Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.298
Filtrar
1.
Int J Nanomedicine ; 19: 4759-4777, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38828199

RESUMEN

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.


Asunto(s)
Analgésicos Opioides , Ganglios Espinales , Morfina , Nanopartículas , Animales , Morfina/administración & dosificación , Morfina/farmacocinética , Morfina/química , Morfina/farmacología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Nanopartículas/química , Ratas , Células PC12 , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/química , Analgésicos Opioides/farmacología , Masculino , Neuralgia/tratamiento farmacológico , Ratones , Lípidos/química , Proteínas Proto-Oncogénicas/metabolismo , Nervios Periféricos/efectos de los fármacos , Oxigenasas de Función Mixta/metabolismo , Proteínas de Unión al ADN , Liposomas
2.
Biomed Pharmacother ; 175: 116665, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38701564

RESUMEN

Opioid receptor agonists are often used when cancer patients undergo surgery or analgesic treatment. As analgesics in clinical care, opioids can provide intraoperative or to chronic cancer pain relief. Immune function plays an important role in anti-cancer therapy, with cellular immunity, comprised principally of T-lymphocytes and natural killer cells, representing the primary anti-cancer immune response. However, it remains unclear whether immune function is further affected with the use of opioids in already immunocompromised cancer patients. This article provides a review of the effects of commonly used clinical opioids, including morphine, oxycodone, fentanyl and tramadol, on immune function in cancer patients. It provides a summary of current evidence regarding the immunomodulatory effects of opioids in the cancer setting and mechanisms underlying these interactions.


Asunto(s)
Analgésicos Opioides , Neoplasias , Humanos , Analgésicos Opioides/uso terapéutico , Analgésicos Opioides/farmacología , Neoplasias/inmunología , Neoplasias/tratamiento farmacológico , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/inmunología , Animales , Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos
3.
Drugs ; 84(6): 717-720, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38771484

RESUMEN

Tegileridine () is a small molecule µ-opioid receptor biased agonist developed by Jiangsu Hengrui Pharmaceuticals Co., Ltd for the treatment of postoperative pain. Tegileridine selectively activates the G-protein-coupled pathway, which mediates strong central analgesic effects and only weakly activates the ß-arrestin-2 pathway implicated in adverse events like respiratory depression and gastrointestinal dysfunction. In January 2024, tegileridine received its first approval in China for the treatment of moderate to severe pain after abdominal surgery. This article summarizes the milestones in the development of tegileridine leading to this first approval for the treatment of moderate to severe pain after abdominal surgery.


Asunto(s)
Aprobación de Drogas , Dolor Postoperatorio , Receptores Opioides mu , Humanos , Dolor Postoperatorio/tratamiento farmacológico , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacología , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/uso terapéutico , China , Tiofenos/farmacología , Tiofenos/uso terapéutico , Tiofenos/efectos adversos , Compuestos de Espiro
4.
Molecules ; 29(7)2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38611824

RESUMEN

Pain affects one-third of the global population and is a significant public health issue. The use of opioid drugs, which are the strongest painkillers, is associated with several side effects, such as tolerance, addiction, overdose, and even death. An increasing demand for novel, safer analgesic agents is a driving force for exploring natural sources of bioactive peptides with antinociceptive activity. Since the G protein-coupled receptors (GPCRs) play a crucial role in pain modulation, the discovery of new peptide ligands for GPCRs is a significant challenge for novel drug development. The aim of this review is to present peptides of human and animal origin with antinociceptive potential and to show the possibilities of their modification, as well as the design of novel structures. The study presents the current knowledge on structure-activity relationship in the design of peptide-based biomimetic compounds, the modification strategies directed at increasing the antinociceptive activity, and improvement of metabolic stability and pharmacodynamic profile. The procedures employed in prolonged drug delivery of emerging compounds are also discussed. The work summarizes the conditions leading to the development of potential morphine replacements.


Asunto(s)
Analgésicos , Péptidos , Animales , Humanos , Analgésicos/farmacología , Analgésicos/uso terapéutico , Péptidos/farmacología , Morfina , Dolor , Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico
5.
Biol Pharm Bull ; 47(4): 872-877, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38658360

RESUMEN

The formalin test is one approach to studying acute pain in rodents. Similar to formalin, injection with glutamate and veratrine can also produce a nociceptive response. This study investigated whether opioid-related compounds could suppress glutamate- and veratrine-induced nociceptive responses in mice at the same dose. The administration of morphine (3 mg/kg), hydromorphone (0.4 mg/kg), or fentanyl (0.03 mg/kg) suppressed glutamate-induced nociceptive response, but not veratrine-induced nociceptive response at the same doses. However, high doses of morphine (10 mg/kg), hydromorphone (2 mg/kg), or fentanyl (0.1 mg/kg) produced a significant reduction in the veratrine-induced nociceptive response. These results indicate that high doses are required when using morphine, hydromorphone, or fentanyl for sodium channel-related neuropathic pain, such as ectopic activity. As a result, concerns have arisen about overdose and abuse if the dose of opioids is steadily increased to relieve pain. In contrast, trimebutine (100 mg/kg) and fentanyl analog isobutyrylfentanyl (iBF; 0.1 mg/kg) suppressed both glutamate- and veratrine-induced nociceptive response. Furthermore, nor-isobutyrylfentanyl (nor-iBF; 1 mg/kg), which is a metabolite of iBF, suppressed veratrine-induced nociceptive response. Besides, the optimal antinociceptive dose of iBF, unlike fentanyl, only slightly increased locomotor activity and did not slow gastrointestinal transit. Cancer pain is a complex condition driven by inflammatory, neuropathic, and cancer-specific mechanisms. Thus, iBF may have the potential to be a superior analgesic than fentanyl.


Asunto(s)
Analgésicos Opioides , Fentanilo , Animales , Fentanilo/farmacología , Fentanilo/análogos & derivados , Masculino , Ratones , Analgésicos Opioides/farmacología , Ácido Glutámico/metabolismo , Bloqueadores de los Canales de Sodio/farmacología , Bloqueadores de los Canales de Sodio/uso terapéutico , Analgésicos/farmacología , Analgésicos/uso terapéutico , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Morfina/farmacología
6.
Pharmacol Biochem Behav ; 240: 173771, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38670466

RESUMEN

In the United States, the societal costs associated with drug use surpass $500 billion annually. The rewarding and reinforcing properties that drive the use of these addictive substances are typically examined concerning the neurobiological effects responsible for their abuse potential. In this review, terms such as "abuse potential," "drug," and "addictive properties" are used due to their relevance to the methodological, theoretical, and conceptual framework for understanding the phenomenon of drug-taking behavior and the associated body of preclinical and clinical literature. The use of these terms is not intended to cast aspersions on individuals with substance use disorders (SUD). Understanding what motivates substance use has been a focus of SUD research for decades. Much of this corpus of work has focused on the shared effects of each drug class to increase dopaminergic transmission within the central reward pathways of the brain, or the "reward center." However, the precise influence of each drug class on dopamine signaling, and the extent thereof, differs considerably. Furthermore, the aforementioned substances have effects on several neurobiological targets that mediate and modulate their addictive properties. The current manuscript sought to review the influence of drug class on the rewarding effects of each of the major pharmacological classes of addictive drugs (i.e., psychostimulants, opioids, nicotine, alcohol, and cannabinoids). Our review suggests that even subtle differences in drug effects can result in significant variability in the subjective experience of the drug, altering rewarding and other reinforcing effects. Additionally, this review will argue that reward (i.e., the attractive and motivational property of a stimulus) alone is not sufficient to explain the abuse liability of these substances. Instead, abuse potential is best examined as a function of both positive and negative reinforcing drug effects (i.e., stimuli that the subject will work to attain and stimuli that the subject will work to end or avoid, respectively). Though reward is central to drug use, the factors that motivate and maintain drug taking are varied and complex, with much to be elucidated.


Asunto(s)
Recompensa , Trastornos Relacionados con Sustancias , Humanos , Trastornos Relacionados con Sustancias/psicología , Animales , Conducta Adictiva/psicología , Dopamina/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cannabinoides/farmacología , Motivación , Nicotina/farmacología , Refuerzo en Psicología , Analgésicos Opioides/farmacología , Cocaína/farmacología
7.
Drug Discov Today ; 29(5): 103950, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38514040

RESUMEN

Drugs targeting the µ-opioid receptor (MOR) remain the most efficacious analgesics for the treatment of pain, but activation of MOR with current opioid analgesics also produces harmful side effects, notably physical dependence, addiction, and respiratory depression. Opioid peptides have been accepted as promising candidates for the development of safer and more efficacious analgesics. To develop peptide-based opioid analgesics, strategies such as modification of endogenous opioid peptides, development of multifunctional opioid peptides, G protein-biased opioid peptides, and peripherally restricted opioid peptides have been reported. This review seeks to provide an overview of the opioid peptides that produce potent antinociception with much reduced side effects in animal models and highlight the potential advantages of peptides as safer opioid analgesics.


Asunto(s)
Analgésicos Opioides , Descubrimiento de Drogas , Péptidos Opioides , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacología , Animales , Humanos , Ligandos , Descubrimiento de Drogas/métodos , Dolor/tratamiento farmacológico , Receptores Opioides mu/metabolismo , Péptidos/farmacología , Péptidos/uso terapéutico
8.
Top Companion Anim Med ; 59: 100861, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38508490

RESUMEN

Pre-emptive analgesia consists of administering drugs such as opioids and nonsteroid anti-inflammatory drugs. This study aims to evaluate the intraoperative antinociceptive effects of diclofenac administered alone in premedication or combined with morphine along with its potential influence on recovery of dogs undergoing ovariohysterectomy. A total of 34 dogs (ASA I or II) admitted for ovariohysterectomy were randomly allocated into three groups according to the drugs given in premedication: Diclofenac (D) (n = 11), Morphine (M) (n = 13) and Diclofenac-Morphine (DM) (n = 10) groups. Induction and maintenance of anesthesia were standardized in all dogs. To assess intraoperative nociception, the heart rate (HR) and mean arterial pressure (MAP) were recorded during the surgery and at predefined time points: St (steady-state), Cut (cutaneous incision), P1 (first ovarian manipulation), P2 (second ovarian manipulation) and Cerv (cervical manipulation). The dynamic variation of HR (ΔHR) and MAP (ΔMAP) over 2 min was calculated at each time point. After extubation, early quality of recovery was assessed. Compared to St, a significant increase in HR and MAP at P1, P2 and Cerv was shown in all groups. MAP in the M group was lower at St than in the other groups. The dynamic variation of HR (ΔHR) and MAP (ΔMAP) was significantly less important at P2 and Cerv compared to P1 only in the DM group. Also, a better quality of recovery was shown in the D group compared to the M and DM groups. Diclofenac may be considered a suitable premedication drug and a part of a multimodal anesthetic approach in dogs.


Asunto(s)
Analgésicos Opioides , Diclofenaco , Animales , Perros , Femenino , Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Diclofenaco/farmacología , Histerectomía/veterinaria , Morfina/farmacología , Ovariectomía/veterinaria , Premedicación/veterinaria , Distribución Aleatoria
9.
Spine (Phila Pa 1976) ; 49(12): 821-828, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38348858

RESUMEN

STUDY DESIGN: Translational research. OBJECTIVE: To evaluate the relative effects of NSAIDs, opioids, and a combination of the two on spinal fusion inhibition in a rodent model. SUMMARY OF BACKGROUND DATA: Nonsteroidal anti-inflammatory drugs (NSAIDs) and opioids are common postoperative analgesic agents. Since NSAIDs inhibit the cyclooxygenase (COX) pathway, they are seldom prescribed following spinal fusion. Opioids may be given instead, but recent evidence suggests opioids also adversely affect spinal fusion quality and success. METHODS: Eighty male Sprague-Dawley rats underwent L4-5 posterior lumbar fusion and were given one of the following analgesia regimens: saline, morphine (6 mg/kg), ketorolac (4 mg/kg), or morphine (3 mg/kg) and ketorolac (2 mg/kg). Serum samples were drawn to evaluate systemic pro-osteoblastic cytokines and vascular endothelial growth factor-A (VEGF-A) levels, which were measured through enzyme-linked immunosorbent assays (ELISA). After six weeks, the rats were sacrificed, and the operated spinal segments underwent manual palpation, microCT, and histologic analysis. RESULTS: Manual palpation scores were significantly diminished in the opioid, NSAID, and multimodal groups when compared with control ( P <0.001). MicroCT fusion scores ( P <0.001) and fusion rates (control: 75% vs . NSAID: 35% vs . opioid: 0% vs . combination: 15%, P <0.001) were significantly diminished in the treatment groups. The bone volume (BV) to tissue volume (TV) ratio (BV/TV) ( P <0.001) and bone mineral density (BMD) ( P <0.001) were all lower in the treatment groups, with the opioid and combined groups having the lowest BMD. Although statistically insignificant ( P <0.09), the concentration of VEGF-A was greater in the control group compared with opioids, NSAIDs, and the combined group. CONCLUSION: Opioids and NSAIDs, both independently and combined, inhibited spinal fusion and caused inferior bony callus. Administration of opioids resulted in the lowest rate of spinal fusion. We propose this may be due to the inhibition of VEGF-A, which limits angiogenesis to the burgeoning fusion mass.


Asunto(s)
Analgésicos Opioides , Antiinflamatorios no Esteroideos , Ratas Sprague-Dawley , Fusión Vertebral , Animales , Fusión Vertebral/métodos , Masculino , Antiinflamatorios no Esteroideos/farmacología , Analgésicos Opioides/farmacología , Ratas , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ketorolaco/farmacología , Vértebras Lumbares/efectos de los fármacos , Morfina/farmacología
10.
J Anesth ; 38(3): 371-376, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38376589

RESUMEN

PURPOSE: The effect-site concentration (Ce) at loss of response (Ce-LOR) to propofol closely correlates both with Ce as electroencephalographic alpha power becomes highest (Ce-alpha) and with Ce at onset of burst suppression (BS) (Ce-OBS), when no opioids are administered. Co-administration of opioids dose-dependently decreases Ce-LOR. We investigated the influence of remifentanil on the relationship between these three Ces. METHODS: After receiving approval from our local ethical committee, with written informed consent, we enrolled 90 participants (ASA-PS I or II) who were scheduled for elective surgery. Participants were randomly assigned to three groups: constant remifentanil Ce 0 ng/ml (Remi_0); 1 ng/mL (Remi_1); and 2 ng/mL (Remi_2). We recorded both raw EEG and EEG-derived parameters on a computer. After reaching remifentanil equilibrium, we administered propofol using a target-controlled infusion pump such that propofol Ce increased to about 0.3 µg/mL/min. After determining Ce-LOR, we administered 0.6 mg/kg of rocuronium and started mask ventilation. The study protocol ended after observation of BS. RESULTS: Three participants were excluded. Ce-LOR in each group (Remi_0, Remi_1, Remi_2) was 2.00 ± 0.58 µg/mL, 1.43 ± 0.49 µg/mL, and 1.37 ± 0.42 µg/mL. Ce-alpha was 2.91 ± 0.63 µg/mL, 2.30 ± 0.41 µg/mL, and 2.12 ± 0.39 µg/mL. Ce-OBS was 3.80 ± 0.69 µg/mL, 3.25 ± 0.68 µg/mL, and 2.90 ± 0.57 µg/mL. In three other instances, Ce was decreased by remifentanil. Generalized linear model analysis revealed that remifentanil had no influence on the relationship between the three Ces. CONCLUSION: During propofol anesthesia, even low concentrations of remifentanil shifted concentration-related electroencephalographic changes.


Asunto(s)
Anestésicos Intravenosos , Electroencefalografía , Propofol , Remifentanilo , Humanos , Remifentanilo/administración & dosificación , Remifentanilo/farmacología , Propofol/administración & dosificación , Propofol/farmacología , Electroencefalografía/efectos de los fármacos , Electroencefalografía/métodos , Masculino , Femenino , Anestésicos Intravenosos/administración & dosificación , Anestésicos Intravenosos/farmacología , Estudios Prospectivos , Adulto , Persona de Mediana Edad , Relación Dosis-Respuesta a Droga , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacología , Piperidinas/farmacología , Piperidinas/administración & dosificación
11.
Int J Mol Sci ; 25(4)2024 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-38396814

RESUMEN

Methadone is an effective and long-lasting analgesic drug that is also used in medication-assisted treatment for people with opioid use disorders. Although there is evidence that methadone activates µ-opioid and Toll-like-4 receptors (TLR-4s), its effects on distinct immune cells, including mast cells (MCs), are not well characterized. MCs express µ-opioid and Toll-like receptors (TLRs) and constitute an important cell lineage involved in allergy and effective innate immunity responses. In the present study, murine bone-marrow-derived mast cells (BMMCs) were treated with methadone to evaluate cell viability by flow cytometry, cell morphology with immunofluorescence and scanning electron microscopy, reactive oxygen species (ROS) production, and intracellular calcium concentration ([Ca2+]i) increase. We found that exposure of BMMCs to 0.5 mM or 1 mM methadone rapidly induced cell death by forming extracellular DNA traps (ETosis). Methadone-induced cell death depended on ROS formation and [Ca2+]i. Using pharmacological approaches and TLR4-defective BMMC cultures, we found that µ-opioid receptors were necessary for both methadone-induced ROS production and intracellular calcium increase. Remarkably, TLR4 receptors were also involved in methadone-induced ROS production as it did not occur in BMMCs obtained from TLR4-deficient mice. Finally, confocal microscopy images showed a significant co-localization of µ-opioid and TLR4 receptors that increased after methadone treatment. Our results suggest that methadone produces MCETosis by a mechanism requiring a novel crosstalk pathway between µ-opioid and TLR4 receptors.


Asunto(s)
Analgésicos Opioides , Trampas Extracelulares , Humanos , Animales , Ratones , Analgésicos Opioides/farmacología , Receptor Toll-Like 4/metabolismo , Metadona/farmacología , Mastocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Médula Ósea/metabolismo , Calcio/metabolismo , Trampas Extracelulares/metabolismo , Receptor Toll-Like 2/metabolismo , Receptores Toll-Like/metabolismo
12.
JCO Oncol Pract ; 20(2): 268-277, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38061003

RESUMEN

PURPOSE: Opioid prescribing trends in medical oncology are poorly defined past 2017, the year after the CDC updated opioid prescription guidelines in noncancer settings. We aim to characterize pain management by medical oncologists by analyzing opioid and gabapentin prescribing trends from 2013 to 2019, identify physician-related factors associated with prescribing patterns, and assess whether CDC guidelines for nononcologic settings changed prescribing patterns. METHODS: The Centers for Medicare & Medicaid Services (CMS) Medicare Part D Prescribers-by Provider, CMS Medicare Part D Prescribers-by Provider and Drug, and CMS Medicare Physician National Downloadable files from 2013 to 2019 were merged by National Provider Identification. The database included physicians' sex, years of practice, regions, and practice settings. Multivariable binary logistic regression identified significant predictors of total opioid, long-acting opioid, and gabapentin prescriptions. RESULTS: Binary logistic regression modeling revealed no significant difference in mean daily total opioid prescriptions from 2013 to 2017. Daily opioid prescriptions by medical oncologists decreased significantly after 2017 (P < .001). Increased opioid prescribing was associated with physician male sex (P < .001), practicing over 10 years (P < .001), and practice in nonurban areas (P < .001). Opioid prescribing was greatest in the South and Midwest United States (P < .001). The same patterns were observed with total long-acting opioid prescriptions, whereas gabapentin prescribing increased from 2013 to 2019 (P < .001). CONCLUSION: Opioid prescriptions by medical oncologists decreased significantly from 2013 to 2019, but this decrease was most substantial from 2017 to 2019. These results may imply that the 2016 CDC guidelines influenced medical oncologists, particularly more junior physicians in urban settings, to manage chronic cancer pain with alternative therapies.


Asunto(s)
Medicare Part D , Oncólogos , Anciano , Masculino , Humanos , Estados Unidos , Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Medicaid , Gabapentina/farmacología , Gabapentina/uso terapéutico , Pautas de la Práctica en Medicina
13.
J Ocul Pharmacol Ther ; 40(1): 34-47, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37878373

RESUMEN

Purpose: Previous literature has investigated opioid prescription trends in ophthalmology at large, however, little has been done looking at differences between subspecialties. We evaluate if significant trends exist among subspecialties in opioid prescribing patterns. This study aims to illuminate potential over-usage of opioids in ophthalmology that could compromise patient quality of life. Methods: Medicare data and "National Plan and Provider Enumeration System (NPPES) Downloadable File" were queried for cases of ophthalmologists with nonsuppressed opioid prescription data from 2014 to 2019. Ophthalmologists with no subspecialty code or missing regional, gender, degree, or graduation information were excluded. Chi-squared analysis, analysis of variance, t-tests, and multivariate logistic regression were utilized. Results: Five thousand one hundred forty-three physician records were included in analysis, 450 of which were by cornea subspecialists. Most cornea cases were male, graduated before 2005, and practiced in the South. All subspecialties had a significantly increased likelihood of making opioid claims and higher prescription rates compared with cornea (P < 0.050) besides glaucoma (P = 0.357). Only oculoplastics had significantly increased likelihood of greater total supply of opioids compared with cornea (odds ratio [OR] = 22.195, 95% confidence interval [CI] = 12.209-40.350, P < 0.001), while pediatrics (OR = 4.036, 95% CI = 1.377-11.831, P = 0.011) and neuro-ophthalmology (OR = 4.158, 95% CI = 1.237-13.975, P = 0.021) in addition to oculoplastics (OR = 64.380, 95% CI = 26.306-157.560, P < 0.001) were predicted to have significantly greater opioid beneficiaries. Males, the South/Midwest, and graduating before 2005, all were generally associated with increased likelihood of greater total opioid claims, supply, beneficiaries, and prescription rate (P < 0.050). Conclusion: Subspecialty, demographic, chronological, and regional trends exist for opioid prescribing patterns in ophthalmology.


Asunto(s)
Analgésicos Opioides , Oftalmología , Humanos , Masculino , Anciano , Estados Unidos , Niño , Femenino , Analgésicos Opioides/uso terapéutico , Analgésicos Opioides/farmacología , Salud Pública , Calidad de Vida , Medicare , Pautas de la Práctica en Medicina , Estudios Retrospectivos
14.
Eur J Pain ; 28(3): 359-368, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37947151

RESUMEN

BACKGROUND: Opioid dose titration is a fundamental process of opioid therapy in cancer pain. AIMS: To assess data opioid dose titration. METHODS: The principal opioid dose titration methods, outcomes, and modalities of administration regarding the different opioid preparations were examined in different clinical contexts. RESULTS: Most studies suggested that opioid-naive patients should be started at doses of 15-30 mg/day of oral morphine equivalents. Opioid-tolerant patients may receive low or higher doses of oral morphine equivalents, depending on the level of opioid tolerance. Generally, dose increments of 30%-50% seem to be indicated to start dose titration. Some patients with severe excruciating cancer pain may present as an emergency requiring a rapid application of powerful analgesic strategies. The intravenous use of opioids may circumvent this problem providing a faster pain relief, due to the large availability and rapid achievement of effective plasma concentrations. DISCUSSION: Opioid dose titration is a delicate passage in patients with cancer pain. This approach may be different according to different clinical conditions. Opioid dose titration requires expertise to optimize cancer pain management while minimizing the development of adverse effects. CONCLUSION: While most approaches are meaningful and partially supported by existing literature, more studies are necessary to establish advantages and disadvantages in different clinical conditions. Optimization of opioid dose titration is of paramount importance. SIGNIFICANCE: This review provides the most recent insights on the different modalities of opioid dose titration in cancer pain management.


Asunto(s)
Dolor en Cáncer , Neoplasias , Humanos , Analgésicos Opioides/farmacología , Dolor en Cáncer/tratamiento farmacológico , Tolerancia a Medicamentos , Morfina/uso terapéutico , Dolor/etiología , Dolor/inducido químicamente , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico
16.
Behav Neurosci ; 138(1): 59-71, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38127524

RESUMEN

Drug exposure during adolescence, when the "reward" circuitry of the brain is developing, can permanently impact reward-related behavior into adulthood. Epidemiological studies show that opioid treatment during adolescence, such as pain management for a dental procedure or surgery, increases the incidence of psychiatric illness including substance use disorders. Moreover, the opioid epidemic currently in the United States is affecting younger individuals raising the impetus to understand the pathogenesis of the negative effects of opioids. One reward-related behavior that develops during adolescence is social behavior. We previously demonstrated that developmental changes in the nucleus accumbens reward region regulate social development in rats during sex-specific adolescent periods: early to mid-adolescence in males (postnatal day, P30-40) and preearly adolescence in females (P20-30). We thus hypothesized that the developmental stage of morphine exposure will differentially impact social behavior development such that drug administered during the female critical period would result in adult sociability deficits in females, but not males, and morphine administered during the male critical period would result in adult sociability deficits in males, but not females. We found that morphine exposure during the female critical period primarily resulted in deficits in sociability in females, while morphine exposure during the male critical period primarily resulted in deficits in sociability primarily in males. However, depending on the test performed and the social parameter measured, social alterations could be found in both sexes that received morphine exposure at either adolescent stage. These data indicate that when drug exposure occurs during adolescence, and how the endpoint data are measured, will play a large role in determining the effects of drug exposures on social development. (PsycInfo Database Record (c) 2024 APA, all rights reserved).


Asunto(s)
Morfina , Cambio Social , Ratas , Femenino , Masculino , Animales , Morfina/farmacología , Analgésicos Opioides/farmacología , Conducta Social , Núcleo Accumbens
17.
J Avian Med Surg ; 37(3): 209-216, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37962314

RESUMEN

Across the Americas, great horned owls (Bubo virginianus) are often presented to veterinarians for conditions requiring pain management. Although recent studies have evaluated opioid drugs in raptor species, information in Strigiformes is lacking. The objective of this study was to evaluate the analgesic effect and duration of action of hydromorphone hydrochloride, a full µ-opioid receptor agonist, in great horned owls. In a randomized, blinded, balanced crossover study, 6 adult birds (5 females and 1 male) received hydromorphone (0.3 and 0.6 mg/kg) or saline (0.9% NaCl) solution (0.03 mL/kg; control) in the left pectoral muscle, with a 7-day washout interval between treatments. Each bird was assigned an agitation-sedation score, and the thermal foot withdrawal threshold (TFWT) was measured at predetermined times before (t = 0 hours) and after treatment administration (t = 0.5, 1.5, 3, and 6 hours). Measurements of the TFWT were obtained with a test box equipped with a thermal perch, which delivered a gradually increasing temperature 40-62°C (104-143.6°F) to the right plantar surface of the owl's foot. Compared with controls, hydromorphone at 0.3 mg/kg dose resulted in significantly higher mean TFWT at 0.5 hours (P < 0.001), 1.5 hours (P = 0.003), and 3 hours (P = 0.005), whereas the 0.6 mg/kg dose resulted in significantly higher mean TFWT from 0.5 hours (P = 0.035) to 1.5 hours (P = 0.001). Both hydromorphone doses were associated with a significant change in the agitation-sedation score (P = 0.001), consistent with mild to moderate sedation. Two owls were observed tremoring after administration of the 0.6 mg/kg dose, which was not noted after the 0.5-hour timepoint; no other adverse effects were identified. This study offers scientific evidence to support the use of a µ-opioid agonist in great horned owls for pain management. Pharmacokinetics and other pharmacodynamic studies of other pain models evaluating hydromorphone and other opioid drugs in this species are still needed.


Asunto(s)
Hidromorfona , Estrigiformes , Animales , Femenino , Masculino , Analgésicos Opioides/farmacología , Estudios Cruzados , Hidromorfona/farmacología
18.
Neuroreport ; 34(18): 853-859, 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-37942736

RESUMEN

The interaction between the µ opioid receptor (MOR) and ß-arrestin2 serves as a model for addressing morphine tolerance. A peptide was designed to alleviate morphine tolerance through interfering with the interaction of MOR and ß-arrestin2. We developed a peptide derived from MOR. The MOR-TAT-pep peptide was expressed in E. coli Bl21(DE3) and purified. The effects of MOR-TAT-pep in alleviating morphine tolerance was examined through behavior tests. The potential mechanism was detected by Western blotting, Mammalian Two-Hybrid and other techniques. The pretreatment with MOR-TAT-pep prior to morphine usage led to an enhanced analgesic effectiveness of morphine and a significant reduction in the development of morphine tolerance. The peptide directly interacted with ß-arrestin2 during morphine treatment and deceased the membrane recruitment of ß-arrestin2. MOR-TAT-pep effectively suppressed the increase of ß-arrestin2 induced by morphine. The MOR-TAT-pep could alleviate morphine tolerance through inhibition of ß-arrestin2.


Asunto(s)
Analgésicos Opioides , Morfina , Animales , Morfina/farmacología , Analgésicos Opioides/farmacología , beta-Arrestina 1 , Receptores Opioides mu/metabolismo , Escherichia coli/metabolismo , Péptidos , Mamíferos/metabolismo
19.
Molecules ; 28(22)2023 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-38005269

RESUMEN

Peptide-based opioid ligands are important candidates for the development of novel, safer, and more effective analgesics to treat pain. To develop peptide-based safer analgesics, we synthesized a mixture-based cyclic pentapeptide library containing a total of 24,624 pentapeptides and screened the mixture-based library samples using a 55 °C warm water tail-withdrawal assay. Using this phenotypic screening approach, we deconvoluted the mixture-based samples to identify a novel cyclic peptide Tyr-[D-Lys-Dap(Ant)-Thr-Gly] (CycloAnt), which produced dose- and time-dependent antinociception with an ED50 (and 95% confidence interval) of 0.70 (0.52-0.97) mg/kg i.p. mediated by the mu-opioid receptor (MOR). Additionally, higher doses (≥3 mg/kg, i.p.) of CycloAnt antagonized delta-opioid receptors (DOR) for at least 3 h. Pharmacological characterization of CycloAnt showed the cyclic peptide did not reduce breathing rate in mice at doses up to 15 times the analgesic ED50 value, and produced dramatically less hyperlocomotion than the MOR agonist, morphine. While chronic administration of CycloAnt resulted in antinociceptive tolerance, it was without opioid-induced hyperalgesia and with significantly reduced signs of naloxone-precipitated withdrawal, which suggested reduced physical dependence compared to morphine. Collectively, the results suggest this dual MOR/DOR multifunctional ligand is an excellent lead for the development of peptide-based safer analgesics.


Asunto(s)
Analgésicos Opioides , Péptidos Cíclicos , Ratones , Animales , Analgésicos Opioides/farmacología , Péptidos Cíclicos/farmacología , Receptores Opioides delta/agonistas , Morfina/farmacología , Analgésicos/farmacología , Analgésicos/uso terapéutico , Receptores Opioides mu/agonistas , Péptidos
20.
Sci Rep ; 13(1): 18164, 2023 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-37875567

RESUMEN

Opioid addiction and the opioid overdose epidemic are becoming more serious, and the development of therapeutic agents is essential for the pharmacological treatment of substance use disorders. The κ-opioid receptor (KOP) is a member of the opioid receptor system that has been gaining attention as a promising molecular target for the treatment of numerous human disorders, including pain, depression, anxiety, and drug addiction. Here, we biologically and pharmacologically evaluated a novel azepane-derived ligand, NP-5497-KA, as a selective KOP agonist. NP-5497-KA had 1000-fold higher selectivity for the KOP over the µ-opioid receptor (MOP), which was higher than nalfurafine (KOP/MOP: 65-fold), and acted as a selective KOP full agonist in the 3',5'-cyclic adenosine monophosphate assay. The oral administration of NP-5497-KA (1-10 mg/kg) dose-dependently suppressed morphine-induced conditioned place preference in C57BL/6 J mice, and its effects were comparable to an intraperitoneal injection of nalfurafine (1-10 µg/kg). Nalfurafine (10 µg/kg) significantly inhibited rotarod performance, whereas NP-5497-KA (10 mg/kg) exerted no effect on rotarod performance. These results indicate that NP-5497-KA may be a novel option for the treatment of opioid use disorder with fewer side effects.


Asunto(s)
Morfina , Trastornos Relacionados con Opioides , Ratones , Animales , Humanos , Morfina/farmacología , Ratones Endogámicos C57BL , Receptores Opioides , Receptores Opioides mu/agonistas , Receptores Opioides kappa/agonistas , Trastornos Relacionados con Opioides/tratamiento farmacológico , Recompensa , Analgésicos Opioides/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA