Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Molecules ; 24(23)2019 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-31779072

RESUMEN

The opioid receptors modulate a variety of biological functions, including pain, mood, and reward. As a result, opioid ligands are being explored as potential therapeutics for a variety of indications. Multifunctional opioid ligands, which act simultaneously at more than one type of opioid receptor, show promise for use in the treatment of addiction, pain, and other conditions. Previously, we reported the creation of bifunctional kappa opioid receptor (KOR) agonist/mu opioid receptor (MOR) partial agonist ligands from the classically delta opioid receptor (DOR) antagonist selective dimethyltyrosine-tetrahydroisoquinoline (Dmt-Tiq) scaffold through the addition of a 7-benzyl pendant on the tetrahydroisoquinoline ring. This study further explores the structure-activity relationships surrounding 7-position pendants on the Dmt-Tiq scaffold. Some analogues maintain a KOR agonist/MOR partial agonist profile, which is being explored in the development of a treatment for cocaine addiction. Others display a MOR agonist/DOR antagonist profile, which has potential to be used in the creation of a less addictive pain medication. Ultimately, we report the synthesis and in vitro evaluation of novel opioid ligands with a variety of multifunctional profiles.


Asunto(s)
Analgésicos Opioides/metabolismo , Peptidomiméticos/química , Peptidomiméticos/farmacología , Tetrahidroisoquinolinas/química , Tetrahidroisoquinolinas/farmacología , Animales , Células CHO , Línea Celular , Cricetulus , Humanos , Dolor/tratamiento farmacológico , Dolor/metabolismo , Manejo del Dolor/métodos , Receptores Opioides/metabolismo , Relación Estructura-Actividad
2.
Handb Exp Pharmacol ; 247: 21-51, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29675582

RESUMEN

The opioid receptor system plays a major role in the regulation of mood, reward, and pain. The opioid receptors therefore make attractive targets for the treatment of many different conditions, including pain, depression, and addiction. However, stimulation or blockade of any one opioid receptor type often leads to on-target adverse effects that limit the clinical utility of a selective opioid agonist or antagonist. Literature precedent suggests that the opioid receptors do not act in isolation and that interactions among the opioid receptors and between the opioid receptors and other proteins may produce clinically useful targets. Multifunctional ligands have the potential to elicit desired outcomes with reduced adverse effects by allowing for the activation of specific receptor conformations and/or signaling pathways promoted as a result of receptor oligomerization or crosstalk. In this chapter, we describe several classes of multifunctional ligands that interact with at least one opioid receptor. These ligands have been designed for biochemical exploration and the treatment of a wide variety of conditions, including multiple kinds of pain, depression, anxiety, addiction, and gastrointestinal disorders. The structures, pharmacological utility, and therapeutic drawbacks of these classes of ligands are discussed.


Asunto(s)
Receptores Opioides/efectos de los fármacos , Afecto/efectos de los fármacos , Analgésicos Opioides/farmacología , Animales , Antidepresivos/farmacología , Humanos , Ligandos , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides kappa/efectos de los fármacos , Receptores Opioides mu/efectos de los fármacos
3.
Biopolymers ; 102(1): 107-14, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24436042

RESUMEN

Most opioid analgesics used in the treatment of pain are mu opioid receptor (MOR) agonists. While effective, there are significant drawbacks to opioid use, including the development of tolerance and dependence. However, the coadministration of a MOR agonist with a delta opioid receptor (DOR) antagonist slows the development of MOR-related side effects, while maintaining analgesia. We have previously reported a series of cyclic mixed efficacy MOR agonist/DOR antagonist ligands. Here we describe the transfer of key features from these cyclic analogs to linear sequences. Using the linear MOR/DOR agonist, Tyr-DThr-Gly-Phe-Leu-Ser-NH2 (DTLES), as a lead scaffold, we replaced Phe(4) with bulkier and/or constrained aromatic residues shown to confer DOR antagonism in our cyclic ligands. These replacements failed to confer DOR antagonism in the DTLES analogs, presumably because the more flexible linear ligands can adopt binding poses that will fit in the narrow binding pocket of the active conformations of both MOR and DOR. Nonetheless, the pharmacological profile observed in this series, high affinity and efficacy for MOR and DOR with selectivity relative to KOR, has also been shown to reduce the development of unwanted side effects. We further modified our lead MOR/DOR agonist with a C-terminal glucoserine to improve bioavailability. The resulting ligand displayed high efficacy and potency at both MOR and DOR and no efficacy at KOR.


Asunto(s)
Péptidos Opioides/química , Péptidos Cíclicos/química , Secuencia de Aminoácidos , Aminoácidos/química , Animales , Sitios de Unión , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Modelos Moleculares , Datos de Secuencia Molecular , Ratas , Relación Estructura-Actividad
4.
bioRxiv ; 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39229079

RESUMEN

Fatal opioid overdoses in the United States have nearly tripled during the past decade, with greater than 92% involving a synthetic opioid like fentanyl. Fentanyl potently activates the µ-opioid receptor to induce both analgesia and respiratory depression. The danger of illicit fentanyl has recently been exacerbated by adulteration with xylazine, an α2-adrenergic receptor agonist typically used as a veterinary anesthetic. In 2023, over a 1,000% increase in xylazine-positive overdoses was reported in some regions of the U.S. Xylazine has been shown to potentiate the lethality of fentanyl in mice, yet a mechanistic underpinning for this effect has not been defined. Herein, we evaluate fentanyl, xylazine, and their combination in whole-body plethysmography (to measure respiration) and pulse oximetry (to measure blood oxygen saturation and heart rate) in male and female CD-1 mice. We show that xylazine decreases breathing rate more than fentanyl by increasing the expiration time. In contrast, fentanyl primarily reduces breathing by inhibiting inspiration, and xylazine exacerbates these effects. Fentanyl but not xylazine decreased blood oxygen saturation, and when combined, xylazine did not change the maximum level of fentanyl-induced hypoxia. Xylazine also reduced heart rate more than fentanyl. Finally, loss in blood oxygen saturation correlated with the frequency of fentanyl-induced apneas, but not breathing rate. Together, these findings provide insight into how the addition of xylazine to illicit fentanyl may increase the risk of overdose.

5.
ACS Chem Neurosci ; 15(15): 2830-2841, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38994846

RESUMEN

Opioid-related overdoses account for almost half of all drug overdose deaths in the United States and cause more preventable deaths every year than car crashes. Fentanyl, a highly potent mu opioid receptor (MOR) agonist and its analogues (fentalogues) are increasingly found in illicit drug samples, both where the primary drug of abuse is an opioid and where it is not. The prevalence of fentalogues in the illicit drug market is thought to be the primary driver of the increased number of opioid-related overdose deaths since 2016. In fact, fentanyl and its analogues are involved in more than 70% of opioid-related overdoses. The standard opioid overdose rescue therapy naloxone is often insufficient to reverse opioid overdoses caused by fentalogue agonists under current treatment paradigms. However, the pharmacology of many fentalogues is unknown. Moreover, within the fentalogue series of compounds, it is possible that antagonists could be identified that might be superior to naloxone as opioid overdose reversal agents. In this report, we explore the pharmacology of 70 fentalogues and identify compounds that behave as MOR antagonists in vitro and demonstrate with one of these reversals of fentanyl-induced respiratory depression in the mouse. Such compounds could provide leads for the development of effective agents for the reversal of opioid overdose.


Asunto(s)
Analgésicos Opioides , Fentanilo , Naloxona , Antagonistas de Narcóticos , Sobredosis de Opiáceos , Fentanilo/farmacología , Fentanilo/análogos & derivados , Animales , Sobredosis de Opiáceos/tratamiento farmacológico , Ratones , Antagonistas de Narcóticos/farmacología , Analgésicos Opioides/farmacología , Relación Estructura-Actividad , Naloxona/farmacología , Receptores Opioides mu/metabolismo , Humanos , Masculino
6.
ACS Chem Neurosci ; 13(21): 3108-3117, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36223082

RESUMEN

The opioid crisis continues to claim many lives, with a particular issue being the ready availability and use (whether intentional or accidental) of fentanyl and fentanyl analogues. Fentanyl is both potent and longer-acting than naloxone, the standard of care for overdose reversal, making it especially deadly. Consequently, there is interest in opioid reversal agents that are better able to counter its effects. The orvinol series of ligands are known for their high-affinity binding to opioid receptors and often extended duration of action; generally, compounds on this scaffold show agonist activity at the kappa and the mu-opioid receptor. Diprenorphine is an unusual member of this series being an antagonist at mu and only a partial agonist at kappa-opioid receptors. In this study, an orvinol antagonist, 14, was designed and synthesized that shows no agonist activity in vitro and is at least as good as naloxone at reversing the effects of mu-opioid receptor agonists in vivo.


Asunto(s)
Antagonistas de Narcóticos , Sobredosis de Opiáceos , Humanos , Antagonistas de Narcóticos/farmacología , Receptores Opioides mu/metabolismo , Naloxona/farmacología , Receptores Opioides kappa/metabolismo , Receptores Opioides/metabolismo , Fentanilo/farmacología , Analgésicos Opioides/farmacología
7.
ACS Chem Neurosci ; 12(1): 216-233, 2021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33346631

RESUMEN

We previously described the development of potent µ-opioid receptor (MOR)-agonist/δ-opioid receptor (DOR)-antagonist peptidomimetic ligands as an approach toward effective analgesics with reduced side effects. In this series, a tetrahydroquinoline (THQ) or substituted phenyl is employed to link two key pharmacophore elements, a dimethyltyrosine amino acid and typically an aromatic pendant. Using new and previously reported analogues, we constructed a structure-activity relationship (SAR) matrix that probes the utility of previously reported amine pendants. This matrix reveals that the MOR-agonist/DOR-antagonist properties of these ligands do not change when a tetrahydroisoquinoline (THIQ) pendant is used, despite removal of substituents on the core phenyl ring. Based on this observation, we retained the THIQ pendant and replaced the phenyl core with simpler aliphatic chain structures. These simpler analogues proved to be potent MOR-agonists with high variability in their effects at the DOR and the κ-opioid receptor (KOR). These data show that the amine of the THIQ pendant may be a novel pharmacophore element that favors high MOR-efficacy, whereas the aromatic ring of the THIQ pendant may produce high MOR-potency. Combined, the two pharmacophores within the THIQ pendant may be a structurally efficient means of converting opioid peptides and peptidomimetics into potent and efficacious MOR-agonists.


Asunto(s)
Peptidomiméticos , Aminas , Analgésicos Opioides/farmacología , Peptidomiméticos/farmacología , Receptores Opioides delta , Receptores Opioides kappa , Receptores Opioides mu , Relación Estructura-Actividad
8.
J Biol Chem ; 284(39): 26732-41, 2009 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-19542234

RESUMEN

Despite extensive characterization of the mu-opioid receptor (MOR), the biochemical properties of the isolated receptor remain unclear. In light of recent reports, we proposed that the monomeric form of MOR can activate G proteins and be subject to allosteric regulation. A mu-opioid receptor fused to yellow fluorescent protein (YMOR) was constructed and expressed in insect cells. YMOR binds ligands with high affinity, displays agonist-stimulated [(35)S]guanosine 5'-(gamma-thio)triphosphate binding to Galpha(i), and is allosterically regulated by coupled G(i) protein heterotrimer both in insect cell membranes and as purified protein reconstituted into a phospholipid bilayer in the form of high density lipoprotein particles. Single-particle imaging of fluorescently labeled receptor indicates that the reconstituted YMOR is monomeric. Moreover, single-molecule imaging of a Cy3-labeled agonist, [Lys(7), Cys(8)]dermorphin, illustrates a novel method for studying G protein-coupled receptor-ligand binding and suggests that one molecule of agonist binds per monomeric YMOR. Together these data support the notion that oligomerization of the mu-opioid receptor is not required for agonist and antagonist binding and that the monomeric receptor is the minimal functional unit in regard to G protein activation and strong allosteric regulation of agonist binding by G proteins.


Asunto(s)
Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Receptores Opioides mu/metabolismo , Regulación Alostérica , Secuencia de Aminoácidos , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacología , Animales , Unión Competitiva/efectos de los fármacos , Línea Celular , Encefalina Ala(2)-MeFe(4)-Gli(5)/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Lipoproteínas HDL/química , Lipoproteínas HDL/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Datos de Secuencia Molecular , Péptidos Opioides/metabolismo , Péptidos Opioides/farmacología , Unión Proteica , Receptores Opioides mu/genética , Receptores Opioides mu/aislamiento & purificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Spodoptera
9.
J Med Chem ; 63(4): 1671-1683, 2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-31986033

RESUMEN

We previously reported a novel SAR campaign that converted a metabolically unstable series of µ-opioid receptor (MOR) agonist/δ-opioid receptor (DOR) antagonist bicyclic core peptidomimetics with promising analgesic activity and reduced abuse liabilities into a more stable series of benzylic core analogues. Herein, we expanded the SAR of that campaign and determined that the incorporation of amines into the benzylic pendant produces enhanced MOR-efficacy in this series, whereas the reincorporation of an aromatic ring into the pendant enhanced MOR-potency. Two compounds, which contain a piperidine (14) or an isoindoline (17) pendant, retained the desired opioid profile in vitro, possessed metabolic half-lives of greater than 1 h in mouse liver microsomes (MLMs), and were active antinociceptive agents in the acetic acid stretch assay (AASA) at subcutaneous doses of 1 mg/kg.


Asunto(s)
Analgésicos Opioides/farmacología , Peptidomiméticos/farmacología , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Analgésicos Opioides/síntesis química , Analgésicos Opioides/metabolismo , Animales , Células CHO , Cricetulus , Masculino , Ratones Endogámicos C57BL , Microsomas Hepáticos/metabolismo , Estructura Molecular , Peptidomiméticos/síntesis química , Peptidomiméticos/metabolismo , Relación Estructura-Actividad
10.
Drug Test Anal ; 12(8): 1212-1221, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32415719

RESUMEN

Opioids are widely misused and account for almost half of overdose deaths in the United States. The cost in terms of lives, health care, and lost productivity is significant and has been declared a national crisis. Fentanyl is a highly potent mu opioid receptor (MOR) agonist and plays a significant role in the current opioid epidemic; fentanyl and its analogs (fentalogs) are increasingly becoming one of the biggest dangers in the opioid crisis. The availability of fentalogs in the illicit market is thought to play a significant role in the recent increase in opioid-related deaths. Although there is both rodent homolog in vivo and in vitro data for some fentalogs, prior to this publication very little was known about the pharmacology of many of these illicit compounds at the human MOR (hMOR). Using gas chromatography-mass spectrometry, nuclear magnetic resonance spectroscopy, and in vitro assays, this study describes the spectral and pharmacological properties of 34 fentalogs. The reported spectra and chemical data will allow for easy identification of novel fentalogs in unknown or mixed samples. Taken together these data are useful for law enforcement and clinical workers as they will aid in the identification of fentalogs in unknown samples and can potentially be used to predict physiological effects after exposure.


Asunto(s)
Analgésicos Opioides/farmacología , Fentanilo/farmacología , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Animales , Células CHO , Cricetulus , Fentanilo/análogos & derivados , Fentanilo/química , Cromatografía de Gases y Espectrometría de Masas , Humanos , Espectroscopía de Resonancia Magnética
11.
J Med Chem ; 62(8): 4142-4157, 2019 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-30924650

RESUMEN

We have previously reported a series of µ-opioid receptor (MOR) agonist/δ-opioid receptor (DOR) antagonist ligands to serve as potential nonaddictive opioid analgesics. These ligands have been shown to be active in vivo, do not manifest withdrawal syndromes or reward behavior in conditioned-place preference assays in mice, and do not produce dependence. Although these attributes are promising, these analogues exhibit poor metabolic stability in mouse liver microsomes, likely due to the central tetrahydroquinoline scaffold in this series. As such, a structure-activity relationship (SAR) campaign was pursued to improve their metabolic stability. This resulted in a shift from our original bicyclic tetrahydroquinoline core to a monocyclic benzylic-core system. By eliminating one of the rings in this scaffold and exploring the SAR of this new core, two promising analogues were discovered. These analogues (5l and 5m) had potency and efficacy values at MOR better or comparable to morphine, retained their DOR-antagonist properties, and showed a 10-fold improvement in metabolic stability.


Asunto(s)
Analgésicos Opioides/metabolismo , Peptidomiméticos , Quinolinas/química , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Animales , Diseño de Fármacos , Ratones , Microsomas Hepáticos/metabolismo , Morfina/química , Morfina/metabolismo , Quinolinas/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad
12.
ACS Chem Neurosci ; 10(8): 3682-3689, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31199621

RESUMEN

The dimethyltyrosine-tetrahydroisoquinoline (Dmt-Tiq) scaffold was originally developed in the production of selective delta opioid receptor (DOR) antagonists. Installation of a 7-benzyl pendant on the tetrahydroisoquinoline core of this classic opioid scaffold introduced kappa opioid receptor (KOR) agonism. Further modification of this pendant resulted in retention of KOR agonism and the addition of mu opioid receptor (MOR) partial agonism, a bifunctional profile with potential to be used in the treatment of cocaine addiction.


Asunto(s)
Peptidomiméticos/farmacología , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Tetrahidroisoquinolinas/farmacología , Animales , Línea Celular , Ratas , Relación Estructura-Actividad
13.
J Med Chem ; 62(8): 4193-4203, 2019 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-30916966

RESUMEN

Short-acting µ-opioid receptor (MOR) agonists have long been used for the treatment of severe, breakthrough pain. However, selective MOR agonists including fentanyl and morphine derivatives are limited clinically due to high risks of dependence, tolerance, and respiratory depression. We recently reported the development of a long-acting, bifunctional MOR agonist/δ-opioid receptor (DOR) antagonist analgesic devoid of tolerance or dependence in mice (AAH8, henceforth referred to as 2B). To address the need for short-acting treatments for breakthrough pain, we present a series of novel, short-acting, high-potency MOR agonist/DOR antagonist ligands with antinociceptive activity in vivo. In this study, we utilized a two-dimensional structure-activity relationship matrix to identify pharmacological trends attributable to combinations of two key pharmacophore elements within the chemotype. This work enhances our ability to modulate efficacy at MOR and DOR, accessing a variety of bifunctional profiles while maintaining high affinity and potency at both receptors.


Asunto(s)
Analgésicos Opioides/química , Diseño de Fármacos , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Analgésicos Opioides/metabolismo , Analgésicos Opioides/uso terapéutico , Animales , Línea Celular , Humanos , Cinética , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Dolor/tratamiento farmacológico , Dolor/patología , Peptidomiméticos , Unión Proteica , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad
15.
ACS Chem Neurosci ; 9(7): 1840-1848, 2018 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-29677442

RESUMEN

The use of opioids for the treatment of pain, while largely effective, is limited by detrimental side effects including analgesic tolerance, physical dependence, and euphoria, which may lead to opioid abuse. Studies have shown that compounds with a µ-opioid receptor (MOR) agonist/δ-opioid receptor (DOR) antagonist profile reduce or eliminate some of these side effects including the development of tolerance and dependence. Herein we report the synthesis and pharmacological evaluation of a series of tetrahydroquinoline-based peptidomimetics with substitutions at the C-8 position. Relative to our lead peptidomimetic with no C-8 substitution, this series affords an increase in DOR affinity and provides greater balance in MOR and DOR binding affinities. Moreover, compounds with carbonyl moieties at C-8 display the desired MOR agonist/DOR antagonist profile whereas alkyl substitutions elicit modest DOR agonism. Several compounds in this series produce a robust antinociceptive effect in vivo and show antinociceptive activity for greater than 2 h after intraperitoneal administration in mice.


Asunto(s)
Analgésicos Opioides/síntesis química , Analgésicos Opioides/farmacología , Dolor/tratamiento farmacológico , Quinolinas/síntesis química , Quinolinas/farmacología , Animales , Línea Celular , Cricetulus , Humanos , Masculino , Ratones Endogámicos C57BL , Estructura Molecular , Dolor/metabolismo , Unión Proteica , Ratas , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad
16.
Br J Pharmacol ; 175(11): 2013-2027, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29352503

RESUMEN

BACKGROUND AND PURPOSE: Agonists at µ-opioid receptors (µ-receptors) are used for pain management but produce adverse effects including tolerance, dependence and euphoria. The co-administration of a µ-receptor agonist with a δ-opioid receptor (δ-receptor) antagonist has been shown to produce antinociception with reduced development of some side effects. We characterized the effects of three µ-receptor agonist/δ-receptor antagonist peptidomimetics in vivo after acute and repeated administration to determine if this profile provides a viable alternative to traditional opioid analgesics. EXPERIMENTAL APPROACH: Three µ-receptor agonist / δ-receptor antagonist peptidomimetics, AAH8, AMB46 and AMB47, and morphine were evaluated for the development of tolerance and dependence after 5 days of twice daily treatment with escalating doses of drug (10-50 mg·kg-1 ). Antinociceptive effects were measured in the warm water tail withdrawal assay before and after repeated drug treatment. Physical dependence was evaluated by naltrexone-precipitated withdrawal jumping. The rewarding effects of AAH8 were evaluated using a conditioned place preference (CPP) assay with twice daily conditioning sessions performed for 5 days. KEY RESULTS: Morphine, AAH8, AMB47 and AMB46 all demonstrated acute antinociceptive effects, but repeated administration only produced tolerance in animals treated with morphine and AMB46. Injection of naltrexone precipitated fewer jumps in mice treated repeatedly with AAH8 as compared with morphine, AMB47 or AMB46. Conditioning with morphine, but not AAH8, produced significant CPP. CONCLUSIONS AND IMPLICATIONS: AAH8 may be a better alternative than traditional opioid analgesics, producing antinociception with less development of tolerance and dependence and may be less rewarding than morphine.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacología , Peptidomiméticos/administración & dosificación , Peptidomiméticos/farmacología , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Animales , Células CHO , Línea Celular Tumoral , Cricetulus , Femenino , Humanos , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Conformación Molecular , Peptidomiméticos/química , Ratas
17.
Psychopharmacology (Berl) ; 233(13): 2479-87, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27117141

RESUMEN

RATIONALE: VRP26 displays mu opioid receptor agonist and delta opioid receptor antagonist activity in vitro, a pharmacological profile purported to produce reduced tolerance, dependence, and rewarding effects. We hypothesized that VRP26 would display reduced adverse effects after chronic administration as compared with the traditional opioid analgesic fentanyl. OBJECTIVE: The aim of this study is to explore the development of tolerance, dependence, and conditioned place preference of VRP26 as compared with the traditional opioid analgesic fentanyl. METHODS: The antinociceptive effects of VRP26 and fentanyl were assessed using the mouse warm water tail withdrawal (WWTW) assay. Measurement of antinociceptive tolerance and physical dependence occurred after 7 days of continuous administration of either fentanyl (0.3 mg/kg/day) or VRP26 (10 mg/kg/day); tolerance was measured by a shift in the antinociceptive dose response curve in the WWTW assay. Physical dependence was determined by observation of withdrawal symptoms after precipitated withdrawal. Rewarding effects were measured by the ability of VRP26 or fentanyl to produce conditioned place preference. RESULTS: Fentanyl produced significant tolerance and dependence, as well as significant conditioned place preference. VRP26 produced neither tolerance nor physical dependence, nor did it produce significant conditioned place preference. CONCLUSIONS: These results suggest that chronic treatment with VRP26 may produce less tolerance or physical dependence than chronic treatment with clinically available mu opioid analgesics such as fentanyl. Additionally, VRP26 produces less rewarding effects than fentanyl. This desirable in vivo profile may be due to the mixed efficacy nature of VRP26 and could provide the framework for safer opioid analgesics.


Asunto(s)
Analgésicos Opioides/farmacología , Fentanilo/farmacología , Antagonistas de Narcóticos/farmacología , Péptidos Cíclicos/farmacología , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Recompensa
18.
J Med Chem ; 59(10): 4985-98, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27148755

RESUMEN

N-Acetylation of the tetrahydroquinoline (THQ) core of a series of µ-opioid receptor (MOR) agonist/δ-opioid receptor (DOR) antagonist ligands increases DOR affinity, resulting in ligands with balanced MOR and DOR affinities. We report a series of N-substituted THQ analogues that incorporate various carbonyl-containing moieties to maintain DOR affinity and define the steric and electronic requirements of the binding pocket across the opioid receptors. 4h produced in vivo antinociception (ip) for 1 h at 10 mg/kg.


Asunto(s)
Quinolinas/química , Quinolinas/farmacología , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Conformación Molecular , Quinolinas/síntesis química , Ratas , Relación Estructura-Actividad
19.
J Med Chem ; 58(22): 8952-69, 2015 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-26524472

RESUMEN

In a previously described peptidomimetic series, we reported the development of bifunctional µ-opioid receptor (MOR) agonist and δ-opioid receptor (DOR) antagonist ligands with a lead compound that produced antinociception for 1 h after intraperitoneal administration in mice. In this paper, we expand on our original series by presenting two modifications, both of which were designed with the following objectives: (1) probing bioavailability and improving metabolic stability, (2) balancing affinities between MOR and DOR while reducing affinity and efficacy at the κ-opioid receptor (KOR), and (3) improving in vivo efficacy. Here, we establish that, through N-acetylation of our original peptidomimetic series, we are able to improve DOR affinity and increase selectivity relative to KOR while maintaining the desired MOR agonist/DOR antagonist profile. From initial in vivo studies, one compound (14a) was found to produce dose-dependent antinociception after peripheral administration with an improved duration of action of longer than 3 h.


Asunto(s)
Analgésicos/síntesis química , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Acetilación , Analgésicos/química , Analgésicos/farmacología , Animales , Disponibilidad Biológica , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Dimensión del Dolor/efectos de los fármacos , Ensayo de Unión Radioligante , Receptores Opioides delta/química , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato
20.
ACS Chem Neurosci ; 6(8): 1428-35, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-25938166

RESUMEN

We previously reported a small series of mixed-efficacy µ opioid receptor (MOR) agonist/δ opioid receptor (DOR) antagonist peptidomimetics featuring a tetrahydroquinoline scaffold and showed the promise of this series as effective analgesics after intraperitoneal administration in mice. We report here an expanded structure-activity relationship study of the pendant region of these compounds and focus in particular on the incorporation of heteroatoms into this side chain. These analogues provide new insight into the binding requirements for this scaffold at MOR, DOR, and the κ opioid receptor (KOR), and several of them (10j, 10k, 10m, and 10n) significantly improve upon the overall MOR agonist/DOR antagonist profile of our previous compounds. In vivo data for 10j, 10k, 10m, and 10n are also reported and show the antinociceptive potency and duration of action of compounds 10j and 10m to be comparable to those of morphine.


Asunto(s)
Analgésicos Opioides/farmacología , Antagonistas de Narcóticos/farmacología , Quinolinas/farmacología , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Analgésicos Opioides/sangre , Analgésicos Opioides/síntesis química , Analgésicos Opioides/química , Animales , Dominio Catalítico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Masculino , Ratones Endogámicos C57BL , Estructura Molecular , Antagonistas de Narcóticos/sangre , Antagonistas de Narcóticos/síntesis química , Antagonistas de Narcóticos/química , Dolor Nociceptivo/tratamiento farmacológico , Quinolinas/sangre , Quinolinas/síntesis química , Quinolinas/química , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA