoquinoline moiety afforded a marked increase in potency. This strategy, when combined with additional structure-activity relationship exploration, has led to a compound only six-fold less potent than norBNI, a widely utilized KOR antagonist tool compound, but significantly more synthetically accessible. The new optimized probe is suitably potent for use as an in vivo tool to investigate the therapeutic potential of KOR antagonists.


Subject(s)
Benzamides/pharmacology , Receptors, Opioid, kappa/antagonists & inhibitors , Structure-Activity Relationship , Sulfonamides/pharmacology , Animals , Arrestins/metabolism , Benzamides/chemistry , CHO Cells , Chemistry Techniques, Synthetic , Cricetulus , Drug Evaluation, Preclinical/methods , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Naltrexone/analogs & derivatives , Naltrexone/chemistry , Narcotic Antagonists/chemistry , Narcotic Antagonists/pharmacology , Receptors, Opioid, kappa/genetics , Sulfonamides/chemistry , Tetrahydroisoquinolines/chemistry , beta-Arrestins
4.
Bioorg Med Chem ; 16(17): 7956-67, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18701308

ABSTRACT

We have previously reported on antitussive effect of (5R,9R,13S,14S)-17-cyclopropylmethyl-6,7-didehydro-4,5-epoxy-5',6'-dihydro-3-methoxy-4'H-pyrrolo[3,2,1-ij]quinolino[2',1':6,7]morphinan-14-ol(1b) methanesulfonate (TRK-850), a selective delta opioid receptor antagonist which markedly reduced the number of coughs in a rat cough model. We designed TRK-850 based on naltrindole (NTI), a typical delta opioid receptor antagonist, to improve its permeability through the blood-brain barrier by introducing hydrophobic moieties to NTI. The ED(50) values of NTI and compound 1b by intraperitoneal injections were 104 microg/kg and 2.07 microg/kg, respectively. This increased antitussive potency probably resulted from the improved brain exposure of compound 1b. However, 1b was extremely unstable toward metabolism by cytochrome P450. In this study, we designed and synthesized compound 1b derivatives to improve the metabolic instability, which resulted in affording highly potent and metabolically stable oral antitussive agent (5R,9R,13S,14S)-17-cyclopropylmethyl-6,7-didehydro-4,5-epoxy-8'-fluoro-5',6'-dihydro-4'H-pyrrolo[3,2,1-ij]quinolino[2',1':6,7]morphinan-3,14-diol (1c) methanesulfonate (TRK-851).


Subject(s)
Antitussive Agents/administration & dosage , Antitussive Agents/chemical synthesis , Drug Design , Naltrexone/analogs & derivatives , Receptors, Opioid, delta/antagonists & inhibitors , Administration, Oral , Animals , Antitussive Agents/chemistry , Capsaicin , Cough/chemically induced , Cough/drug therapy , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Male , Mice , Mice, Inbred Strains , Molecular Conformation , Naltrexone/administration & dosage , Naltrexone/chemical synthesis , Naltrexone/chemistry , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship
5.
J Med Chem ; 51(15): 4404-11, 2008 Aug 14.
Article in English | MEDLINE | ID: mdl-18637671

ABSTRACT

We have previously reported antitussive effects of naltrindole (NTI), a typical delta opioid receptor antagonist, in a rat model. The ED50 values of NTI by intraperitoneal and peroral injections were 104 microg/kg and 1840 microg/kg, respectively, comparable to those of codeine. Codeine, one of the most reliable centrally acting antitussive drugs, has micro agonist activity and thus the same side effects as morphine, e.g., constipation, dependency, and respiratory depression. Because NTI is a delta opioid antagonist, its derivatives have potential as highly potent antitussives, free from the mu opioid agonist side effects. We attempted to optimize the NTI derivatives to develop novel antitussive agents. On the basis of the studies of structure-antitussive activity relationships of alkyl substituted NTI derivatives, we designed NTI derivatives with extra ring fused structures. As a clinical candidate, we identified a highly potent new compound, (5R,9R,13S,14S)-17-cyclopropylmethyl-6,7-didehydro-4,5-epoxy-5',6'-dihydro-3-methoxy-4'H-pyrrolo[3,2,1-ij]quinolino[2',1':6,7]morphinan-14-ol (5b) methanesulfonate (TRK-850) which was effective even by oral administration (ED50 6.40 microg/kg).


Subject(s)
Antitussive Agents/chemical synthesis , Antitussive Agents/therapeutic use , Naltrexone/analogs & derivatives , Alkylation , Animals , Antitussive Agents/chemistry , CHO Cells , Capsaicin/pharmacology , Cough/chemically induced , Cough/drug therapy , Cricetinae , Cricetulus , Male , Mice , Molecular Structure , Naltrexone/chemical synthesis , Naltrexone/chemistry , Naltrexone/therapeutic use , Rats , Structure-Activity Relationship
6.
Int J Pharm ; 340(1-2): 104-18, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17482393

ABSTRACT

This research is based on the recognized need for an in vitro release method for drug implants that better simulate physiological conditions at the site of implantation ('biorelevance'). In this paper, we describe the evaluation of a 'biorelevant' approach for in vitro drug release testing of a biodegradable implant of naltrexone in a pre-clinical stage of development. A miniature, capillary cell culture device was modified and tested as a biorelevant alternative for a standard commercially available flow-through cell. The real-time data generated through 90 days indicated a 48% lower rate of release for the capillary system. The profiles using both systems followed zero-order kinetics after an initial period of burst release. In vitro release data from the capillary device resulted in a 1-to-1 correlation with dog plasma pharmacokinetic data, and furthermore, the capillary device potentially simulated the lag-time in absorption more effectively than the flow-through cell. Scanning electron micrographs revealed that the sheath was continuous with no signs of cracks at the end of in vitro and in vivo studies. However, at the interface of the sheath and the core, intercalating, "finger-like" projections were observed consistent with penetration of the medium. No macroscopic or clinical toxicity signs were observed during the in vivo implantation study.


Subject(s)
Bioreactors , Drug Evaluation, Preclinical/instrumentation , Drug Implants , Naltrexone/chemistry , Narcotic Antagonists/chemistry , Technology, Pharmaceutical/instrumentation , Animals , Chemistry, Pharmaceutical , Diffusion , Dogs , Drug Compounding , Drug Implants/adverse effects , Drug Implants/chemistry , Equipment Design , Foreign-Body Reaction/etiology , Foreign-Body Reaction/pathology , Kinetics , Models, Biological , Naltrexone/adverse effects , Naltrexone/blood , Naltrexone/pharmacokinetics , Narcotic Antagonists/adverse effects , Narcotic Antagonists/blood , Narcotic Antagonists/pharmacokinetics , Reproducibility of Results , Solubility , Subcutaneous Tissue/ultrastructure , Surface Properties
7.
Int J Pharm ; 340(1-2): 119-25, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17482777

ABSTRACT

The development of a 'biorelevant' approach for accelerating drug release from an implant is described. A miniature, capillary system has been shown previously to be suitable for real-time release tests for a biodegradable, naltrexone implant. Whereas the real-time study under physiological condition was essential for evaluation of the system, the accelerated (short-term) method provides for a faster assessment of in vitro drug release that would be useful in product development and quality control. Increased temperature was employed as the mechanism for accelerating drug release. Release rates were investigated and compared using modifications of two devices: the flow-through cell and the new, potentially more 'biorelevant' capillary device. The data generated for accelerated release using both devices through 45 days indicated approximately two-fold and four-fold increases in release rates at 45 and 55 degrees C, respectively, as compared to the real-time release rate. The similar activation energy values for both devices obtained from Arrhenius plots demonstrated that the release mechanism had been consistent; and that the rates of release could be used for long-term prediction. The rate of release reverted to that observed in real-time data, however, upon a reduction of temperature to 38 degrees C. The results demonstrated that temperature was the sole factor involved in modification of the release rate in vitro. The profiles using both systems followed zero-order kinetics after an initial period of burst release.


Subject(s)
Bioreactors , Drug Evaluation, Preclinical/instrumentation , Drug Implants , Naltrexone/chemistry , Narcotic Antagonists/chemistry , Technology, Pharmaceutical/instrumentation , Chemistry, Pharmaceutical , Drug Compounding , Equipment Design , Kinetics , Models, Chemical , Reproducibility of Results , Solubility , Temperature
8.
Bioorg Med Chem ; 12(2): 417-21, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14723960

ABSTRACT

Naltrexone (1), which is a member of the group of competitive opioid antagonists, shows a strong affinity for mu-receptors and its derivatives have been notable as novel receptor antagonists. In this paper, the preparation of several naltrexone derivatives is described; these were used to investigate the role of the oxygenated functional groups in facilitating binding to a series of the opioid receptors. The derivatives showed affinity for opioid mu-receptors which was similar to that of naltrexone, but these compounds, which had masked hydroxyl functional groups, displayed a moderate activity. These results suggest that every oxygenated functional group in naltrexone (1) plays an important role in binding to the opioid receptor.


Subject(s)
Naltrexone/analogs & derivatives , Narcotic Antagonists , Narcotic Antagonists/chemistry , Narcotic Antagonists/metabolism , Receptors, Opioid/metabolism , Animals , Biochemistry/methods , Brain/metabolism , Crystallography, X-Ray , Drug Evaluation, Preclinical/methods , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism , Guinea Pigs , Ligands , Mice , Naltrexone/chemistry , Narcotic Antagonists/pharmacology , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL