Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 71
Filtrer
1.
Drug Alcohol Depend Rep ; 12: 100268, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39262668

RÉSUMÉ

More than 90 % of opioid overdose deaths in North America are now caused by synthetic opioids, and while they are not as prevalent in the European illicit drug market, there are indications that they may become so in the near future. Multiple publications have argued that neither higher doses of naloxone nor more potent opioid receptor antagonists are needed to reverse a synthetic opioid overdose. However, the unique physicochemical properties of synthetic opioids result in a very rapid onset of respiratory depression compared to opium-based molecules, reducing the margin of opportunity to reverse an overdose. While intravenous administration rapidly delivers the high naloxone concentrations needed to reverse a synthetic opioid overdose, this option is often unavailable to first responders. A translational mechanistic model of opioid overdose developed by the FDA's Division of Applied Regulatory Science provides an unbiased approach to evaluate the effectiveness of overdose reversal strategies. Reports using this model demonstrated the naloxone tools (2 mg intramuscular and 4 mg intranasal) used by many first responders can result in an unacceptable loss of life following a synthetic opioid (fentanyl, carfentanil) overdose. Moreover, sequential (2.5 minutes between doses) administration of up to four doses of intranasal naloxone was no more effective at reducing the incidence of cardiac arrest (a surrogate endpoint for lethality) than a single dose, suggesting that attempts at titration may not provide the rapid absorption required to reverse a synthetic opioid overdose. This model was also used to compare the effectiveness of intranasal naloxone to intranasal nalmefene, a recently FDA-approved opioid receptor antagonist with a more rapid absorption and a higher affinity at mu-opioid receptors compared to intranasal naloxone. Intranasal nalmefene resulted in large and clinically meaningful reductions in the incidence of cardiac arrest compared to intranasal naloxone. Furthermore, simultaneous administration of four doses of intranasal naloxone was needed to reduce the incidence of cardiac arrest to levels approaching those produced by a single dose of intranasal nalmefene. These data are consistent with evidence that synthetics have indeed disrupted conventional wisdom in the treatment of opioid overdose.

3.
Front Psychiatry ; 15: 1399803, 2024.
Article de Anglais | MEDLINE | ID: mdl-38952632

RÉSUMÉ

Introduction: Using a validated translational model that quantitatively predicts opioid-induced respiratory depression and cardiac arrest, we compared cardiac arrest events caused by synthetic opioids (fentanyl, carfentanil) following rescue by intranasal (IN) administration of the µ-opioid receptor antagonists naloxone and nalmefene. Methods: This translational model was originally developed by Mann et al. (Clin Pharmacol Ther 2022) to evaluate the effectiveness of intramuscular (IM) naloxone. We initially implemented this model using published codes, reproducing the effects reported by Mann et al. on the incidence of cardiac arrest events following intravenous doses of fentanyl and carfentanil as well as the reduction in cardiac arrest events following a standard 2 mg IM dose of naloxone. We then expanded the model in terms of pharmacokinetic and µ-opioid receptor binding parameters to simulate effects of 4 mg naloxone hydrochloride IN and 3 mg nalmefene hydrochloride IN, both FDA-approved for the treatment of opioid overdose. Model simulations were conducted to quantify the percentage of cardiac arrest in 2000 virtual patients in both the presence and absence of IN antagonist treatment. Results: Following simulated overdoses with both fentanyl and carfentanil in chronic opioid users, IN nalmefene produced a substantially greater reduction in the incidence of cardiac arrest compared to IN naloxone. For example, following a dose of fentanyl (1.63 mg) producing cardiac arrest in 52.1% (95% confidence interval, 47.3-56.8) of simulated patients, IN nalmefene reduced this rate to 2.2% (1.0-3.8) compared to 19.2% (15.5-23.3) for IN naloxone. Nalmefene also produced large and clinically meaningful reductions in the incidence of cardiac arrests in opioid naïve subjects. Across dosing scenarios, simultaneous administration of four doses of IN naloxone were needed to reduce the percentage of cardiac arrest events to levels that approached those produced by a single dose of IN nalmefene. Conclusion: Simulations using this validated translational model of opioid overdose demonstrate that a single dose of IN nalmefene produces clinically meaningful reductions in the incidence of cardiac arrest compared to IN naloxone following a synthetic opioid overdose. These findings are especially impactful in an era when >90% of all opioid overdose deaths are linked to synthetic opioids such as fentanyl.

4.
J Clin Pharmacol ; 64(7): 828-839, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38436495

RÉSUMÉ

An open-label, randomized, crossover study in healthy volunteers compared the reversal of remifentanil-induced respiratory depression by intranasal (IN) naloxone hydrochloride (4 mg) to IN nalmefene (2.7 mg) (NCT04828005). Subjects were administered a hypercapnic gas mixture which produces an elevation in minute ventilation (MV), a result of the ventilatory response to hypercapnia. Subjects breathed a hypercapnic gas mixture through a tight-fitting mask for an initial period of 46 min prior to a series of mask "holidays" introduced to reduce subject discomfort and encourage study completion. Ten minutes after initiating the hypercapnic gas mixture, a remifentanil bolus was administered, and an infusion continued for the study duration. Subjects were administered either naloxone or nalmefene 15 min after initiating the remifentanil infusion and MV monitored for 21 min followed by a mask holiday. Both nalmefene and naloxone produced a time-dependent reversal of remifentanil-induced reductions in MV measured 2.5-20 min post administration. At the primary endpoint (5 min post administration), nalmefene increases in MV (5.75 L/min) were nearly twice that produced by naloxone (3.01 L/min) (P < .0009); the point estimate favors nalmefene, demonstrating non-inferiority and superiority. In this model of opioid-induced respiratory depression, nalmefene has a more rapid onset of action than naloxone, which required 20 min to achieve a comparable reversal of respiratory depression. Both nalmefene and naloxone were well tolerated by healthy volunteers. This rapid onset of action may prove particularly valuable in an era when over 90% of fatalities are linked to synthetic opioid overdose.


Sujet(s)
Administration par voie nasale , Analgésiques morphiniques , Études croisées , Volontaires sains , Naloxone , Naltrexone , Antagonistes narcotiques , Rémifentanil , Insuffisance respiratoire , Humains , Naloxone/administration et posologie , Naloxone/pharmacologie , Mâle , Adulte , Naltrexone/analogues et dérivés , Naltrexone/pharmacologie , Naltrexone/administration et posologie , Insuffisance respiratoire/induit chimiquement , Analgésiques morphiniques/administration et posologie , Analgésiques morphiniques/effets indésirables , Analgésiques morphiniques/pharmacologie , Femelle , Antagonistes narcotiques/administration et posologie , Antagonistes narcotiques/pharmacologie , Rémifentanil/administration et posologie , Rémifentanil/pharmacologie , Jeune adulte , Adulte d'âge moyen
6.
Clin Pharmacol Drug Dev ; 13(1): 58-69, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37496452

RÉSUMÉ

Nalmefene is a high-affinity, long-duration opioid antagonist that was approved in 1995 as an injection for the treatment of opiate overdose, but subsequently withdrawn (2008) for reasons other than safety or effectiveness. The dramatic rise in opioid overdose deaths over the past 7-8 years catalyzed the development of an intranasal (IN) formulation of nalmefene for the emergency treatment of opioid overdose. The studies described here compare the pharmacokinetic properties and safety profiles of an IN formulation containing nalmefene (2.7 mg in 0.1 mL) to an approved 1 mg intramuscular (IM) dose. IN nalmefene produced maximum plasma concentrations that were significantly higher than observed following the IM dose (12.2 and 1.77 ng/mL, respectively). The time to reach maximum plasma concentrations was also faster following IN administration (0.25 and 0.33 hours, respectively) with significant differences in plasma concentrations manifested as early as 2.5 minutes after administration (NCT04759768). The plasma half-life of nalmefene was similar following IM and IN administration (10.6-11.4 hours). Furthermore, dose-normalized nalmefene exposure was similar for both 1 spray in each nostril and 2 sprays in the same nostril compared to a single spray in each nostril (NCT05219669). There were no sex differences in the pharmacokinetic properties of either IN or IM nalmefene. In an era when almost 90% of opioid overdose deaths have been linked to high-potency synthetic opioids, the ability to rapidly deliver high concentrations of nalmefene could represent an important tool for reducing both morbidity and mortality.


Sujet(s)
Mauvais usage des médicaments prescrits , Surdose d'opiacés , Humains , Surdose d'opiacés/traitement médicamenteux , Mauvais usage des médicaments prescrits/traitement médicamenteux , Naltrexone , Antagonistes narcotiques
7.
Pharmacol Ther ; 233: 108019, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-34637841

RÉSUMÉ

Overdose deaths are often viewed as the leading edge of the opioid epidemic which has gripped the United States over the past two decades (Skolnick, 2018a). This emphasis is perhaps unsurprising because opioid overdose is both the number-one cause of death for individuals between 25 and 64 years old (Dezfulian et al., 2021) and a significant contributor to the decline in average lifespan (Dowell et al., 2017). Exacerbated by the COVID 19 pandemic, it was estimated there were 93,400 drug overdose deaths in the United States during the 12 months ending December 2020, with more than 69,000 (that is, >74%) of these fatalities attributed to opioid overdose (Ahmad et al., 2021). However, the focus on mortality statistics (Ahmad et al., 2021; Shover et al., 2020) tends to obscure the broader medical impact of nonfatal opioid overdose. Analyses of multiple databases indicate that for each opioid-induced fatality, there are between 6.4 and 8.4 non-fatal overdoses, exacting a significant burden on both the individual and society. Over the past 7-8 years, there has been an alarming increase in the misuse of synthetic opioids ("synthetics"), primarily fentanyl and related piperidine-based analogs. Within the past 2-3 years, a structurally unrelated class of high potency synthetics, benzimidazoles exemplified by etonitazene and isotonitazene ("iso"), have also appeared in illicit drug markets (Thompson, 2020; Ujvary et al. 2021). In 2020, it was estimated that over 80% of fatal opioid overdoses in the United States now involve synthetics (Ahmad et al., 2021). The unique physicochemical and pharmacological properties of synthetics described in this review are responsible for both the morbidity and mortality associated with their misuse as well as their widespread availability. This dramatic increase in the misuse of synthetics is often referred to as the "3rd wave" (Pardo et al., 2019; Volkow and Blanco, 2020) of the opioid epidemic. Among the consequences resulting from misuse of these potent opioids is the need for higher doses of the competitive antagonist, naloxone, to reverse an overdose. The development of more effective reversal agents such as those described in this review is an essential component of a tripartite strategy (Volkow and Collins, 2017) to reduce the biopsychosocial impact of opioid misuse in the "synthetic era".


Sujet(s)
Traitements médicamenteux de la COVID-19 , Mauvais usage des médicaments prescrits , Surdose d'opiacés , Adulte , Analgésiques morphiniques/effets indésirables , Mauvais usage des médicaments prescrits/traitement médicamenteux , Mauvais usage des médicaments prescrits/épidémiologie , Humains , Adulte d'âge moyen , Naloxone/usage thérapeutique , Surdose d'opiacés/traitement médicamenteux , Surdose d'opiacés/épidémiologie , États-Unis/épidémiologie
8.
Clin Pharmacol Ther ; 109(3): 578-590, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33113208

RÉSUMÉ

The only medication available currently to prevent and treat opioid overdose (naloxone) was approved by the US Food and Drug Administration (FDA) nearly 50 years ago. Because of its pharmacokinetic and pharmacodynamic properties, naloxone has limited utility under some conditions and would not be effective to counteract mass casualties involving large-scale deployment of weaponized synthetic opioids. To address shortcomings of current medical countermeasures for opioid toxicity, a trans-agency scientific meeting was convened by the US National Institute of Allergy and Infectious Diseases/National Institutes of Health (NIAID/NIH) on August 6 and 7, 2019, to explore emerging alternative approaches for treating opioid overdose in the event of weaponization of synthetic opioids. The meeting was initiated by the Chemical Countermeasures Research Program (CCRP), was organized by NIAID, and was a collaboration with the National Institute on Drug Abuse/NIH (NIDA/NIH), the FDA, the Defense Threat Reduction Agency (DTRA), and the Biomedical Advanced Research and Development Authority (BARDA). This paper provides an overview of several presentations at that meeting that discussed emerging new approaches for treating opioid overdose, including the following: (1) intranasal nalmefene, a competitive, reversible opioid receptor antagonist with a longer duration of action than naloxone; (2) methocinnamox, a novel opioid receptor antagonist; (3) covalent naloxone nanoparticles; (4) serotonin (5-HT)1A receptor agonists; (5) fentanyl-binding cyclodextrin scaffolds; (6) detoxifying biomimetic "nanosponge" decoy receptors; and (7) antibody-based strategies. These approaches could also be applied to treat opioid use disorder.


Sujet(s)
Analgésiques morphiniques/effets indésirables , Mauvais usage des médicaments prescrits/thérapie , Mesures sanitaires préventives , Naloxone/usage thérapeutique , Antagonistes narcotiques/usage thérapeutique , Épidémie d'opioïdes , Troubles liés aux opiacés/thérapie , Animaux , Congrès comme sujet , Mauvais usage des médicaments prescrits/étiologie , Mauvais usage des médicaments prescrits/mortalité , Humains , Naloxone/effets indésirables , Antagonistes narcotiques/effets indésirables , Épidémie d'opioïdes/mortalité , Troubles liés aux opiacés/complications , Troubles liés aux opiacés/mortalité , Pronostic , Appréciation des risques , Facteurs de risque
9.
J Neural Transm (Vienna) ; 127(2): 279-286, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31893308

RÉSUMÉ

The legalization of cannabis for both recreational and medical use in the USA has resulted in a dramatic increase in the number of emergency department visits and hospital admissions for acute cannabinoid overdose (also referred to as cannabis intoxication and cannabis poisoning). Both "edibles" (often sold as brownies, cookies, and candies) containing large amounts of Δ9-tetrahydrocannabinol and synthetic cannabinoids (many possessing higher potencies and efficacies than Δ9-tetrahydrocannabinol) are responsible for a disproportionate number of emergency department visits relative to smoked cannabis. Symptoms of acute cannabinoid overdose range from extreme lethargy, ataxia, and generalized psychomotor impairment to feelings of panic and anxiety, agitation, hallucinations, and psychosis. Treatment of acute cannabinoid overdose is currently supportive and symptom driven. Converging lines of evidence indicating many of the symptoms which can precipitate an emergency department visit are mediated through activation of cannabinoid1 receptors. Here, we review the evidence that cannabinoid1 receptor antagonists, originally developed for indications ranging from obesity to smoking cessation and schizophrenia, provide a molecular approach to treating acute cannabinoid overdose.


Sujet(s)
Antagonistes des récepteurs de cannabinoïdes/pharmacologie , Cannabinoïdes/effets indésirables , Mauvais usage des médicaments prescrits/traitement médicamenteux , Récepteur cannabinoïde de type CB1/antagonistes et inhibiteurs , Humains
10.
Eur Neuropsychopharmacol ; 29(12): 1312-1320, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31753777

RÉSUMÉ

Both positive and negative (null or neutral) results are essential for the progress of science and its self-correcting nature. However, there is general reluctance to publish negative results, and this may be due a range of factors (e.g., the widely held perception that negative results are more difficult to publish, the preference to publish positive findings that are more likely to generate citations and funding for additional research). It is particularly challenging to disclose negative results that are not consistent with previously published positive data, especially if the initial publication appeared in a high impact journal. Ideally, there should be both incentives and support to reduce the costs associated with investing efforts into preparing publications with negative results. We describe here a set of criteria that can help scientists, reviewers and editors to publish technically sound, scientifically high-impact negative (or null) results originating from rigorously designed and executed studies. Proposed criteria emphasize the importance of collaborative efforts and communication among scientists (also including the authors of original publications with positive results).


Sujet(s)
Résultats négatifs/normes , Évaluation de la recherche par les pairs/normes , Périodiques comme sujet/normes , Humains , Évaluation de la recherche par les pairs/méthodes
11.
J Pharmacol Exp Ther ; 371(2): 409-415, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-30940694

RÉSUMÉ

The dramatic rise in overdose deaths linked to synthetic opioids (e.g., fentanyl, carfentanil) may require more potent, longer-duration opiate antagonists than naloxone. Both the high affinity of nalmefene at µ opiate receptors and its long half-life led us to examine the feasibility of developing an intranasal (IN) formulation as a rescue medication that could be especially useful in treating synthetic opioid overdose. In this study, the pharmacokinetic properties of IN nalmefene were compared with an intramuscular (i.m.) injection in a cohort of healthy volunteers. Nalmefene was absorbed slowly following IN administration, with a median time to reach Cmax (Tmax) of 2 hours. Addition of the absorption enhancer dodecyl maltoside (Intravail, Neurelis, Inc., Encinitas, CA) reduced Tmax to 0.25 hour and increased Cmax by ∼2.2-fold. The pharmacokinetic properties of IN nalmefene (3 mg) formulated with dodecyl maltoside has characteristics consistent with an effective rescue medication: its onset of action is comparable to an i.m. injection of nalmefene (1.5 mg) previously approved to treat opioid overdose. Furthermore, the Cmax following IN administration was ∼3-fold higher than following i.m. dosing, comparable to previously reported plasma concentrations of nalmefene observed 5 minutes following a 1-mg i.v. dose. The high affinity, very rapid onset, and long half-life (>7 hours) of IN nalmefene present distinct advantages as a rescue medication, particularly against longer-lived synthetic opioids.


Sujet(s)
Analgésiques morphiniques/effets indésirables , Développement de médicament/méthodes , Mauvais usage des médicaments prescrits/traitement médicamenteux , Naltrexone/analogues et dérivés , Antagonistes narcotiques/administration et posologie , Médicaments de synthèse/effets indésirables , Administration par voie nasale , Adolescent , Adulte , Analgésiques morphiniques/sang , Études croisées , Méthode en double aveugle , Mauvais usage des médicaments prescrits/sang , Femelle , Humains , Injections musculaires , Mâle , Adulte d'âge moyen , Naltrexone/administration et posologie , Naltrexone/sang , Antagonistes narcotiques/sang , Médicaments de synthèse/métabolisme , Résultat thérapeutique , Jeune adulte
12.
Drug Metab Dispos ; 47(7): 690-698, 2019 07.
Article de Anglais | MEDLINE | ID: mdl-30992306

RÉSUMÉ

Naloxone (17-allyl-4,5α-epoxy-3,14-dihydroxymorphinan-6-one HCl), a µ-opioid receptor antagonist, is administered intranasally to reverse an opioid overdose but its short half-life may necessitate subsequent doses. The addition of naltrexone [17-(cyclopropylmethyl)-4,5α-epoxy-3,14-dihydroxymorphinan-6-one], another µ-receptor antagonist, which has a reported half-life of 3 1/2 hours, may extend the available time to receive medical treatment. In a phase 1 pharmacokinetic study, healthy adults were administered naloxone and naltrexone intranasally, separately and in combination. When administered with naloxone, the C max value of naltrexone decreased 62% and the area under the concentration-time curve from time zero to infinity (AUC0-inf) decreased 38% compared with when it was given separately; lower concentrations of naltrexone were observed as early as 5 minutes postdose. In contrast, the C max and AUC0-inf values of naloxone decreased only 18% and 16%, respectively, when given with naltrexone. This apparent interaction was investigated further to determine if naloxone and naltrexone shared a transporter. Neither compound was a substrate for organic cation transporter (OCT) 1, OCT2, OCT3, OCTN1, or OCTN2. There was no evidence of the involvement of a transmembrane transporter when they were tested separately or in combination at concentrations of 10 and 500 µM using Madin-Darby canine kidney II cell monolayers at pH 7.4. The efflux ratios of naloxone and naltrexone increased to six or greater when the apical solution was pH 5.5, the approximate pH of the nasal cavity; there was no apparent interaction when the two were coincubated. The importance of understanding how opioid antagonists are absorbed by the nasal epithelium is magnified by the rise in overdose deaths attributed to long-lived synthetic opioids and the realization that better strategies are needed to treat opioid overdoses.


Sujet(s)
Naloxone/pharmacocinétique , Naltrexone/pharmacocinétique , Antagonistes narcotiques/pharmacocinétique , Administration par voie nasale , Adolescent , Adulte , Études croisées , Méthode en double aveugle , Interactions médicamenteuses , Femelle , Humains , Mâle , Adulte d'âge moyen , Naloxone/administration et posologie , Naloxone/sang , Naltrexone/administration et posologie , Naltrexone/sang , Antagonistes narcotiques/administration et posologie , Antagonistes narcotiques/sang , Jeune adulte
13.
J Clin Pharmacol ; 59(7): 947-957, 2019 07.
Article de Anglais | MEDLINE | ID: mdl-30698833

RÉSUMÉ

Based on its high affinity for µ opiate receptors and reported half-life after oral administration, the pharmacokinetic properties of intranasal naltrexone were examined to evaluate its potential to treat opioid overdose. This study was prompted by the marked rise in overdose deaths linked to synthetic opioids like fentanyl, which may require more potent, longer-lived opiate antagonists than naloxone. Both the maximum plasma concentration (Cmax ) and the time (Tmax ) to reach Cmax for intranasal naltrexone (4 mg) were comparable to values reported for a Food and Drug Administration-approved 4-mg dose of intranasal naloxone. The addition of the absorption enhancer dodecyl maltoside (Intravail) increased Cmax by ∼3-fold and reduced the Tmax from 0.5 to 0.17 hours. Despite these very rapid increases in plasma concentrations of naltrexone, its short half-life following intranasal administration (∼2.2 hours) could limit its usefulness as a rescue medication, particularly against longer-lived synthetic opioids. Nonetheless, the ability to rapidly attain high plasma concentrations of naltrexone may be useful in other indications, including an as-needed dosing strategy to treat alcohol use disorder.


Sujet(s)
Maltose/analogues et dérivés , Naltrexone/sang , Naltrexone/pharmacocinétique , Alcaloïdes opiacés/antagonistes et inhibiteurs , Administration par voie nasale , Administration par voie orale , Adulte , Aire sous la courbe , Lignée cellulaire , Études croisées , Mauvais usage des médicaments prescrits , Femelle , Période , Humains , Concentration en ions d'hydrogène , Injections musculaires , Mâle , Maltose/pharmacocinétique , Adulte d'âge moyen , Naltrexone/administration et posologie , Absorption nasale , Perméabilité/effets des médicaments et des substances chimiques , Jeune adulte
14.
Eur J Pharmacol ; 835: 147-153, 2018 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-30092179

RÉSUMÉ

Naloxone is a specific, high affinity opioid antagonist that has been used to treat suspected or confirmed overdose for more than 40 years. Naloxone use was initially confined to an emergency room setting, but the dramatic rise in opioid overdose events over the past two decades has, with increasing frequency, shifted naloxone use to first responders including police, emergency medical technicians, and the friends and family of overdose victims. The opioids responsible for overdose events have also evolved, from prescription opioids to heroin and most recently, very high potency synthetic opioids such as fentanyl. In 2016, synthetic opioids were linked to more overdose fatalities than either prescription opioids or heroin. In this review, I will discuss the evolution and use of naloxone products by first responders and the development of additional rescue medications in response to the unprecedented dangers posed by synthetic opioids.


Sujet(s)
Naloxone/pharmacologie , Antagonistes narcotiques/pharmacologie , Troubles liés aux opiacés/épidémiologie , Mauvais usage des médicaments prescrits/traitement médicamenteux , Humains , Naloxone/usage thérapeutique , Antagonistes narcotiques/usage thérapeutique
15.
Subst Use Misuse ; 53(13): 2257-2264, 2018 11 10.
Article de Anglais | MEDLINE | ID: mdl-29927691

RÉSUMÉ

BACKGROUND: Cannabis use disorder (CUD) as described/defined in DSM 5, is characterized by impaired control of marijuana use and related personal, health, and legal consequences. CUD is a serious public health problem, affecting nearly 6 million individuals in the United States. There are no FDA approved medications to treat this disorder. The lack of available treatment options contributes to uncertainties by drug sponsors about formulary and reimbursement decision-making for CUD pharmacotherapies. OBJECTIVE: To addresses this gap by presenting the first findings on managed care payers' perceptions of CUD treatments and clinical trial end points. METHODS: An online survey was conducted with 50 payers from managed care organizations. The survey inquired about perceptions of unmet need in CUD treatment, relevant clinical trial end points, disease knowledge, and likelihood of review of new pharmacotherapies. RESULTS: The majority of payers (62%) reported that they were at least moderately familiar with CUD treatment end points. Most (80%) rated the unmet need for new pharmacotherapies for CUD as at least moderately important. Payers rated the most important end points for clinical trials as abstinence and decreased resource utilization. Most participants said an FDA approved CUD treatment would be formally reviewed by payers within 6 months (58%) or a year (36%). CONCLUSIONS: Based on these findings, payers see an unmet need for CUD treatment. Furthermore, FDA-approved pharmacotherapies for CUD will likely be reviewed quickly by payers, especially if data are provided on the likelihood of achieving abstinence and reduced resource utilization.


Sujet(s)
Attitude du personnel soignant , Remboursement par l'assurance maladie , Abus de marijuana/rééducation et réadaptation , Prise de décision , Diagnostic and stastistical manual of mental disorders (USA) , Agrément de médicaments , Humains , Couverture d'assurance , Évaluation des besoins , Psychoanaleptiques/usage thérapeutique , Mécanismes de remboursement , Enquêtes et questionnaires , États-Unis , Food and Drug Administration (USA)
16.
Sci Transl Med ; 10(434)2018 03 28.
Article de Anglais | MEDLINE | ID: mdl-29593105

RÉSUMÉ

The FDA's "abstinence" outcome measure for approval of new medications to treat opioid-use disorders has been difficult to achieve; developing and validating alternative meaningful outcomes could facilitate drug development.


Sujet(s)
Troubles liés aux opiacés/traitement médicamenteux , Troubles liés aux opiacés/prévention et contrôle , Agrément de médicaments/statistiques et données numériques , Développement de médicament/statistiques et données numériques , Humains , États-Unis , Food and Drug Administration (USA)
18.
Annu Rev Pharmacol Toxicol ; 58: 143-159, 2018 01 06.
Article de Anglais | MEDLINE | ID: mdl-28968188

RÉSUMÉ

The widespread abuse of prescription opioids and a dramatic increase in the availability of illicit opioids have created what is commonly referred to as the opioid epidemic. The magnitude of this epidemic is startling: About 4% of the adult US population misuses prescription opioids, and in 2015, more than 33,000 deaths were attributable to overdose with licit and illicit opioids. Increasing the availability of medication-assisted treatments (such as buprenorphine and naltrexone), the use of abuse-deterrent formulations, and the adoption of US Centers for Disease Control and Prevention prescribing guidelines all constitute short-term approaches to quell this epidemic. However, with more than 125 million Americans suffering from either acute or chronic pain, the development of effective alternatives to opioids, enabled at least in part by a fuller understanding of the neurobiological bases of pain, offers the best long-term solution for controlling and ultimately eradicating this epidemic.


Sujet(s)
Analgésiques morphiniques/effets indésirables , Analgésiques morphiniques/usage thérapeutique , Douleur chronique/traitement médicamenteux , Animaux , Mauvais usage des médicaments prescrits/prévention et contrôle , Ordonnances médicamenteuses/normes , Humains , Substances illicites/effets indésirables , Substances illicites/législation et jurisprudence , Troubles liés à une substance/prévention et contrôle , États-Unis
19.
Psychopharmacology (Berl) ; 234(9-10): 1347-1355, 2017 05.
Article de Anglais | MEDLINE | ID: mdl-27995279

RÉSUMÉ

OBJECTIVE: This paper provides an overview of the role of type 2 metabotropic glutamate receptors (mGluR2) in addiction and behaviors reflecting addictive processes. RESULTS: AZD8529, an mGluR2 positive allosteric modulator (PAM), failed to separate from placebo in a phase II schizophrenia trial. The demonstration by Athina Markou's laboratory that AZD8529 attenuated both nicotine self-administration and cue-induced reinstatement was a key factor in the decision to move this compound into a smoking cessation study. CONCLUSION: Here, we highlight Markou laboratory's contribution to this project, as well as several innovative features of the phase II clinical trial that has already completed enrollment with top line results expected in early 2017.


Sujet(s)
Indoles/usage thérapeutique , Oxadiazoles/usage thérapeutique , Récepteurs métabotropes au glutamate/métabolisme , Troubles liés à une substance/traitement médicamenteux , Troubles liés à une substance/métabolisme , Dispositifs de sevrage tabagique , Régulation allostérique/effets des médicaments et des substances chimiques , Régulation allostérique/physiologie , Animaux , Relation dose-effet des médicaments , Humains , Indoles/pharmacologie , Mâle , Nicotine/administration et posologie , Oxadiazoles/pharmacologie , Récepteurs métabotropes au glutamate/agonistes , Récepteurs métabotropes au glutamate/antagonistes et inhibiteurs , Autoadministration/méthodes , Résultat thérapeutique
20.
Neuron ; 92(2): 294-297, 2016 Oct 19.
Article de Anglais | MEDLINE | ID: mdl-27764663

RÉSUMÉ

Limited options for treating moderate-severe pain led to an overreliance on opioids and the current opioid epidemic. Addressing the factors contributing to a dearth of effective alternatives and re-energizing the development of pain therapeutics is necessary to quell this epidemic.


Sujet(s)
Analgésiques/usage thérapeutique , Douleur chronique/traitement médicamenteux , Découverte de médicament , Épidémies , Troubles liés aux opiacés/épidémiologie , Douleur aigüe/traitement médicamenteux , Analgésiques morphiniques/usage thérapeutique , Humains , Gestion de la douleur , Éthers phényliques/usage thérapeutique , Prégabaline/usage thérapeutique , Proline/analogues et dérivés , Proline/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE