Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.031
Filter
1.
J Med Chem ; 67(12): 10447-10463, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38869493

ABSTRACT

In recent years, synthetic opioids have emerged as a predominant cause of drug-overdose-related fatalities, causing the "opioid crisis." To design safer therapeutic agents, we accidentally discovered µ-opioid receptor (MOR) antagonists based on fentanyl with a relatively uncomplicated chemical composition that potentiates structural modifications. Here, we showed the development of novel atropisomeric fentanyl analogues that exhibit more potent antagonistic activity against MOR than naloxone, a morphinan MOR antagonist. Derivatives displaying stable axial chirality were synthesized based on the amide structure of fentanyl. The aS- and aR-enantiomers exerted antagonistic and agonistic effects on the MOR, respectively, and each atropisomer interacted with the MOR by assuming a distinct binding mode through molecular docking. These findings suggest that introducing atropisomerism into fentanyl may serve as a key feature in the molecular design of future MOR antagonists to help mitigate the opioid crisis.


Subject(s)
Fentanyl , Receptors, Opioid, mu , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism , Fentanyl/pharmacology , Fentanyl/analogs & derivatives , Fentanyl/chemistry , Stereoisomerism , Humans , Molecular Docking Simulation , Structure-Activity Relationship , Animals , Narcotic Antagonists/chemistry , Narcotic Antagonists/pharmacology , Molecular Conformation , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemistry , Analgesics, Opioid/chemical synthesis , CHO Cells , Cricetulus
2.
Int J Nanomedicine ; 19: 4759-4777, 2024.
Article in English | MEDLINE | ID: mdl-38828199

ABSTRACT

Background: Opioids are irreplaceable analgesics owing to the lack of alternative analgesics that offer opioid-like pain relief. However, opioids have many undesirable central side effects. Restricting opioids to peripheral opioid receptors could reduce those effects while maintaining analgesia. Methods: To achieve this goal, we developed Tet1-LNP (morphine), a neural-targeting lipid nanoparticle encapsulating morphine that could specifically activate the peripheral opioid receptor in the dorsal root ganglion (DRG) and significantly reduce the side effects caused by the activation of opioid receptors in the brain. Tet1-LNP (morphine) were successfully prepared using the thin-film hydration method. In vitro, Tet1-LNP (morphine) uptake was assessed in differentiated neuron-like PC-12 cells and dorsal root ganglion (DRG) primary cells. The uptake of Tet1-LNP (morphine) in the DRGs and the brain was assessed in vivo. Von Frey filament and Hargreaves tests were used to assess the antinociception of Tet1-LNP (morphine) in the chronic constriction injury (CCI) neuropathic pain model. Morphine concentration in blood and brain were evaluated using ELISA. Results: Tet1-LNP (morphine) had an average size of 131 nm. Tet1-LNP (morphine) showed high cellular uptake and targeted DRG in vitro. CCI mice treated with Tet1-LNP (morphine) experienced prolonged analgesia for nearly 32 h compared with 3 h with free morphine (p < 0.0001). Notably, the brain morphine concentration in the Tet1-LNP (morphine) group was eight-fold lower than that in the morphine group (p < 0.0001). Conclusion: Our study presents a targeted lipid nanoparticle system for peripheral neural delivery of morphine. We anticipate Tet1-LNP (morphine) will offer a safe formulation for chronic neuropathic pain treatment, and promise further development for clinical applications.


Subject(s)
Analgesics, Opioid , Ganglia, Spinal , Morphine , Nanoparticles , Animals , Morphine/administration & dosage , Morphine/pharmacokinetics , Morphine/chemistry , Morphine/pharmacology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Nanoparticles/chemistry , Rats , PC12 Cells , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Male , Neuralgia/drug therapy , Mice , Lipids/chemistry , Proto-Oncogene Proteins/metabolism , Peripheral Nerves/drug effects , Mixed Function Oxygenases/metabolism , DNA-Binding Proteins , Liposomes
3.
Anal Chim Acta ; 1312: 342686, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38834256

ABSTRACT

BACKGROUND: Fentanyl and its derivatives are a type of potent opioid analgesics, with the characteristics of diverse structure, high toxicity, extremely low content, and high fatality rate. Currently, they have become one of the most serious problems in international drug abuse control due to their extensive use in drug production and use. Therefore, the development of a rapid, sensitive, and accurate method for detecting trace fentanyl is of great significance. In this study, in view of its complex structure and trace concentration, a new molecular imprinting electrochemical sensor was developed through molecular simulations followed by experimental validation to detect trace fentanyl. RESULTS: The process consisted of first obtaining the optimal functional monomer and its molar ratio through molecular simulations. The recognition sites of fentanyl-imprinted polymers were predicted to guide the synthesis of imprinted membranes with precision approach to ensure an efficient and accurate reaction process. Reduced graphene oxide (ErGO) was then deposited on glassy carbon electrode surface by electrochemical reduction to yield large numbers of active sites suitable for catalyzing reactions of fentanyl piperidine for promoted efficient electron transfer and amplified sensitivity of the sensor. Accordingly, fentanyl molecularly imprinted film was formed through one-step electropolymerization to yield greatly improved sensing selectivity due to the specific recognition of molecularly imprinted polymer. Under optimal experimental conditions, the fentanyl sensor showed an extended detection range of 3.84 × 10-9 mol L-1-1.72 × 10-6 mol L-1 and a detection limit of 1.28 × 10-9 mol L-1. SIGNIFICANCE: A distinctive feature of this sensor is its molecularly imprinted polymerized membrane, which offers excellent specific recognition, thereby boosting the sensor's selectivity. Throughout the sensor's development process, molecular simulations were employed to steer the synthesis of molecularly imprinted polymers and predict the recognition sites of fentanyl-imprinted polymers. The experimental outcomes proved to align with the simulation data. The final sensor exhibited outstanding selectivity, repeatability, stability, and high sensitivity. The sensor was effectively used to reliably track fentanyl in human serum samples, with acceptable analytical reliability, suggesting its potential for practical applications.


Subject(s)
Electrochemical Techniques , Fentanyl , Molecular Imprinting , Fentanyl/analysis , Fentanyl/blood , Fentanyl/chemistry , Molecularly Imprinted Polymers/chemistry , Electrodes , Limit of Detection , Graphite/chemistry , Molecular Dynamics Simulation , Analgesics, Opioid/blood , Analgesics, Opioid/analysis , Analgesics, Opioid/chemistry , Humans
4.
Int J Pharm ; 659: 124295, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38823469

ABSTRACT

Opioids are powerful analgesics; however, their significant systemic adverse effects and the need for frequent administration restrict their use. Nalbuphine (NA) is a κ-agonist narcotic with limited adverse effects, but needs to be frequently administrated due to its short elimination half-life. Whereas sebacoyl dinalbuphine ester (SDE) is a NA prodrug, which can effectively prolong the analgesic effect, but lacks immediate pain relief. Therefore, in this study, a rapid and sustained local delivery formulation to introduce NA and SDE directly into surgical sites was developed. An amphiphilic nanostructured lipid carrier (NLC) poloxamer 407 (P407) gel (NLC-Gel) was developed to permit concurrent delivery of hydrophobic SDE from the NLC core and hydrophilic NA from P407, offering a dual rapid and prolonged analgesic effect. Benefiting from the thermal-sensitive characteristic of P407, the formulation can be injected in liquid phase and instantly transit into gel at wound site. NLC-Gel properties, including particle size, drug release, rheology, and stability, were assessed. In vivo evaluation using a rat spinal surgery model highlighted the effect of the formulation through pain behavior test and hematology analysis. NLC-Gels demonstrated an analgesic effect comparable with that of commercial intramuscular injected SDE formulation (IM SDE), with only 15 % of the drug dosage. The inclusion of supplemental NA in the exterior gel (PA12-Gel + NA) provided rapid drug onset owing to swift NA dispersion, addressing acute pain within hours along with prolonged analgesic effects. Our findings suggest that this amphiphilic formulation significantly enhanced postoperative pain management in terms of safety and efficacy.


Subject(s)
Analgesics, Opioid , Drug Carriers , Drug Liberation , Gels , Nalbuphine , Pain, Postoperative , Poloxamer , Rats, Sprague-Dawley , Nalbuphine/administration & dosage , Pain, Postoperative/drug therapy , Animals , Male , Poloxamer/chemistry , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/chemistry , Drug Carriers/chemistry , Rats , Lipids/chemistry , Particle Size , Nanostructures/administration & dosage , Nanostructures/chemistry , Esters/chemistry
5.
Molecules ; 29(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38893511

ABSTRACT

The opioid crisis in the United States is a significant public health issue, with a nearly threefold increase in opioid-related fatalities between 1999 and 2014. In response to this crisis, society has made numerous efforts to mitigate its impact. Recent advancements in understanding the structural intricacies of the κ opioid receptor (KOR) have improved our knowledge of how opioids interact with their receptors, triggering downstream signaling pathways that lead to pain relief. This review concentrates on the KOR, offering crucial structural insights into the binding mechanisms of both agonists and antagonists to the receptor. Through comparative analysis of the atomic details of the binding site, distinct interactions specific to agonists and antagonists have been identified. These insights not only enhance our understanding of ligand binding mechanisms but also shed light on potential pathways for developing new opioid analgesics with an improved risk-benefit profile.


Subject(s)
Analgesics, Opioid , Receptors, Opioid, kappa , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, kappa/chemistry , Humans , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Animals , Binding Sites , Ligands , Signal Transduction/drug effects , Protein Binding , Structure-Activity Relationship , Narcotic Antagonists/chemistry , Pain/drug therapy , Pain/metabolism
6.
Bioorg Chem ; 149: 107507, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38850778

ABSTRACT

Opioids are currently the most effective and widely used painkillers in the world. Unfortunately, the clinical use of opioid analgesics is limited by serious adverse effects. Many researchers have been working on designing and optimizing structures in search of novel µ opioid receptor(MOR) agonists with improved analgesic activity and reduced incidence of adverse effects. There are many strategies to develop MOR drugs, mainly focusing on new low efficacy agonists (potentially G protein biased agonists), MOR agonists acting on different Gα subtype, targeting opioid receptors in the periphery, acting on multiple opioid receptor, and targeting allosteric sites of opioid receptors, and others. This review summarizes the design methods, clinical applications, and structure-activity relationships of small-molecule agonists for MOR based on these different design strategies, providing ideas for the development of safer novel opioid ligands with therapeutic potential.


Subject(s)
Analgesics, Opioid , Receptors, Opioid, mu , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Humans , Structure-Activity Relationship , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemistry , Animals , Molecular Structure
7.
ACS Sens ; 9(6): 3198-3204, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38775258

ABSTRACT

Fentanyl is a potent synthetic opioid with an alarmingly low lethal dosage of 2 mg. The equipment necessary to detect fentanyl in field settings (e.g., hand-held spectrometers) is restricted to highly trained, well-funded, and specialized personnel. Established point-of-need technologies, such as lateral flow immunochromatographic strips, are available; however, they often involve multiple contact-based steps (e.g., collection, mixing) that pose a higher risk to users handling unknown substances. Herein, we developed a colorimetric displacement assay capable of contactless detection of fentanyl in liquid or solid samples. The basis of our assay relies on the presence of fentanyl to displace a redox mediator, ferrocene carboxylic acid, inclusively bound in the cavity of a supramolecular host, CB[7]. The displacement is only possible in the presence of high affinity binding guests, like fentanyl (KA ∼ 106 M-1). The liberated redox guest can then react with indicator reagents that are free in solution, producing either: (i) a distinct blue color to indicate the presence of fentanyl or (ii) a pale blue tint in the absence of fentanyl. We demonstrate rapid and specific detection of fentanyl free base and fentanyl derivatives (e.g., acetyl fentanyl and furanyl fentanyl) against a panel of 9 other common drugs of abuse (e.g., morphine, cocaine, and heroin). Furthermore, we highlight the intended use of this assay by testing grains of fentanyl derivatives on a surface with a drop (i.e., 25 µL) of the assay reagent. We anticipate that this approach can be applied broadly to identify the presence of fentanyl at the point of need.


Subject(s)
Colorimetry , Fentanyl , Fentanyl/analysis , Colorimetry/methods , Metallocenes/chemistry , Ferrous Compounds/chemistry , Surface Properties , Limit of Detection , Analgesics, Opioid/analysis , Analgesics, Opioid/chemistry
8.
J Med Chem ; 67(11): 9173-9193, 2024 06 13.
Article in English | MEDLINE | ID: mdl-38810170

ABSTRACT

While in the process of designing more effective synthetic opioid rescue agents, we serendipitously identified a new chemotype of potent synthetic opioid. Here, we report that conformational constraint of a piperazine ring converts a mu opioid receptor (MOR) antagonist into a potent MOR agonist. The prototype of the series, which we have termed atoxifent (2), possesses potent in vitro agonist activity. In mice, atoxifent displayed long-lasting antinociception that was reversible with naltrexone. Repeated dosing of atoxifent produced antinociceptive tolerance and a level of withdrawal like that of fentanyl. In rats, while atoxifent produced complete loss of locomotor activity like fentanyl, it failed to produce deep respiratory depression associated with fentanyl-induced lethality. Assessment of brain biodistribution demonstrated ample distribution of atoxifent into the brain with a Tmax of approximately 0.25 h. These results indicate enhanced safety for atoxifent-like molecules compared to fentanyl.


Subject(s)
Analgesics, Opioid , Fentanyl , Receptors, Opioid, mu , Respiratory Insufficiency , Animals , Mice , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/drug therapy , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/chemistry , Rats , Male , Fentanyl/pharmacology , Fentanyl/chemical synthesis , Fentanyl/chemistry , Structure-Activity Relationship , Piperazines/pharmacology , Piperazines/chemistry , Piperazines/chemical synthesis , Piperazines/therapeutic use , Piperazines/pharmacokinetics , Humans , Rats, Sprague-Dawley , Tissue Distribution , Brain/metabolism , Brain/drug effects , Naltrexone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/chemical synthesis , Naltrexone/chemistry , Naltrexone/therapeutic use
9.
Bioorg Chem ; 148: 107489, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38797065

ABSTRACT

The number of opioid-related overdose deaths and individuals that have suffered from opioid use disorders have significantly increased over the last 30 years. FDA approved maintenance therapies to treat opioid use disorder may successfully curb drug craving and prevent relapse but harbor adverse effects that reduce patient compliance. This has created a need for new chemical entities with improved patient experience. Previously our group reported a novel lead compound, NAT, a mu-opioid receptor antagonist that potently antagonized the antinociception of morphine and showed significant blood-brain barrier permeability. However, NAT belongs to thiophene containing compounds which are known structural alerts for potential oxidative metabolism. To overcome this, 15 NAT derivatives with various substituents at the 5'-position of the thiophene ring were designed and their structure-activity relationships were studied. These derivatives were characterized for their binding affinity, selectivity, and functional activity at the mu opioid receptor and assessed for their ability to antagonize the antinociceptive effects of morphine in vivo. Compound 12 showed retention of the basic pharmacological attributes of NAT while improving the withdrawal effects that were experienced in opioid-dependent mice. Further studies will be conducted to fully characterize compound 12 to examine whether it would serve as a new lead for opioid use disorder treatment and management.


Subject(s)
Receptors, Opioid, mu , Animals , Structure-Activity Relationship , Mice , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/antagonists & inhibitors , Humans , Molecular Structure , Thiophenes/chemistry , Thiophenes/pharmacology , Thiophenes/chemical synthesis , Thiophenes/therapeutic use , Male , Dose-Response Relationship, Drug , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemistry , Narcotic Antagonists/pharmacology , Narcotic Antagonists/chemistry , Morphine/pharmacology
10.
ACS Chem Neurosci ; 15(11): 2091-2098, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38747710

ABSTRACT

Xylazine (also known as "tranq") is a potent nonopioid veterinary sedative that has recently experienced a surge in use as a drug adulterant, most often combined with illicitly manufactured fentanyl. This combination may heighten the risk of fatal overdose. Xylazine has no known antidote approved for use in humans, and age-adjusted overdose deaths involving xylazine were 35 times higher in 2021 than 2018. In April 2023, the Biden Administration declared xylazine-laced fentanyl an emerging drug threat in the United States. In 2022, the Drug Enforcement Agency (DEA) reported nearly a quarter of seized fentanyl powder contained xylazine. This dramatic increase in prevalence has solidified the status of xylazine as an emerging drug of abuse and an evolving threat to public health. The following narrative review outlines the synthesis, pharmacokinetics, pharmacodynamics, and adverse effects of xylazine, as well as the role it may play in the ongoing opioid epidemic.


Subject(s)
Xylazine , Xylazine/pharmacology , Humans , Animals , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/chemistry , Fentanyl/pharmacology , Fentanyl/chemistry , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Drug Overdose/epidemiology
11.
J Control Release ; 370: 490-500, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38685384

ABSTRACT

Misuse of prescription opioid drugs is the leading cause of the opioid crisis and overdose-related death. Abuse deterrent formulations (ADFs) have been developed to discourage attempts to tamper with the formulation and alter the ingestion methods. However, abusers develop complex extraction strategies to circumvent the ADF technologies. For comprehensive deterrence of drug abuse, we develop tannic acid nanoparticles (NPs) that protect encapsulated opioids from solvent extraction and thermal challenge (crisping), complementing the existing formulation strategy to deter injection abuse. Here, we develop a hybrid ADF tablet (NP-Tab), consisting of iron-crosslinked tannic acid NPs encapsulating thebaine (model opioid compound), xanthan gum, and chitosan (gel-forming polymers), and evaluate its performance in common abuse conditions. NP-Tab tampered by crushing and suspended in aqueous solvents forms an instantaneous gel, which is difficult to pull or push through a 21-gauge needle. NPs insulate the drug from organic solvents, deterring solvent extraction. NPs also promote thermal destruction of the drug to make crisping less rewarding. However, NP-Tab releases thebaine in the simulated gastric fluid without delay, suggesting that its analgesic effect may be unaffected if consumed orally as prescribed. These results demonstrate that NP-Tab can provide comprehensive drug abuse deterrence, resisting aqueous/organic solvent extraction, injection, and crisping, while retaining its therapeutic effect upon regular usage.


Subject(s)
Analgesics, Opioid , Chitosan , Nanoparticles , Opioid-Related Disorders , Nanoparticles/chemistry , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/chemistry , Opioid-Related Disorders/prevention & control , Chitosan/chemistry , Animals , Tannins/chemistry , Tannins/administration & dosage , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/administration & dosage , Abuse-Deterrent Formulations , Male , Tablets , Polymers/chemistry
12.
J Med Chem ; 67(9): 7603-7619, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38687204

ABSTRACT

The design of bifunctional compounds is a promising approach toward the development of strong analgesics with reduced side effects. We here report the optimization of the previously published lead peptide KGFF09, which contains opioid receptor agonist and neuropeptide FF receptor antagonist pharmacophores and is shown to induce potent antinociception and reduced side effects. We evaluated the novel hybrid peptides for their in vitro activity at MOP, NPFFR1, and NPFFR2 and selected four of them (DP08/14/32/50) for assessment of their acute antinociceptive activity in mice. We further selected DP32 and DP50 and observed that their antinociceptive activity is mostly peripherally mediated; they produced no respiratory depression, no hyperalgesia, significantly less tolerance, and strongly attenuated withdrawal syndrome, as compared to morphine and the recently FDA-approved TRV130. Overall, these data suggest that MOP agonist/NPFF receptor antagonist hybrids might represent an interesting strategy to develop novel analgesics with reduced side effects.


Subject(s)
Receptors, Neuropeptide , Receptors, Opioid, mu , Animals , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism , Mice , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/metabolism , Male , Analgesics/pharmacology , Analgesics/chemistry , Analgesics/therapeutic use , Analgesics/chemical synthesis , Humans , Structure-Activity Relationship , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemistry
13.
Chem Pharm Bull (Tokyo) ; 72(3): 271-279, 2024.
Article in English | MEDLINE | ID: mdl-38432909

ABSTRACT

Codeine is a common analgesic drug that is a pro-drug of morphine. It also has a high risk of abuse as a recreational drug because of its extensive distribution as an OTC drug. Therefore, sensitive and selective screening methods for codeine are crucial in forensic analytical chemistry. To date, a commercial analytical kit has not been developed for dedicated codeine determination, and there is a need for an analytical method to quantify codeine in the field. In the present work, potential modulation was combined with electrochemiluminescence (ECL) for sensitive determination of codeine. The potential modulated technique involved applying a signal to electrodes by superimposing an AC potential on the DC potential. When tris(2,2'-bipyridine)ruthenium(II) ([Ru(bpy)3]2+) was used as an ECL emitter, ECL activity was confirmed for codeine. A detailed investigation of the electrochemical reaction mechanism suggested a characteristic ECL reaction mechanism involving electrochemical oxidation of the opioid framework. Besides the usual ECL reaction derived from the amine framework, selective detection of codeine was possible under the measurement conditions, with clear luminescence observed in an acidic solution. The sensitivity of codeine detection by potential modulated-ECL was one order of magnitude higher than that obtained with the conventional potential sweep method. The proposed method was applied to codeine determination in actual prescription medications and OTC drug samples. Codeine was selectively determined from other compounds in medications and showed good linearity with a low detection limit (150 ng mL-1).


Subject(s)
Analgesics, Opioid , Codeine , Amines , Analgesics, Opioid/analysis , Analgesics, Opioid/chemistry , Codeine/analysis , Codeine/chemistry , Electrodes , Nonprescription Drugs , Luminescence
14.
J Biochem ; 175(4): 337-355, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38382631

ABSTRACT

Morphinan-based opioids, derived from natural alkaloids like morphine, codeine and thebaine, have long been pivotal in managing severe pain. However, their clinical utility is marred by significant side effects and high addiction potential. This review traces the evolution of the morphinan scaffold in light of advancements in biochemistry and molecular biology, which have expanded our understanding of opioid receptor pharmacology. We explore the development of semi-synthetic and synthetic morphinans, their receptor selectivity and the emergence of biased agonism as a strategy to dissociate analgesic properties from undesirable effects. By examining the molecular intricacies of opioid receptors and their signaling pathways, we highlight how receptor-type selectivity and signaling bias have informed the design of novel analgesics. This synthesis of historical and contemporary perspectives provides an overview of the morphinan landscape, underscoring the ongoing efforts to mitigate the problems facing opioids through smarter drug design. We also highlight that most morphinan derivatives show a preference for the G protein pathway, although detailed experimental comparisons are still necessary. This fact underscores the utility of the morphinan skeleton in future opioid drug discovery.


Subject(s)
Morphinans , Morphinans/chemistry , Morphinans/metabolism , Morphinans/pharmacology , Morphine/pharmacology , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Analgesics, Opioid/chemistry , Molecular Biology
15.
Eur J Pharm Biopharm ; 197: 114230, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38373555

ABSTRACT

Opioid misuse is a public health crisis in the United States. In response, the FDA has approved drug products with abuse-deterrent features to reduce the risk of prescription opioid abuse. Abuse-deterrent formulations (ADFs) typically employ physical or chemical barriers or incorporate agonist-antagonist combinations as mechanisms to deter misuse. This study aims to assess the impact of abuse-deterrent properties, specifically ion-exchange resin complexation as a chemical barrier, on a model drug, promethazine hydrochloride (PMZ) tablets. Various formulations were developed through twin-screw wet granulation (TSWG) followed by twin-screw melt granulation (TSMG). In the TSWG process, the drug interacts with the resin through an exchange reaction, forming a drug-resin complex. Additionally, the study explored factors influencing the complex formation between the drug and resin, using the drug loading status as an indicator. DSC and ATR studies were carried out to confirm the formation of the drug-resin complex. Subsequently, hot melt granulation was employed to create a matrix tablet incorporating Kollidon® SR and Kollicoat® MAE 100P, thereby enabling sustained release properties. The drug-resin complex embedded in the matrix effectively deters abuse through methods like smoking, snorting, or parenteral injection, unless the drug can be extracted. In order to assess this, solvent extraction studies were conducted using an FDA-recommended solvents, determining the potential for abuse. Further investigations involved dissolution tests in change-over media, confirming the extended-release properties of the formulation. Results from dissolution studies comparing the ground and intact tablets provided positive evidence of the formulation's effectiveness in deterring abuse. Finally, alcohol-induced dose-dumping studies were conducted in compliance with FDA guidelines, concluding that the formulation successfully mitigates dose dumping in the presence of alcohol.


Subject(s)
Abuse-Deterrent Formulations , Opioid-Related Disorders , Humans , Analgesics, Opioid/chemistry , Analgesics, Opioid/therapeutic use , Opioid-Related Disorders/drug therapy , Drug Compounding , Delayed-Action Preparations
16.
Drug Test Anal ; 16(3): 323-326, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37482925

ABSTRACT

New synthetic opioids (NSOs) with diverse chemical structures continue to appear on recreational drug markets worldwide. U-type opioids have become one of the largest groups of non-fentanyl-related NSOs. Starting in 2020, a previously unreported U-compound coined "ß-U10" (2-naphthyl U-47700; N-[2-(dimethylamino)cyclohexyl]-N-methylnaphthalene-2-carboxamide) was identified in Australia and the United States. ß-U10 is a positional isomer of α-U10 (1-naphthyl U-47700), more commonly known as "U10." Here, the first comparative in vitro pharmacological characterization of naphthyl U-47700 (U10 and ß-U10), together with the structural analogue U-47700 and fentanyl, is reported. Application of a cell-based µ-opioid receptor (MOR) activation (ß-arrestin 2 recruitment) assay demonstrated ß-U10 (EC50 = 348 nM; Emax = 150% vs. hydromorphone) to be less potent than U-47700 (EC50 = 116 nM; Emax = 154%) and fentanyl (EC50 = 9.35 nM; Emax = 146%) but considerably more active than the α-isomer (EC50 value in the µM range). For the latter, maximum receptor activation could not be reached at 100 µM. The difference in MOR activation potential for U10 and ß-U10 stresses the importance of (analytical) differentiation between closely related analytes. The emergence of ß-U10 on the recreational drug market is an example of the continuing emergence of non-fentanyl-related NSOs and further emphasizes the need to closely monitor fluctuations in the drug supply.


Subject(s)
Analgesics, Opioid , Illicit Drugs , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemistry , Benzamides , Fentanyl/pharmacology , Illicit Drugs/pharmacology
17.
Nat Commun ; 14(1): 8064, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38052802

ABSTRACT

Despite the increasing number of GPCR structures and recent advances in peptide design, the development of efficient technologies allowing rational design of high-affinity peptide ligands for single GPCRs remains an unmet challenge. Here, we develop a computational approach for designing conjugates of lariat-shaped macrocyclized peptides and a small molecule opioid ligand. We demonstrate its feasibility by discovering chemical scaffolds for the kappa-opioid receptor (KOR) with desired pharmacological activities. The designed De Novo Cyclic Peptide (DNCP)-ß-naloxamine (NalA) exhibit in vitro potent mixed KOR agonism/mu-opioid receptor (MOR) antagonism, nanomolar binding affinity, selectivity, and efficacy bias at KOR. Proof-of-concept in vivo efficacy studies demonstrate that DNCP-ß-NalA(1) induces a potent KOR-mediated antinociception in male mice. The high-resolution cryo-EM structure (2.6 Å) of the DNCP-ß-NalA-KOR-Gi1 complex and molecular dynamics simulations are harnessed to validate the computational design model. This reveals a network of residues in ECL2/3 and TM6/7 controlling the intrinsic efficacy of KOR. In general, our computational de novo platform overcomes extensive lead optimization encountered in ultra-large library docking and virtual small molecule screening campaigns and offers innovation for GPCR ligand discovery. This may drive the development of next-generation therapeutics for medical applications such as pain conditions.


Subject(s)
Analgesics, Opioid , Receptors, Opioid, kappa , Male , Mice , Animals , Receptors, Opioid, kappa/metabolism , Ligands , Analgesics, Opioid/chemistry , Receptors, Opioid, mu/metabolism , Peptides, Cyclic/chemistry
18.
Molecules ; 28(19)2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37836768

ABSTRACT

In medicinal chemistry, the copper-catalyzed click reaction is used to prepare ligand candidates. This reaction is so clean that the bioactivities of the products can be determined without purification. Despite the advantages of this in situ screening protocol, the applicability of this method for transmembrane proteins has not been validated due to the incompatibility with copper catalysts. To address this point, we performed ligand screening for the µ, δ, and κ opioid receptors using this protocol. As we had previously reported the 7-azanorbornane skeleton as a privileged scaffold for the G protein-coupled receptors, we performed the click reactions between various 7-substituted 2-ethynyl-7-azanorbornanes and azides. Screening assays were performed without purification using the CellKeyTM system, and the putative hit compounds were re-synthesized and re-evaluated. Although the "hit" compounds for the µ and the δ receptors were totally inactive after purifications, three of the four "hits" for the κ receptor were true agonists for this receptor and also showed activities for the δ receptor. Although false positive/negative results exist as in other screening projects for soluble proteins, this in situ method is effective in identifying novel ligands for transmembrane proteins.


Subject(s)
Copper , Receptors, Opioid, kappa , Receptors, Opioid, kappa/metabolism , Ligands , Membrane Proteins , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/chemistry
19.
J Med Chem ; 66(15): 10304-10341, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37467430

ABSTRACT

A new generation of dual-target µ opioid receptor (MOR) agonist/dopamine D3 receptor (D3R) antagonist/partial agonists with optimized physicochemical properties was designed and synthesized. Combining in vitro cell-based on-target/off-target affinity screening, in silico computer-aided drug design, and BRET functional assays, we identified new structural scaffolds that achieved high affinity and agonist/antagonist potencies for MOR and D3R, respectively, improving the dopamine receptor subtype selectivity (e.g., D3R over D2R) and significantly enhancing central nervous system multiparameter optimization scores for predicted blood-brain barrier permeability. We identified the substituted trans-(2S,4R)-pyrrolidine and trans-phenylcyclopropyl amine as key dopaminergic moieties and tethered these to different opioid scaffolds, derived from the MOR agonists TRV130 (3) or loperamide (6). The lead compounds 46, 84, 114, and 121 have the potential of producing analgesic effects through MOR partial agonism with reduced opioid-misuse liability via D3R antagonism. Moreover, the peripherally limited derivatives could have therapeutic indications for inflammation and neuropathic pain.


Subject(s)
Analgesics, Opioid , Opioid-Related Disorders , Humans , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemistry , Dopamine , Ligands , Analgesics/pharmacology , Receptors, Dopamine D3/agonists , Receptors, Opioid, mu/agonists
20.
Leg Med (Tokyo) ; 64: 102295, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37418781

ABSTRACT

Benzimidazole opioids were originally developed from the late 1950s to 1970s as analgesics for medical use, although a lot of them could not be approved as licit medicines because of their severe side effects and physical dependence. Such benzimidazole opioid analogs as abused drug, however, have recently been found in illicit drug markets throughout the world. Isotonitazene is one such benzimidazole opioids, whose analgesic potency can be as much as 500 times greater than that of morphine, according to previous animal studies. In line with this potency, a couple of hundred fatalities related to it were reported to date. In this study, a well validated method for the quantification of isotonitazene in human hair samples using liquid chromatography (LC)-tandem mass spectrometry (MS/MS) was established, and could be applied to authentic samples which were seized by the police security bureau. Isotonitazene concentrations in the seized hair averaged 6.11 pg/mg. The LLOQ and LOD of this method were 1.25 and 2.5 pg/mg, respectively; the calibration curve of the substance in hair samples showed a good linearity in the concentration range of 2.5-250 pg/mg (r > 0.999); the extraction recovery rates were 87.3-105% in the tested range; the inter- and intra-day precisions and accuracies (%biases) were not greater than 9.09% for each determination. Isotonitazene in human hair showed good stability at room temperature and under dark storage conditions for 30 days. As for matrix effect in hair samples, moderate ion suppression of target substances could be found. This is the first report for the analysis of isotonitazene in human hair samples.


Subject(s)
Analgesics, Opioid , Illicit Drugs , Animals , Humans , Analgesics, Opioid/analysis , Analgesics, Opioid/chemistry , Tandem Mass Spectrometry/methods , Chromatography, Liquid/methods , Benzimidazoles/analysis , Illicit Drugs/analysis , Hair/chemistry , Substance Abuse Detection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...