Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 62(2): 641-653, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30571123

ABSTRACT

Herein, the synthesis and pharmacological characterization of an extended library of differently substituted N-methyl-14- O-methylmorphinans with natural and unnatural amino acids and three dipeptides at position 6 that emerged as potent µ/δ opioid receptor (MOR/DOR) agonists with peripheral antinociceptive efficacy is reported. The current study adds significant value to our initial structure-activity relationships on a series of zwitterionic analogues of 1 (14- O-methyloxymorphone) by targeting additional amino acid residues. The new derivatives showed high binding and potent agonism at MOR and DOR in vitro. In vivo, the new 6-amino acid- and 6-dipeptide-substituted derivatives of 1 were highly effective in inducing antinociception in the writhing test in mice after subcutaneous administration, which was antagonized by naloxone methiodide demonstrating activation of peripheral opioid receptors. Such peripheral opioid analgesics may represent alternatives to presently available drugs for a safer pain therapy.


Subject(s)
Analgesics, Opioid/chemical synthesis , Oxymorphone/analogs & derivatives , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Analgesics, Opioid/metabolism , Analgesics, Opioid/therapeutic use , Animals , Cell Membrane/metabolism , Dipeptides/chemistry , Humans , Male , Mice , Morphine/therapeutic use , Oxymorphone/chemistry , Oxymorphone/metabolism , Oxymorphone/therapeutic use , Pain/chemically induced , Pain/drug therapy , Pain/pathology , Protein Binding , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
2.
Curr Pharm Des ; 19(42): 7362-72, 2013.
Article in English | MEDLINE | ID: mdl-23448472

ABSTRACT

The κ opioid receptor (KOR) plays a significant role in many physiological functions, including pain relief, stress, depression, drug abuse, anxiety and psychotic behaviors. KORs are widely distributed in the central and peripheral nervous systems, and are specifically activated by endogenous opioids derived from prodynorphin. They are members of the G protein-coupled receptor superfamily, and the crystal structure of the human KOR was recently elucidated. KORs were initially studied for their involvement in mediation of pain as stimulation of KOR produces analgesia and minimizes abuse liability and other side effects. Nowadays, the KOR is rapidly emerging as an important target for the treatment of a variety of other human disorders. Specifically, the KOR system has become increasingly implicated as a modulator of stress-related and addictive behaviors. Several selective KOR partial agonists and antagonists have been developed as potential antidepressants, anxiolytic and anti-addiction medications. Although many KOR ligands have not demonstrated desirable pharmacological properties, some have been shown to be viable drug candidates. Herein, we describe chemical and pharmacological developments on KOR ligands, advantages and challenges, and potential therapeutic applications of ligands for KORs. In the second part, recent advances in the KOR drug design utilizing computational approaches are presented, with focus on the discovery of a new naturally derived scaffold, sewarine, as a novel class of selective KOR ligands with antagonist properties, using a pharmacophore-based virtual screening strategy.


Subject(s)
Drug Discovery , Receptors, Opioid, kappa/metabolism , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Humans , Ligands , Receptors, Opioid, kappa/antagonists & inhibitors
3.
Curr Pharm Des ; 19(42): 7415-34, 2013.
Article in English | MEDLINE | ID: mdl-23448479

ABSTRACT

Although the µ opioid receptor (MOR) was pharmacologically and biochemically identified in binding studies forty years ago, its structure, function, and true complexity only have emerged after its cloning in 1993. Continuous efforts from many laboratories have greatly advanced our understanding of MORs, ranging from their anatomic distribution to cellular and molecular mechanisms, and from cell lines to in vivo systems. The MOR is recognized as the main target for effective pain relief, but its involvement in many other physiological functions has also been recognized. This review provides a synopsis on the history of research on MORs and ligands acting at the MOR with the focus on their clinical and potential use as therapeutic drugs, or as valuable research tools. Since the elucidation of the chemical structure of morphine and the characterization of endogenous opioid peptides, research has stimulated the development of new generations of MOR ligands with distinct pharmacological profiles (agonist, antagonist, mixed agonist/antagonist and partial agonist) or site of action (central/peripheral). Discovery of therapeutically useful morphine-like drugs and innovative drugs with new scaffolds, with several outstanding representatives, is discussed. Extensive efforts on modifications of endogenous peptides to attain stable and MOR selective analogs are overviewed with stimulating results for the development of peptide-based pharmaceuticals. With pharmacophore modeling as an important tool in drug discovery, application of modern computational methodologies for the development of morphinans as new MOR ligands is described. Moreover, the crystal structure of the MOR available today will enable the application of structure-based approaches to design better drugs for the management of pain, addiction and other human diseases, where MORs play a key role.


Subject(s)
Receptors, Opioid, mu/metabolism , Humans , Ligands
4.
Mol Brain ; 5: 4, 2012 Jan 30.
Article in English | MEDLINE | ID: mdl-22289619

ABSTRACT

BACKGROUND: An important limiting factor in the development of centrally acting pharmaceuticals is the blood-brain barrier (BBB). Transport of therapeutic peptides through this highly protective physiological barrier remains a challenge for peptide drug delivery into the central nervous system (CNS). Because the most common strategy to treat moderate to severe pain consists of the activation of opioid receptors in the brain, the development of active opioid peptide analogues as potential analgesics requires compounds with a high resistance to enzymatic degradation and an ability to cross the BBB. RESULTS: Herein we report that tetrapeptide analogues of the type H-Dmt1-Xxx2-Yyy3-Gly4-NH2 are transported into the brain after intravenous and subcutaneous administration and are able to activate the µ- and δ opioid receptors more efficiently and over longer periods of time than morphine. Using the hot water tail flick test as the animal model for antinociception, a comparison in potency is presented between a side chain conformationally constrained analogue containing the benzazepine ring (BVD03, Yyy3: Aba), and a "ring opened" analogue (BVD02, Yyy3: Phe). The results show that in addition to the increased lipophilicity through amide bond N-methylation, the conformational constraint introduced at the level of the Phe3 side chain causes a prolonged antinociception. Further replacement of NMe-D-Ala2 by D-Arg2 in the tetrapeptide sequence led to an improved potency as demonstrated by a higher and maintained antinociception for AN81 (Xxx2: D-Arg) vs. BVD03 (Xxx2: NMe-D-Ala). A daily injection of the studied opioid ligands over a time period of 5 days did however result in a substantial decrease in antinociception on the fifth day of the experiment. The compact opioid agonist-NK1 antagonist hybrid SBCHM01 could not circumvent opioid induced tolerance. CONCLUSIONS: We demonstrated that the introduction of a conformational constraint has an important impact on opioid receptor activation and subsequent antinociception in vivo. Further amino acid substitution allowed to identify AN81 as an opioid ligand able to access the CNS and induce antinociception at very low doses (0.1 mg/kg) over a time period up to 7 hours. However, tolerance became apparent after repetitive i.v. administration of the investigated tetrapeptides. This side effect was also observed with the dual opioid agonist-NK1 receptor antagonist SBCHM01.


Subject(s)
Neurokinin-1 Receptor Antagonists , Nociception/drug effects , Oligopeptides/pharmacology , Opioid Peptides/agonists , Receptors, Opioid, mu/agonists , Recombinant Proteins/pharmacology , Analgesics/pharmacology , Animals , Dose-Response Relationship, Drug , Injections, Intravenous , Ligands , Mice , Mice, Inbred C57BL , Morphine/administration & dosage , Morphine/pharmacology , Oligopeptides/chemistry , Opioid Peptides/administration & dosage , Opioid Peptides/pharmacology , Receptors, Neurokinin-1/metabolism , Receptors, Opioid, mu/metabolism , Recombinant Proteins/chemistry , Time Factors
5.
J Med Chem ; 54(4): 980-8, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21235243

ABSTRACT

The synthesis and the effect of a combination of 6-glycine and 14-phenylpropoxy substitutions in N-methyl- and N-cycloproplymethylmorphinans on biological activities are described. Binding studies revealed that all new 14-phenylpropoxymorphinans (11-18) displayed high affinity to opioid receptors. Replacement of the 14-methoxy group with a phenylpropoxy group led to an enhancement in affinity to all three opioid receptor types, with most pronounced increases in δ and κ activities, hence resulting in a loss of µ receptor selectivity. All compounds (11-18) showed potent and long-lasting antinociceptive effects in the tail-flick test in rats after subcutaneous administration. For the N-methyl derivatives 13 and 14, analgesic potencies were in the range of their 14-methoxy analogues 9 and 10, respectively. Even derivatives 15-18 with an N-cyclopropylmethyl substituent acted as potent antinociceptive agents, being several fold more potent than morphine. Subcutaneous administration of compounds 13 and 14 produced significant and prolonged antinociceptive effects mediated through peripheral opioid mechanisms in carrageenan-induced inflammatory hyperalgesia in rats.


Subject(s)
Analgesics/chemical synthesis , Analgesics/pharmacology , Morphinans/chemical synthesis , Morphinans/pharmacology , N-substituted Glycines/chemical synthesis , N-substituted Glycines/pharmacology , Receptors, Opioid/metabolism , Analgesics/chemistry , Animals , Binding, Competitive , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Magnetic Resonance Spectroscopy , Morphinans/chemistry , N-substituted Glycines/chemistry , Pain/drug therapy , Rats , Spectrometry, Mass, Electrospray Ionization , Spectroscopy, Fourier Transform Infrared , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...