Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 392
Filter
Add more filters

Publication year range
1.
Acta Pharmacol Sin ; 45(8): 1752-1764, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38570601

ABSTRACT

Morphine and morphine-6-glucuronide (M6G) produce central nervous system (CNS) effects by activating mu-opioid receptors, while naloxone is used mainly for the reversal of opioid overdose, specifically for the fatal complication of respiratory depression, but also for alleviating opioid-induced side effects. In this study we developed a physiologically-based pharmacokinetic-pharmacodynamic (PBPK-PD) model to simultaneously predict pharmacokinetics and CNS effects (miosis, respiratory depression and analgesia) of morphine as well as antagonistic effects of naloxone against morphine. The pharmacokinetic and pharmacodynamic parameters were obtained from in vitro data, in silico, or animals. Pharmacokinetic and pharmacodynamic simulations were conducted using 39 and 36 clinical reports, respectively. The pharmacokinetics of morphine and M6G following oral or intravenous administration were simulated, and the PBPK-PD model was validated using clinical observations. The Emax model correlated CNS effects with free concentrations of morphine and M6G in brain parenchyma. The predicted CNS effects were compared with observations. Most clinical observations fell within the 5th-95th percentiles of simulations based on 1000 virtual individuals. Most of the simulated area under the concentration-time curve or peak concentrations also fell within 0.5-2-fold of observations. The contribution of morphine to CNS effects following intravenous or oral administration was larger than that of M6G. Pharmacokinetics and antagonistic effects of naloxone on CNS effects were also successfully predicted using the developed PBPK-PD model. In conclusion, the pharmacokinetics and pharmacodynamics of morphine and M6G, antagonistic effects of naloxone against morphine-induced CNS effects may be successfully predicted using the developed PBPK-PD model based on the parameters derived from in vitro, in silico, or animal studies.


Subject(s)
Models, Biological , Morphine , Naloxone , Narcotic Antagonists , Naloxone/pharmacokinetics , Naloxone/pharmacology , Humans , Morphine/pharmacokinetics , Morphine/administration & dosage , Morphine/pharmacology , Narcotic Antagonists/pharmacokinetics , Narcotic Antagonists/pharmacology , Narcotic Antagonists/administration & dosage , Animals , Morphine Derivatives/pharmacokinetics , Central Nervous System/drug effects , Central Nervous System/metabolism , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/administration & dosage , Male , Computer Simulation , Administration, Oral , Adult , Administration, Intravenous , Female
2.
Drug Metab Dispos ; 50(2): 168-173, 2022 02.
Article in English | MEDLINE | ID: mdl-34728519

ABSTRACT

Naltrexone, an opioid antagonist primarily metabolized by aldo-keto reductase 1C4 (AKR1C4), treats pediatric conditions involving compulsiveness (e.g., autism spectrum, Prader-Willi, eating disorders, non-suicidal self-injury). Pharmacokinetic variability is apparent in adults, yet no data are available for children. This study aimed to examine the impact of age and genetic variation on naltrexone biotransformation. Human liver cytosol (HLC) samples (n = 158) isolated from children and adult organ donors were incubated with therapeutically relevant concentrations of naltrexone (0.1, 1 µM). Naltrexone biotransformation was determined by ultraperformance mass spectrometry quantification of the primary metabolite, 6-beta-naltrexol (6ßN), and 6ßN formation rates (pmol/mg protein/min) were calculated. HLCs from organ donors, age range 0-79 y (mean 16.0 ± 18.2 y), 37% (n = 60) female, 20% (n = 33) heterozygous and 1.2% (n = 2) homozygous for co-occurring AKR1C4 variants (S145C/L311V) showed >200-fold range in 6ßN formation (0.37-76.5 pmol/mg protein/min). Source of donor samples was found to be a substantial contributor to variability. Model estimates for a trimmed data set of source-adjusted pediatric samples (aged 0-18 y) suggested that AKR1C4 genetic variation, age, and sex explained 36% of the variability in 6ßN formation. Although activity increased steadily from birth and peaked in middle childhood (2-5 years), genetic variation (S145C/L311V) demonstrated a greater effect on activity than did age. Naltrexone biotransformation is highly variable in pediatric and adult livers and can be partly accounted for by individual factors feasible to obtain (e.g., genetic variability, age, sex). These data may inform a precision therapeutics approach (e.g., exposure optimization) to further study Naltrexone responsiveness in children and adults. SIGNIFICANCE STATEMENT: Biotransformation of the commonly used opioid antagonist naltrexone is highly variable and may contribute to reduced therapeutic response. Age, sex, and genetic variation in the drug-metabolizing enzyme, AKR1C4, are potential factors contributing to this variability. In pediatric samples, genetic variation (S145C/L311V) demonstrates a greater impact on activity than age. Additionally, the source of donor samples was identified as an important contributor and must be accounted for to confidently elucidate the biological variables most impactful to drug biotransformation.


Subject(s)
Naltrexone , Narcotic Antagonists , Adolescent , Adult , Aged , Biotransformation , Child , Child, Preschool , Cytosol/metabolism , Female , Humans , Infant , Infant, Newborn , Metabolic Clearance Rate , Middle Aged , Naltrexone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Young Adult
3.
Pharm Res ; 39(5): 963-975, 2022 May.
Article in English | MEDLINE | ID: mdl-35386013

ABSTRACT

OBJECTIVE: To examine the pharmacokinetics and safety of FMXIN001, a new intranasal powder-based naloxone formulation, in comparison to Narcan® nasal liquid spray. METHODS: FMXIN001, was developed by blending drug microspheres with larger lactose monohydrate particles, that serve as diluent and carrier, as well as a disaggregating agent. Scanning electron microscopy and X-ray were used to characterize the formulation and in vitro deposition was investigated using a nasal cast. We compared the pharmacokinetics and safety of FMXIN001 versus Narcan® in two clinical trials: a pilot study with 14 healthy adults and a pivotal trial in 42 healthy adults (NCT04713709). The studies were open-label, single-dose, randomized, two-period, two-treatment, two-sequence crossover studies to assess the pharmacokinetics and safety of FMXIN001 versus Narcan® nasal spray. RESULTS: FMXIN001 comprises naloxone microspheres (5-30 µM) and lactose particles (40-240 µM). Upon in vitro testing, naloxone deposits mainly to the middle turbinates region and the upper part of the nasal cavity of a nasal cast. In human subjects, FMXIN001 produced significantly higher exposure at the initial time points of 4, 10, and 30 min, post-administration, compared to Narcan®. Both treatments were safe and well tolerated. FMXIN001, powder-based spray, results in similar overall exposure to Narcan®, but with more rapid absorption in the first 30 min. CONCLUSIONS: FMXIN001 is expected to have a shorter onset of action for a more effective therapeutic intervention to manage opioid overdose. Rapid administration of naloxone in cases of opioid overdose is imperative, given the alarming increase in mortality rates.


Subject(s)
Drug Overdose , Opiate Overdose , Administration, Intranasal , Adult , Drug Overdose/drug therapy , Humans , Lactose , Naloxone/pharmacokinetics , Naloxone/therapeutic use , Narcotic Antagonists/pharmacokinetics , Narcotic Antagonists/therapeutic use , Nasal Sprays , Pilot Projects , Powders
4.
Br J Anaesth ; 128(3): 584-595, 2022 03.
Article in English | MEDLINE | ID: mdl-34872716

ABSTRACT

BACKGROUND: The safety profile of buprenorphine has encouraged its widespread use. However, fatalities have been attributed to benzodiazepine/buprenorphine combinations, by poorly understood mechanisms of toxicity. Mechanistic hypotheses include (i) benzodiazepine-mediated increase in brain buprenorphine (pharmacokinetic hypothesis); (ii) benzodiazepine-mediated potentiation of buprenorphine interaction with opioid receptors (receptor hypothesis); and (iii) combined effects of buprenorphine and benzodiazepine on respiratory parameters (pharmacodynamic hypothesis). METHODS: We studied the neuro-respiratory effects of buprenorphine (30 mg kg-1, i.p.), diazepam (20 mg kg-1, s.c.), and diazepam/buprenorphine combination in rats using arterial blood gas analysis, plethysmography, and diaphragm electromyography. Pretreatments with various opioid and gamma-aminobutyric acid receptor antagonists were tested. Diazepam impact on brain 11C-buprenorphine kinetics and binding to opioid receptors was studied using positron emission tomography imaging. RESULTS: In contrast to diazepam and buprenorphine alone, diazepam/buprenorphine induced early-onset sedation (P<0.05) and respiratory depression (P<0.001). Diazepam did not alter 11C-buprenorphine brain kinetics or binding to opioid receptors. Diazepam/buprenorphine-induced effects on inspiratory time were additive, driven by buprenorphine (P<0.0001) and were blocked by naloxonazine (P<0.01). Diazepam/buprenorphine-induced effects on expiratory time were non-additive (P<0.001), different from buprenorphine-induced effects (P<0.05) and were blocked by flumazenil (P<0.01). Diazepam/buprenorphine-induced effects on tidal volume were non-additive (P<0.01), different from diazepam- (P<0.05) and buprenorphine-induced effects (P<0.0001) and were blocked by naloxonazine (P<0.05) and flumazenil (P<0.05). Compared with buprenorphine, diazepam/buprenorphine decreased diaphragm contraction amplitude (P<0.01). CONCLUSIONS: Pharmacodynamic parameters and antagonist pretreatments indicate that diazepam/buprenorphine-induced respiratory depression results from a pharmacodynamic interaction between both drugs on ventilatory parameters.


Subject(s)
Buprenorphine , Diazepam , Respiratory Insufficiency , Animals , Male , Rats , Analgesics, Opioid/pharmacokinetics , Benzodiazepines/pharmacokinetics , Blood Gas Analysis/methods , Buprenorphine/adverse effects , Buprenorphine/pharmacokinetics , Diazepam/adverse effects , Diazepam/pharmacokinetics , Drug Interactions/physiology , Flumazenil/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Rats, Sprague-Dawley , Receptors, Opioid/metabolism , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/metabolism
5.
J Biomed Sci ; 28(1): 62, 2021 Sep 09.
Article in English | MEDLINE | ID: mdl-34503531

ABSTRACT

BACKGROUND: One of the most prominent opioid analgesics in the United States is the high potency agonist fentanyl. It is used in the treatment of acute and chronic pain and as an anesthetic adjuvant. When used inappropriately, however, ingestion of just a few milligrams of fentanyl or other synthetic opioid can cause opioid-induced respiratory depression (OIRD), often leading to death. Currently, the treatment of choice for OIRD is the opioid receptor antagonist naloxone. Recent reports, however, suggest that higher doses or repeated dosing of naloxone (due to recurrence of respiratory depression) may be required to reverse fully fentanyl-induced respiratory depression, rendering this treatment inadequate. To combat this synthetic opioid overdose crisis, this research aims at identifying a novel opioid reversal agent with enhanced efficacy towards fentanyl and other synthetic opioids. METHODS: A series of naltrexone analogues were characterized for their ability to antagonize the effects of fentanyl in vitro utilizing a modified forskolin-induced cAMP accumulation assay. Lead analogue 29 was chosen to undergo further PK studies, followed by in vivo pharmacological analysis to determine its ability to antagonize opioid-induced antinociception in the hot plate assay. RESULTS: A series of potent MOR antagonists were identified, including the highly potent analogue 29 (IC50 = 2.06 nM). Follow-up PK studies revealed 29 to possess near 100% bioavailability following IP administration. Brain concentrations of 29 surpassed plasma concentrations, with an apparent terminal half-life of ~ 80 min in mice. In the hot plate assay, 29 dose-dependently (0.01-0.1 mg/kg; IP) and fully antagonized the antinociception induced by oxycodone (5.6 mg/kg; IP). Furthermore, the dose of 29 that is fully effective in preventing oxycodone-induced antinociception (0.1 mg/kg) was ineffective against locomotor deficits caused by the KOR agonist U50,488. CONCLUSIONS: Methods have been developed that have utility to identify enhanced rescue agents for the treatment of OIRD. Analogue 29, possessing potent MOR antagonist activity in vitro and in vivo, provides a promising lead in our search for an enhanced synthetic opioid rescue agent.


Subject(s)
Analgesics, Opioid/adverse effects , Fentanyl/adverse effects , Naltrexone , Narcotic Antagonists , Animals , Drug Design , Male , Mice , Mice, Inbred C57BL , Naltrexone/chemical synthesis , Naltrexone/pharmacokinetics , Naltrexone/pharmacology , Narcotic Antagonists/chemical synthesis , Narcotic Antagonists/pharmacokinetics , Narcotic Antagonists/pharmacology
6.
Pharm Res ; 38(7): 1221-1234, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34114163

ABSTRACT

PURPOSE: Opioids have been the main factor for drug overdose deaths in the United States. Current naloxone delivery systems are effective in mitigating the opioid effects only for hours. Naloxone-loaded poly(lactide-co-glycolide) (PLGA) microparticles were prepared as quick- and long-acting naloxone delivery systems to extend the naloxone effect as an opioid antidote. METHODS: The naloxone-PLGA microparticles were made using an emulsification solvent extraction approach with different formulation and processing parameters. Two PLGA polymers with the lactide:glycolide (L:G) ratios of 50:50 and 75:25 were used, and the drug loading was varied from 21% to 51%. Two different microparticles of different sizes with the average diameters of 23 µm and 50 µm were produced using two homogenization-sieving conditions. All the microparticles were critically characterized, and three of them were evaluated with ß-arrestin recruitment assays. RESULTS: The naloxone encapsulation efficiency (EE) was in the range of 70-85%. The EE was enhanced when the theoretical naloxone loading was increased from 30% to 60%, the L:G ratio was changed from 50:50 to 75:25, and the average size of the particles was reduced from 50 µm to 23 µm. The in vitro naloxone release duration ranged from 4 to 35 days. Reducing the average size of the microparticles from 50 µm to 23 µm helped eliminate the lag phase and obtain the steady-state drug release profile. The cellular pharmacodynamics of three selected formulations were evaluated by applying DAMGO, a synthetic opioid peptide agonist to a µ-opioid receptor, to recruit ß-arrestin 2. CONCLUSIONS: Naloxone released from the three selected formulations could inhibit DAMGO-induced ß-arrestin 2 recruitment. This indicates that the proposed naloxone delivery system is adequate for opioid reversal during the naloxone release duration.


Subject(s)
Drug Carriers/chemistry , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Opiate Overdose/drug therapy , Animals , CHO Cells , Cricetulus , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Liberation , Humans , Microspheres , Naloxone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Surface Properties , Time Factors
7.
Cardiovasc Drugs Ther ; 35(4): 733-743, 2021 08.
Article in English | MEDLINE | ID: mdl-33484395

ABSTRACT

PURPOSE: Myocardial opioid receptors were demonstrated in animals and humans and seem to colocalize with membranous and sarcolemmal calcium channels of the excitation-contraction coupling in the left ventricle (LV). Therefore, this study investigated whether blockade of the cardiac opioid system by naltrexone would affect cardiac function and neurohumoral parameters in Wistar rats with volume overload-induced heart failure. METHODS: Volume overload in Wistar rats was induced by an aortocaval fistula (ACF). Left ventricular cardiac opioid receptors were identified by immunohistochemistry and their messenger ribonucleic acid (mRNA) as well as their endogenous ligand mRNA quantified by real-time polymerase chain reaction (RT-PCR). Following continuous delivery of either the opioid receptor antagonist naltrexone or vehicle via minipumps (n = 5 rats each), hemodynamic and humoral parameters were assessed 28 days after ACF induction. Sham-operated animals served as controls. RESULTS: In ACF rats mu-, delta-, and kappa-opioid receptors colocalized with voltage-gated L-type Ca2+ channels in left ventricular cardiomyocytes. Chronic naltrexone treatment of ACF rats reduced central venous pressure (CVP) and left ventricular end-diastolic pressure (LVEDP), and improved systolic and diastolic left ventricular functions. Concomitantly, rat brain natriuretic peptide (rBNP-45) and angiotensin-2 plasma concentrations which were elevated during ACF were significantly diminished following naltrexone treatment. In parallel, chronic naltrexone significantly reduced mu-, delta-, and kappa-opioid receptor mRNA, while it increased the endogenous opioid peptide mRNA compared to controls. CONCLUSION: Opioid receptor blockade by naltrexone leads to improved LV function and decreases in rBNP-45 and angiotensin-2 plasma levels. In parallel, naltrexone resulted in opioid receptor mRNA downregulation and an elevated intrinsic tone of endogenous opioid peptides possibly reflecting a potentially cardiodepressant effect of the cardiac opioid system during volume overload.


Subject(s)
Myocytes, Cardiac/drug effects , Naltrexone/pharmacokinetics , Angiotensin II/blood , Animals , Disease Models, Animal , Heart Function Tests , Narcotic Antagonists/pharmacokinetics , Nerve Tissue Proteins/metabolism , Rats , Rats, Wistar , Receptors, Opioid/metabolism , Treatment Outcome , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , Water Intoxication/metabolism , Water Intoxication/physiopathology
8.
Eur J Clin Pharmacol ; 77(12): 1901-1908, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34327552

ABSTRACT

PURPOSE: Remifentanil has been shown to increase the bioavailability of nasally administered naloxone. The aim of this study was to explore the nature of this observation. METHODS: We analysed samples from three pharmacokinetic studies to determine the serum concentrations of naloxone-3-glucuronide (N3G), the main metabolite of naloxone, with or without exposure to remifentanil. To enable direct comparison of the three studies, the data are presented as metabolic ratios (ratio of metabolite to mother substance, N3G/naloxone) and dose-corrected values of the area under the curve and maximum concentration (Cmax). RESULTS: Under remifentanil exposure, the time to maximum concentration (Tmax) for N3G was significantly higher for intranasal administration of 71 min compared to intramuscular administration of 40 min. The dose-corrected Cmax of N3G after intranasal administration of naloxone under remifentanil exposure was significantly lower (4.5 ng/mL) than in subjects not exposed to remifentanil (7.8-8.4 ng/mL). The metabolic ratios after intranasal administration rose quickly after 30-90 min and were 2-3 times higher at 360 min compared to intravenous and intramuscular administration. Remifentanil exposure resulted in a much slower increase of the N3G/naloxone ratio after intranasal administration compared to intranasal administration with the absence of remifentanil. After remifentanil infusion was discontinued, this effect gradually diminished. From 240 min there was no significant difference between the ratios observed after intranasal naloxone administration. CONCLUSION: Remifentanil increases the bioavailability of naloxone after nasal administration by reducing the pre-systemic metabolism of the swallowed part of the nasal dose.


Subject(s)
Analgesics, Opioid/pharmacology , Naloxone/analogs & derivatives , Naloxone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Remifentanil/pharmacology , Administration, Intranasal , Area Under Curve , Dose-Response Relationship, Drug , Healthy Volunteers , Humans , Injections, Intramuscular , Metabolic Clearance Rate , Naloxone/administration & dosage , Naloxone/blood , Narcotic Antagonists/administration & dosage
9.
Ther Drug Monit ; 42(2): 264-270, 2020 04.
Article in English | MEDLINE | ID: mdl-31929398

ABSTRACT

BACKGROUND: In the United States, drug addiction has become a nationwide health crisis. Recently, buprenorphine (BUP), a maintenance therapy approved by the Food and Drug Administration, has been increasingly used in pregnant women for the treatment of opioid use disorder. Pregnancy is associated with various anatomic and physiological changes, which may result in altered drug pharmacokinetics (PKs). Previously, we reported that dose-adjusted plasma concentrations of BUP are lower during pregnancy than after pregnancy. The mechanism(s) responsible for this difference has not yet been defined. Our study aimed to evaluate alterations in cytochromes P450 (CYP)- and uridine diphosphate glucunosyltransferases (UGT)-mediated metabolism of BUP during pregnancy to determine the mechanism(s) responsible for this observation. METHODS: Data from 2 clinical studies were included in the current analysis. Study 1 was a prospective, open-labeled, nonrandomized longitudinal BUP PK study in pregnant women with a singleton gestation, stabilized on twice-daily sublingual BUP opioid substitution therapy. Each subject participated in up to 3 studies during and after pregnancy (the second, third trimester, and postpartum). The design of study 2 was similar to study 1, with patients evaluated at different time points during the pregnancy (first, second-half of pregnancy), as well as during the postpartum period. In addition, the dosing frequency of BUP study 2 participants was not restricted to twice-daily dosing. At each study visit, blood samples were collected before a BUP dose, followed by multiple collection times (10-12) after the dose, for up to 12 hours or till the end of the dosing interval. Plasma concentrations of BUP and 3 metabolites were quantified using validated ultraperformance liquid chromatography-tandem mass spectrometric assays. RESULTS: In total, 19, 18, and 14 subjects completed the PK study during 1/2 trimester, third trimester, and postpartum, respectively. The AUC ratios of norbuprenorphine and norbuprenorphine glucuronide to buprenorphine, a measure of CYP3A mediated N-demethylation, were 1.89, 1.84, and 1.33 during the first and second, third trimesters, and postpartum, respectively. The AUC ratios of buprenorphine glucuronide to BUP, indicative of UGT activity, were 0.71, 2.07, and 0.3 at first/second trimesters, third trimester, and postpartum, respectively. Linear mixed-effect modeling analysis indicated that the AUC ratios of CYP- and UGT-mediated metabolism of BUP were significantly higher during pregnancy compared with postpartum. CONCLUSIONS: The CYP and UGT activities were significantly increased as determined by the metabolic ratios of BUP during pregnancy compared with the postpartum period. The increased UGT activity appeared to account for a substantial part of the observed change in metabolic activity during pregnancy. This is in agreement with the need for BUP dose increment in pregnant women to reach similar BUP exposure and therapeutic effect as in nonpregnant subjects.


Subject(s)
Buprenorphine/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Glucuronosyltransferase/metabolism , Narcotic Antagonists/pharmacokinetics , Adult , Buprenorphine/analogs & derivatives , Buprenorphine/blood , Cytochrome P-450 CYP3A/metabolism , Female , Humans , Longitudinal Studies , Narcotic Antagonists/therapeutic use , Opiate Substitution Treatment/methods , Opioid-Related Disorders/drug therapy , Postpartum Period/metabolism , Pregnancy , Pregnancy Trimesters/metabolism , Young Adult
10.
Drug Metab Dispos ; 47(7): 690-698, 2019 07.
Article in English | MEDLINE | ID: mdl-30992306

ABSTRACT

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.


Subject(s)
Naloxone/pharmacokinetics , Naltrexone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Administration, Intranasal , Adolescent , Adult , Cross-Over Studies , Double-Blind Method , Drug Interactions , Female , Humans , Male , Middle Aged , Naloxone/administration & dosage , Naloxone/blood , Naltrexone/administration & dosage , Naltrexone/blood , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/blood , Young Adult
11.
Nephrol Dial Transplant ; 34(4): 692-702, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30189012

ABSTRACT

BACKGROUND: The pharmacokinetics of oxycodone in patients with end-stage renal disease (ESRD) requiring haemodialysis are largely unknown. Therefore, we investigated the pharmacokinetics of oxycodone/naloxone prolonged release and their metabolites in patients with ESRD during and between haemodialysis sessions. METHODS: Single doses of oxycodone/naloxone (5/2.5 or 10/5 mg) were administered in nine patients with ESRD using a cross-over design on the day of dialysis and on a day between dialysis sessions. Plasma, dialysate and urine concentrations of oxycodone, naloxone and their metabolites were determined up to 48 h post-dosing using a liquid chromatography-tandem mass spectrometry system. RESULTS: Haemodialysis performed 6-10 h after dosing removed ∼10% of the administered dose of oxycodone predominantly as unconjugated oxycodone and noroxycodone or conjugated oxymorphone and noroxymorphone. The haemodialysis clearance of oxycodone based on its recovery in dialysate was (mean ± SD) 8.4 ± 2.1 L/h. The geometric mean (coefficient of variation) plasma elimination half-life of oxycodone during the 4-h haemodialysis period was 3.9 h (39%) which was significantly shorter than the 5.7 h (22%) without haemodialysis. Plasma levels of the active metabolite oxymorphone in its unconjugated form were very low. CONCLUSIONS: Oxycodone is removed during haemodialysis. The pharmacokinetics including the relatively short half-life of oxycodone in patients with ESRD with or without haemodialysis and the absence of unconjugated active metabolites indicate that oxycodone can be used at usual doses in patients requiring dialysis.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Kidney Failure, Chronic/drug therapy , Naloxone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Oxycodone/pharmacokinetics , Renal Dialysis/methods , Adult , Aged , Analgesics, Opioid/administration & dosage , Cross-Over Studies , Female , Humans , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/therapy , Kidney Function Tests , Male , Middle Aged , Morphinans/administration & dosage , Morphinans/pharmacokinetics , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Oxycodone/administration & dosage , Oxymorphone/administration & dosage , Oxymorphone/pharmacokinetics , Prognosis , Tissue Distribution
12.
Br J Anaesth ; 123(2): e204-e214, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30915992

ABSTRACT

BACKGROUND: Naloxone, an opioid receptor antagonist, is used as a pharmacological tool to detect tonic endogenous activation of opioid receptors in experimental pain models. We describe a pharmacokinetic model linking naloxone pharmacokinetics to its main metabolite after high-dose naloxone infusion. METHODS: Eight healthy volunteers received a three-stage stepwise high-dose i.v. naloxone infusion (total dose 3.25 mg kg-1). Naloxone and naloxone-3-glucuronide (N3G) plasma concentrations were sampled from infusion onset to 334 min after infusion discontinuation. Pharmacokinetic analysis was performed using non-linear mixed effect models (NONMEM). The predictive performances of Dowling's and Yassen's models were evaluated, and target-controlled infusion simulations were performed. RESULTS: Three- and two-compartment disposition models with linear elimination kinetics described the naloxone and N3G concentration time-courses, respectively. Two covariate models were developed: simple (weight proportional) and complex (with the shallow peripheral volume of distribution linearly increasing with body weight). The median prediction error (MDPE) and wobble for Dowling's model were -32.5% and 33.4%, respectively. For Yassen's model, the MDPE and wobble were 1.2% and 19.9%, respectively. CONCLUSIONS: A parent-metabolite pharmacokinetic model was developed for naloxone and N3G after high-dose naloxone infusion. No saturable pharmacokinetics were observed. Whereas Dowling's model was inaccurate and over-predicted naloxone concentrations, Yassen's model accurately predicted naloxone pharmacokinetics. The newly developed covariate models may be used for high-dose TCI-naloxone for experimental and clinical practice. CLINICAL TRIALS REGISTRATION: NCT01992146.


Subject(s)
Naloxone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Adolescent , Adult , Humans , Male , Young Adult
13.
Addict Biol ; 24(3): 438-446, 2019 05.
Article in English | MEDLINE | ID: mdl-29441714

ABSTRACT

Opioid addiction is a serious public health concern with severe health and social implications; therefore, extensive therapeutic efforts are required to keep users drug free. The two main pharmacological interventions, in the treatment of addiction, involve management with methadone an mu (µ)-opioid agonist and treatment with naltrexone, µ-opioid, kappa (κ)-opioid and delta (δ)-opioid antagonist. MET and NAL are believed to help individuals to derive maximum benefit from treatment and undergo a full recovery. The aim of this study was to determine the localization and distribution of MET and NAL, over a 24-hour period in rodent brain, in order to investigate the differences in their respective regional brain distributions. This would provide a better understanding of the role of each individual drug in the treatment of addiction, especially NAL, whose efficacy is controversial. Tissue distribution was determined by using mass spectrometric imaging (MSI), in combination with quantification via liquid chromatography tandem mass spectrometry. MSI image analysis showed that MET was highly localized in the striatal and hippocampal regions, including the nucleus caudate, putamen and the upper cortex. NAL was distributed with high intensities in the mesocorticolimbic system including areas of the cortex, caudate putamen and ventral pallidum regions. Our results demonstrate that MET and NAL are highly localized in the brain regions with a high density of µ-receptors, the primary sites of heroin binding. These areas are strongly implicated in the development of addiction and are the major pathways that mediate brain stimulation during reward.


Subject(s)
Brain/metabolism , Methadone/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Opioid-Related Disorders/metabolism , Animals , Caudate Nucleus/chemistry , Cerebral Cortex/chemistry , Hippocampus/chemistry , Male , Methadone/pharmacokinetics , Naltrexone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Opioid-Related Disorders/prevention & control , Putamen/chemistry , Rats, Sprague-Dawley
14.
Prehosp Emerg Care ; 23(5): 647-653, 2019.
Article in English | MEDLINE | ID: mdl-30596290

ABSTRACT

Objective: Naloxone is an opioid receptor antagonist that reverses life-threatening effects of opioid overdose. Since the 1970s, naloxone products have been developed as injectable solutions, and more recently as nasal sprays. Naloxone products have saved many lives in emergency settings. These products are routinely carried by public safety first-responders including fire fighters (FF), law enforcement officers (LEO), and emergency medical services (EMS). Now, they are also distributed through community access programs to the public. While public safety medications are monitored, those publically distributed are not, so expired products can be possibly found on-hand in an emergency. This study analyzed the quality and stability of expired Naloxone HCl Solutions for Injection, to assess their remaining efficacies and potential risks. Methods: The samples were collected from EMS or law enforcement training supplies and expired returns, with expiration dates ranging from 1990 to 2018. Using standardized techniques, the remaining naloxone was quantified, and the main degradation products, nornaloxone (also known as noroxymorphone) and other possible species, were monitored and quantified systematically. Results: Most tested samples were found containing more than 90% of labeled naloxone, including those stored for nearly 30 years. The naloxone degradation was slow, but generally correlated with storage time length. There was no significant amount of degradation products detected across all samples. Nornaloxone was detected from some older samples, but all less than 1%. Therefore, although it is an opioid agonist, the risk caused by nornaloxone should be low. Conclusion: This quality assessment demonstrates that expired naloxone products may still meet USP standards, even after many years. Further pharmaceutical, clinical, and regulatory investigation should be conducted to confirm our findings, especially for new naloxone products with different formulations and routes of administration. Extending the shelf-life of naloxone products may have important financial and public health consequences in addressing future drug shortages and meeting the needs for this critical drug.


Subject(s)
Emergency Medical Services , Emergency Service, Hospital , Morphinans/pharmacokinetics , Naloxone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Analgesics, Opioid/therapeutic use , Death , Drug Stability , Emergency Responders , Humans , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Police
15.
Am J Emerg Med ; 37(1): 143-150, 2019 01.
Article in English | MEDLINE | ID: mdl-30355476

ABSTRACT

INTRODUCTION: Opioid use disorder (OUD) is increasing in prevalence throughout the world, with approximately three million individuals in the United States affected. Buprenorphine is a medication designed, researched, and effectively used to assist in OUD recovery. OBJECTIVE: This narrative review discusses an approach to initiating buprenorphine in the emergency department (ED) for opioid-abuse recovery. DISCUSSION: Buprenorphine is a partial mu-opioid receptor agonist with high affinity and low intrinsic activity. Buprenorphine's long half-life, high potency, and 'ceiling effect' for both euphoric sensation and adverse effects make it an optimal treatment alternative for patients presenting to the ED with opioid withdrawal. While most commonly provided as a sublingual film or tablet, buprenorphine can also be delivered via transbuccal, transdermal, subdermal (implant), subcutaneous, and parenteral routes. Prior to ED administration, caution is recommended to avoid precipitation of buprenorphine-induced opioid withdrawal. Following the evaluation of common opioid withdrawal symptoms, a step-by-step approach to buprenorphine can by utilized to reach a sustained withdrawal relief. A multimodal medication-assisted treatment (MAT) plan involving pharmacologic treatment, as well as counseling and behavioral therapy, is essential to maintaining opioid remission. Patients may be safely discharged with safe-use counseling, close outpatient follow-up, and return precautions for continued management of their OUD. Establishing a buprenorphine program in the ED involves a multifactorial approach to establish a pro-buprenorphine culture. CONCLUSIONS: Buprenorphine is an evidence-based, safe, effective treatment option for OUD in an ED-setting. Though successfully utilized by many ED-based treatment programs, the stigma of 'replacing one opioid with another' remains a barrier. Evidence-based discussions on the safety and benefits of buprenorphine are essential to promoting a culture of acceptance and optimizing ED OUD treatment.


Subject(s)
Buprenorphine/therapeutic use , Emergency Service, Hospital , Narcotic Antagonists/therapeutic use , Opiate Substitution Treatment , Opioid-Related Disorders/drug therapy , Substance Withdrawal Syndrome/drug therapy , Behavior Therapy , Buprenorphine/adverse effects , Buprenorphine/pharmacokinetics , Buprenorphine/pharmacology , Combined Modality Therapy , Directive Counseling , Dosage Forms , Humans , Narcotic Antagonists/adverse effects , Narcotic Antagonists/pharmacokinetics , Narcotic Antagonists/pharmacology
16.
J Pharmacokinet Pharmacodyn ; 46(6): 513-529, 2019 12.
Article in English | MEDLINE | ID: mdl-31396799

ABSTRACT

The primary goal of this work was to develop a computational tool to enable personalized prediction of pharmacological disposition and associated responses for opioids and antidotes. Here we present a computational framework for physiologically-based pharmacokinetic (PBPK) modeling of an opioid (morphine) and an antidote (naloxone). At present, the model is solely personalized according to an individual's mass. These PK models are integrated with a minimal pharmacodynamic model of respiratory depression induction (associated with opioid administration) and reversal (associated with antidote administration). The model was developed and validated on human data for IV administration of morphine and naloxone. The model can be further extended to consider different routes of administration, as well as to study different combinations of opioid receptor agonists and antagonists. This work provides the framework for a tool that could be used in model-based management of pain, pharmacological treatment of opioid addiction, appropriate use of antidotes for opioid overdose and evaluation of abuse deterrent formulations.


Subject(s)
Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacokinetics , Antidotes/adverse effects , Antidotes/pharmacokinetics , Analgesics, Opioid/administration & dosage , Antidotes/administration & dosage , Humans , Male , Morphine/adverse effects , Morphine/pharmacokinetics , Naloxone/administration & dosage , Naloxone/adverse effects , Naloxone/pharmacokinetics , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/adverse effects , Narcotic Antagonists/pharmacokinetics , Opioid-Related Disorders/drug therapy , Pain/drug therapy , Receptors, Opioid/metabolism
17.
Br J Clin Pharmacol ; 84(9): 2075-2087, 2018 09.
Article in English | MEDLINE | ID: mdl-29873094

ABSTRACT

AIMS: Buprenorphine (BUP) is approved by the US Food and Drug Administration for the treatment of opioid addiction. The current dosing regimen of BUP in pregnant women is based on recommendations designed for nonpregnant adults. However, physiological changes during pregnancy may alter BUP exposure and efficacy. The objectives of this study were to develop a physiologically-based pharmacokinetic (PBPK) model for BUP in pregnant women, to predict changes in BUP exposure at different stages of pregnancy, and to demonstrate the utility of PBPK modelling in optimizing BUP pharmacotherapy during pregnancy. METHODS: A full PBPK model for BUP was initially built and validated in healthy subjects. A fetoplacental compartment was included as a combined compartment in this model to simulate pregnancy induced anatomical and physiological changes. Further, gestational changes in physiological parameters were incorporated in this model. The PBPK model predictions of BUP exposure in pregnancy and during the postpartum period were compared to published data from a prospective clinical study. RESULTS: The predicted BUP plasma concentration-time profiles in the virtual pregnant populations are consistent with the observed data in the 2nd and 3rd trimesters, and the postpartum period. The differences in the predicted means of dose normalized area under the plasma drug concentration-time curve up to 12 h, average concentration and maximum concentration were within ±25% of the corresponding observed means with the exception of average concentration in the 3rd trimester (-26.3%). CONCLUSION: PBPK model-based simulation may be a useful tool to optimize BUP pharmacotherapy during pregnancy, obviating the need to perform pharmacokinetic studies in each trimester and the postpartum period that normally require intensive blood sampling.


Subject(s)
Buprenorphine/pharmacokinetics , Models, Biological , Narcotic Antagonists/pharmacokinetics , Opiate Substitution Treatment/methods , Opioid-Related Disorders/rehabilitation , Pregnancy Complications/rehabilitation , Administration, Sublingual , Adult , Area Under Curve , Buprenorphine/administration & dosage , Buprenorphine/adverse effects , Computer Simulation , Female , Humans , Maternal-Fetal Exchange/drug effects , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/adverse effects , Opiate Substitution Treatment/adverse effects , Placenta/metabolism , Pregnancy , Prospective Studies , Tissue Distribution
18.
Eur J Clin Pharmacol ; 74(12): 1547-1553, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30143830

ABSTRACT

PURPOSE: Pharmacodynamic studies of naloxone require opioid agonism. Steady state condition may be achieved by remifentanil TCI (target controlled infusion). Opioid agonism can be measured by pupillometry. It is not known whether there are arteriovenous concentration differences for naloxone. The aim was thus to further develop a model for studying pharmacokinetic/pharmacodynamic aspects of naloxone and to explore whether a significant arteriovenous concentration difference for naloxone in humans was present. METHODS: Relevant authorities approved this study. Healthy volunteers (n = 12) were given 1.0 mg intravenous (IV) naloxone after steady state opioid agonism was obtained by TCI of remifentanil (1.3 ng/ml). Opioid effect was measured by pupillometry. Arterial and venous samples were collected simultaneously before and for 2 h after naloxone administration for quantification of naloxone and remifentanil. RESULTS: Arterial remifentanil was in steady state at 12 min. One milligram IV naloxone reversed the effect of remifentanil to 93% of pre-opioid pupil-size within 4 min. The estimated duration of antagonism was 118 min. At that time, the concentration of naloxone was 0.51 ng/ml. The time course of arterial and venous serum concentrations for naloxone was similar, although arterial AUC (area under the curve) was slightly lower (94%) than the venous AUC (p = 0.03). There were no serious adverse events. CONCLUSION: Onset of reversal by IV naloxone was rapid and lasted 118 min. The minimum effective concentration was 0.5 ng/ml. Using TCI remifentanil to obtain a steady-state opioid agonism may be a useful tool to compare new naloxone products.


Subject(s)
Analgesics, Opioid/pharmacology , Naloxone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Remifentanil/pharmacology , Adult , Arteries/metabolism , Drug Interactions , Female , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Pupil/drug effects , Veins/metabolism , Young Adult
19.
Eur J Clin Pharmacol ; 74(7): 873-883, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29568976

ABSTRACT

PURPOSE: This study aimed to develop a model for pharmacodynamic and pharmacokinetic studies of naloxone antagonism under steady-state opioid agonism and to compare a high-concentration/low-volume intranasal naloxone formulation 8 mg/ml to intramuscular 0.8 mg. METHODS: Two-way crossover in 12 healthy volunteers receiving naloxone while receiving remifentanil by a target-controlled infusion for 102 min. The group were subdivided into three different doses of remifentanil. Blood samples for serum naloxone concentrations, pupillometry and heat pain threshold were measured. RESULTS: The relative bioavailability of intranasal to intramuscular naloxone was 0.75. Pupillometry showed difference in antagonism; the effect was significant in the data set as a whole (p < 0.001) and in all three subgroups (p < 0.02-p < 0.001). Heat pain threshold showed no statistical difference. CONCLUSIONS: A target-controlled infusion of remifentanil provides good conditions for studying the pharmacodynamics of naloxone, and pupillometry was a better modality than heat pain threshold. Intranasal naloxone 0.8 mg is inferior for a similar dose intramuscular. Our design may help to bridge the gap between studies in healthy volunteers and the patient population in need of naloxone for opioid overdose. TRIAL REGISTRATION: clinicaltrials.gov : NCT02307721.


Subject(s)
Analgesics, Opioid/administration & dosage , Models, Biological , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Piperidines/administration & dosage , Administration, Intranasal , Adult , Analgesics, Opioid/pharmacology , Cross-Over Studies , Female , Healthy Volunteers , Humans , Injections, Intramuscular , Male , Miosis/chemically induced , Miosis/drug therapy , Naloxone/blood , Naloxone/pharmacokinetics , Naloxone/pharmacology , Narcotic Antagonists/blood , Narcotic Antagonists/pharmacokinetics , Narcotic Antagonists/pharmacology , Pain/drug therapy , Piperidines/pharmacology , Pupil/drug effects , Remifentanil , Young Adult
20.
Drug Dev Res ; 79(5): 234-238, 2018 08.
Article in English | MEDLINE | ID: mdl-30188587

ABSTRACT

Preclinical Research & Development Background: Samidorphan, a µ-opioid receptor antagonist, is in clinical development for central nervous system related diseases. The discriminative stimulus effects of samidorphan were assessed in rats trained to discriminate the effects of a known morphinan of abuse, morphine, from that of saline. METHODS: Escalating doses of samidorphan were substituted for morphine and rats were allowed to respond on two levers for food reward. Doses of samidorphan were chosen based on other pharmacodynamic assays in which samidorphan blocked the effects of morphine (such as blocking analgesia in a hot plate test; data not shown). In addition, a pharmacokinetic study was conducted to determine if these doses would reflect predicted exposure levels that translate to human equivalent doses. RESULTS: Rats discriminating morphine from vehicle responded predominantly on the vehicle lever after receiving samidorphan. In addition, samidorphan was rapidly absorbed, and plasma concentrations of the doses tested in this study bracket therapeutically relevant concentrations. CONCLUSIONS: In summary, samidorphan produced saline-like behavioral responses over a wide dose range.


Subject(s)
Discrimination, Psychological , Naltrexone/analogs & derivatives , Narcotic Antagonists/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Humans , Male , Morphine/pharmacology , Naltrexone/pharmacokinetics , Naltrexone/pharmacology , Narcotic Antagonists/pharmacokinetics , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL