Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 233
Filter
Add more filters

Publication year range
1.
Arch Toxicol ; 95(12): 3681-3693, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34604914

ABSTRACT

New synthetic opioids (NSOs) pose a public health concern since their emergence on the illicit drug market and are gaining increasing importance in forensic toxicology. Like many other new psychoactive substances, NSOs are consumed without any preclinical safety data or any knowledge on toxicokinetic (TK) data. Due to ethical reasons, controlled human TK studies cannot be performed for the assessment of these relevant data. As an alternative animal experimental approach, six pigs per drug received a single intravenous dose of 100 µg/kg body weight (BW) of U-47700 or 1000 µg/kg BW of tramadol to evaluate whether this species is suitable to assess the TK of NSOs. The drugs were determined in serum and whole blood using a fully validated method based on solid-phase extraction and LC-MS/MS. The concentration-time profiles and a population (pop) TK analysis revealed that a three-compartment model best described the TK data of both opioids. Central volumes of distribution were 0.94 L/kg for U-47700 and 1.25 L/kg for tramadol and central (metabolic) clearances were estimated at 1.57 L/h/kg and 1.85 L/h/kg for U-47700 and tramadol, respectively. The final popTK model parameters for pigs were upscaled via allometric scaling techniques. In comparison to published human data, concentration-time profiles for tramadol could successfully be predicted with single species allometric scaling. Furthermore, possible profiles for U-47700 in humans were simulated. The findings of this study indicate that unlike a multiple species scaling approach, pigs in conjunction with TK modeling are a suitable tool for the assessment of TK data of NSOs and the prediction of human TK data.


Subject(s)
Benzamides/pharmacokinetics , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Tramadol/pharmacokinetics , Administration, Intravenous , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/toxicity , Animals , Benzamides/toxicity , Humans , Illicit Drugs/pharmacokinetics , Illicit Drugs/toxicity , Male , Models, Biological , Species Specificity , Swine , Tissue Distribution , Toxicokinetics , Tramadol/toxicity
2.
Addict Biol ; 26(2): e12906, 2021 03.
Article in English | MEDLINE | ID: mdl-32378298

ABSTRACT

Naphthylpyrovalerone (naphyrone) is a pyrovalerone cathinone that potently inhibits monoamine transporters and provides stimulatory-entactogenic effects. Little is known about the safety of naphyrone or its effects in vivo, and more research is needed to acquire knowledge about its fundamental effects on physiology and behaviour. Our objective was to investigate naphyrone's pharmacokinetics, acute toxicity, hyperthermic potential and stimulatory and psychotomimetic properties in vivo in male Wistar rats. Pharmacokinetics after 1 mg/kg subcutaneous (sc.) naphyrone were measured over 6 h in serum, the brain, liver and lungs. Rectal temperature (degree Celsius) was measured over 10 h in group-versus individually housed rats after 20 mg/kg sc. In the behavioural experiments, 5, 10 or 20 mg/kg of naphyrone was administered 15 or 60 min prior to testing. Stimulation was assessed in the open field, and sensorimotor processing in a prepulse inhibition (PPI) task. Peak concentrations of naphyrone in serum and tissue were reached at 30 min, with a long-lasting elevation in the brain/serum ratio, consistent with observations of lasting hyperlocomotion in the open field and modest increases in body temperature. Administration of 20 mg/kg transiently enhanced PPI. Naphyrone crosses the blood-brain barrier rapidly and is eliminated slowly, and its long-lasting effects correspond to its pharmacokinetics. No specific signs of acute toxicity were observed; therefore, clinical care and harm-reduction guidance should be in line with that available for other stimulants and cathinones.


Subject(s)
Body Temperature Regulation/drug effects , Central Nervous System Stimulants/pharmacokinetics , Illicit Drugs/pharmacokinetics , Pentanones/pharmacokinetics , Pyrrolidines/pharmacokinetics , Animals , Body Temperature/drug effects , Central Nervous System Stimulants/pharmacology , Illicit Drugs/pharmacology , Male , Pentanones/pharmacology , Pyrrolidines/pharmacology , Rats , Rats, Wistar
3.
Drug Metab Rev ; 52(1): 44-65, 2020 02.
Article in English | MEDLINE | ID: mdl-31826670

ABSTRACT

Adverse pharmacokinetic interactions between illicit substances and clinical drugs are of a significant health concern. Illicit substances are taken by healthy individuals as well as by patients with medical conditions such as mental illnesses, acquired immunodeficiency syndrome, diabetes mellitus and cancer. Many individuals that use illicit substances simultaneously take clinical drugs meant for targeted treatment. This concomitant usage can lead to life-threatening pharmacokinetic interactions between illicit substances and clinical drugs. Optimal levels and activity of drug-metabolizing enzymes and drug-transporters are crucial for metabolism and disposition of illicit substances as well as clinical drugs. However, both illicit substances and clinical drugs can induce changes in the expression and/or activity of drug-metabolizing enzymes and drug-transporters. Consequently, with concomitant usage, illicit substances can adversely influence the therapeutic outcome of coadministered clinical drugs. Likewise, clinical drugs can adversely affect the response of coadministered illicit substances. While the interactions between illicit substances and clinical drugs pose a tremendous health and financial burden, they lack a similar level of attention as drug-drug, food-drug, supplement-drug, herb-drug, disease-drug, or other substance-drug interactions such as alcohol-drug and tobacco-drug interactions. This review highlights the clinical pharmacokinetic interactions between clinical drugs and commonly used illicit substances such as cannabis, cocaine and 3, 4-Methylenedioxymethamphetamine (MDMA). Rigorous efforts are warranted to further understand the underlying mechanisms responsible for these clinical pharmacokinetic interactions. It is also critical to extend the awareness of the life-threatening adverse interactions to both health care professionals and patients.


Subject(s)
Illicit Drugs/pharmacokinetics , Prescription Drugs/pharmacokinetics , Animals , Drug Interactions , Humans , Illicit Drugs/adverse effects , Illicit Drugs/pharmacology , Prescription Drugs/adverse effects , Prescription Drugs/pharmacology , Substance-Related Disorders/metabolism
4.
Liver Int ; 40(1): 32-44, 2020 01.
Article in English | MEDLINE | ID: mdl-31654604

ABSTRACT

The hepatitis C virus (HCV) prevalence is extremely high in patients who consume and inject illicit drugs. Concerns about poor adherence and fear of interaction with drugs of abuse could constitute further disincentive for treatment initiation in these patients. We discussed the pharmacokinetics (PKs) and pharmacodynamics (PD) of currently prescribed direct antiviral agents (NSA5 inhibitors: daclatasvir, elbasvir, ledipasvir, pibrentasvir, velpatasvir; NS5B inhibitor: sofosbuvir; NS3/4A protease inhibitors: glecaprevir, grazoprevir, voxilaprevir) and most common substances of abuse (opioids: buprenorphine, fentanyl, heroin, methadone, morphine, oxycodone; stimulants: amphetamines, cathinones, cocaine; cannabinoids; ethanol). Overall, most direct-acting antivirals (DAAs) are substrates and inhibitors of the transmembrane transporter P-glycoprotein (P-gp), and several of them are metabolized by cytochrome P450 enzymes. Clinically relevant interactions are associated with P-gp and CYP3A modulators. Most substances of abuse are eliminated by Phase I and Phase II metabolizing enzymes, but none of them are either major inhibitors or inducers. PK studies did not show any relevant interactions between DAA and methadone or buprenorphine. Based on pharmacological considerations, neither efficacy loss nor adverse drug event associated with detrimental interaction are expected with opioids, stimulants, cannabinoids and ethanol. In summary, our literature review shows that the interaction potential of DAA with most opioids and illicit drugs is limited and should not be a hurdle to the initiate DAA.


Subject(s)
Analgesics, Opioid/adverse effects , Antiviral Agents/adverse effects , Ethanol/adverse effects , Hepatitis C, Chronic/drug therapy , Illicit Drugs/adverse effects , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Drug Interactions , Ethanol/pharmacokinetics , Humans , Illicit Drugs/pharmacokinetics , Substance-Related Disorders/complications
5.
Xenobiotica ; 49(12): 1388-1395, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30739533

ABSTRACT

Recently, there has been a rise in abuse of synthetic cannabinoids (SCBs). The consumption of SCBs results in various effects and can induce toxic reactions, including paranoia, seizures, tachycardia and even death. 1-Naphthyl 1-(4-fluorobenzyl)-1H-indole-3-carboxylate (FDU-PB-22) is a third generation SCB whose metabolic pathway has not been fully characterized. In this study, we conducted in vitro pharmacokinetic analysis of FDU-PB-22 metabolism. Metabolic reactions containing FDU-PB-22 and human liver microsomes (HLMs) were independent of NADPH but not UDP-glucuronic acid (UDPGA), suggesting that UDP-glucuronosyltransferases (UGTs) are the primary enzymes involved in this metabolism. It was further determined that the metabolite extensively formed after incubating FDU-PB-22 with UDPGA in HLMs was the glucuronide of FDU-PB-22 3-carboxyindole (FBI-COOH). Various hepatic UGTs showed enzymatic activity for FBI-COOH. A series of UGT inhibitors showed moderate to strong inhibition of FBI-COOH-glucuronidation in HLMs, suggesting that multiple UGT isoforms are involved in FBI-COOH-glucuronidation in the liver. Interestingly, an extra-hepatic isoform, UGT1A10, exhibited the highest activity with a Km value of 38 µM and a Vmax value of 5.90 nmol/min/mg. Collectively, these results suggest that both genetic mutations of and the co-administration of inhibitors for FDU-PB-22-metabolizing UGTs will likely increase the risk of FDU-PB-22-induced toxicity.


Subject(s)
Cannabinoids/chemistry , Cannabinoids/pharmacokinetics , Indoles/chemistry , Indoles/pharmacokinetics , Microsomes, Liver/enzymology , Enzyme Inhibitors/pharmacology , Glucuronosyltransferase/antagonists & inhibitors , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Illicit Drugs/metabolism , Illicit Drugs/pharmacokinetics , Inactivation, Metabolic , Microsomes, Liver/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Uridine Diphosphate Glucuronic Acid/metabolism
6.
Drug Metab Rev ; 50(2): 125-139, 2018 05.
Article in English | MEDLINE | ID: mdl-29540067

ABSTRACT

New psychoactive substances (NPS), often referred as 'legal highs' or 'designer drugs', are derivatives and analogs of existing psychoactive drugs that are introduced in the recreational market to circumvent existing legislation on drugs of abuse. This work aims to review the state-of-the-art regarding chemical, molecular pharmacology, and in vitro and in vivo data on toxicokinetics of the potent synthetic cathinone α-pyrrolidinovalerophenone (α-PVP or flakka or zombie drug). Chemical, pharmacological, toxicological, and clinical effects of α-PVP were searched in PubMed (U.S. National Library of Medicine) and governmental websites without limitation of the period. α-PVP is a wide spread and easy to get special type of synthetic cathinone with seemingly powerful cocaine-like stimulant effects, high brain penetration, high liability for abuse and with increased risk of adverse effects such as tachycardia, agitation, hypertension, hallucinations, delirium, mydriasis, self-injury, aggressive behavior, and suicidal ideations. α-PVP undergoes extensive metabolism via different pathways and the α-PVP itself or its metabolites ß-hydroxy-α-PVP and α-PVP lactam represent the main targets for toxicological analysis in urine. There is a limited knowledge regarding the short- and long-term effects of α-PVP and metabolites, and pharmacogenetic influence, hence further clinical and forensic toxicological studies are required. Moreover, since α-PVP cannot be detected with classic routine analysis procedures, statements on the frequency of their consumption cannot be made.


Subject(s)
Illicit Drugs/pharmacology , Illicit Drugs/pharmacokinetics , Psychotropic Drugs/pharmacology , Psychotropic Drugs/pharmacokinetics , Pyrrolidines/pharmacology , Pyrrolidines/pharmacokinetics , Animals , Designer Drugs/pharmacokinetics , Designer Drugs/pharmacology , Humans , Illicit Drugs/chemistry , Psychotropic Drugs/chemistry , Pyrrolidines/chemistry , Structure-Activity Relationship
7.
J Pharmacol Exp Ther ; 367(3): 543-550, 2018 12.
Article in English | MEDLINE | ID: mdl-30266766

ABSTRACT

Novel synthetic cannabinoids are appearing in recreational drug markets worldwide. Pharmacological characterization of these new drugs is needed to inform clinicians, toxicologists, and policy makers who monitor public health. [1-(5-Fluoropentyl)-1H-indol-3-yl](1-naphthyl)methanone (AM-2201) is an abused synthetic cannabinoid that was initially created as a research tool for investigating the endocannabinoid system. Here we measured the pharmacodynamic effects of AM-2201 in rats, and simultaneously determined plasma pharmacokinetics for the parent drug and its metabolites. Male Sprague-Dawley rats were fitted with surgically implanted temperature transponders and indwelling jugular catheters under pentobarbital anesthesia. One week later, rats received subcutaneous injection of AM-2201 (0.1, 0.3, and 1.0 mg/kg) or its vehicle, and serial blood specimens were withdrawn via catheters. Core temperatures and catalepsy were measured just prior to each blood withdrawal, and plasma was assayed for drug and metabolites using liquid chromatography-tandem mass spectrometry. We found that AM-2201 produced dose-related hypothermia and catalepsy that peaked at 2 hours and lasted up to 8 hours. AM-2201 plasma concentrations rose linearly with increasing dose and ranged from 0.14 to 67.9 µg/l. Concentrations of three metabolites, AM-2201 N-(4-hydroxypentyl) (≤0.17 µg/l), naphthalen-1-yl-(1-pentylindol-3-yl)methanone (JWH-018) N-(5-hydroxypentyl) (≤1.14 µg/l), and JWH-018 N-pentanoic acid (≤0.88 µg/l) were detectable but much lower. Peak AM-2201, JWH-018 N-(5-hydroxypentyl), and JWH-018 N-pentanoic acid concentrations occurred at 1.3, 2.4, and 6.5 hours, respectively. Concentrations of AM-2201, JWH-018 N-(5-hydroxypentyl), and JWH-018 N-pentanoic acid were negatively correlated with body temperature, but, given the low concentrations of metabolites detected, AM-2201 is likely the major contributor to pharmacodynamic effects under our experimental conditions.


Subject(s)
Cannabinoids/pharmacology , Cannabinoids/pharmacokinetics , Indoles/pharmacology , Indoles/pharmacokinetics , Animals , Chromatography, Liquid/methods , Illicit Drugs/pharmacokinetics , Illicit Drugs/pharmacology , Indoles/metabolism , Male , Naphthalenes/metabolism , Pentanoic Acids/metabolism , Rats , Rats, Sprague-Dawley , Substance-Related Disorders/metabolism , Tandem Mass Spectrometry/methods
8.
Int J Legal Med ; 132(6): 1645-1657, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30219927

ABSTRACT

This case report centres on the post-mortem distribution of the synthetic cannabinoid MDMB-CHMICA and its metabolites in the case of a 27-year-old man found dead after falling from the 24th floor of a high-rise building. Toxicological analysis of post-mortem samples confirmed, besides consumption of the synthetic cannabinoids MDMB-CHMICA (1.7 ng/mL femoral blood) and EG-018, the abuse of THC (9.3 ng/mL femoral blood), amphetamine (1050 ng/mL femoral blood), MDMA (275 ng/mL femoral blood), and cocaine. Regarding EG-018 and cocaine, only traces were detected in heart blood as well as in the brain (EG-018) and urine (cocaine), respectively, which is why no quantification was conducted in the femoral blood sample. It was concluded from femoral blood analysis that, at the time of death, the man was under the influence of the synthetic cannabinoid MDMB-CHMICA, THC, amphetamine and MDMA. Comprehensive screenings of all post-mortem specimens were conducted to elucidate the post-mortem distribution of MDMB-CHMICA and its metabolites. The MDMB-CHMICA concentrations ranged between 0.01 ng/mL (urine) and 5.5 ng/g (brain). Comparably low concentrations were detected in cardiac and femoral blood (2.1 ng/mL and 1.7 ng/mL, respectively) as well as in the psoas major muscle (1.2 ng/g). Higher concentrations were found in the lung (2.6 ng/g), liver (2.6 ng/g), and kidney (3.8 ng/g). Gastric content yielded a MDMB-CHMICA concentration of 2.4 ng/g (1.1 µg absolute). Screening for MDMB-CHMICA metabolites resulted in the detection of mainly monohydroxylated metabolites in the blood, kidney, and liver specimens. Results indicated that monohydroxylated metabolites of MDMB-CHMICA are appropriate target analytes for detecting MDMB-CHMICA intake.


Subject(s)
Cannabinoids/analysis , Cannabinoids/pharmacokinetics , Illicit Drugs/analysis , Illicit Drugs/pharmacokinetics , Adult , Brain Chemistry , Forensic Toxicology , Gastrointestinal Contents/chemistry , Humans , Kidney/chemistry , Liver/chemistry , Lung/chemistry , Male , Psoas Muscles/chemistry , Substance Abuse Detection , Substance-Related Disorders/metabolism
9.
Ther Drug Monit ; 40(1): 38-51, 2018 02.
Article in English | MEDLINE | ID: mdl-29120973

ABSTRACT

Illicit fentanyl and its analogues are very dangerous synthetic opioids, with high abuse potential and severe adverse effects including coma and death. They are used as adulterants in street heroin, cocaine, and methamphetamine, or as heroin substitutes sold to unaware users with a high risk of overdoses. Fentanyl and its analogues have also been identified in counterfeit medicinal products, such as oxycodone, hydrocodone, and alprazolam tablets, or as components of speedball mixtures together with cocaine or other stimulants. In recent years, a number of epidemics involving acute intoxications and deaths related to illicit fentanyl or its analogues have occurred in the United States, Europe, Canada, Australia, and Japan. In several cases, fatalities involved polysubstance use. A review of the most recent case reports or case series of acute intoxications and fatalities involving illicit fentanyl and its newest analogues is herein provided, together with the available information on intoxication symptoms, eventual death cause, and metabolites detected in different biological fluids and reported concentrations.


Subject(s)
Cause of Death , Fentanyl/adverse effects , Fentanyl/pharmacokinetics , Illicit Drugs/adverse effects , Illicit Drugs/pharmacokinetics , Analgesics, Opioid/adverse effects , Drug Contamination , Fentanyl/analogs & derivatives , Humans , Tissue Distribution
10.
Epilepsy Behav ; 88: 162-171, 2018 11.
Article in English | MEDLINE | ID: mdl-30286443

ABSTRACT

RATIONALE: Treatment with a highly purified oral solution of cannabidiol (CBD), derived from the plant Cannabis sativa L., demonstrated some evidence of central nervous system (CNS)-related adverse events in patients enrolled in phase 3 trials for treatment of childhood-onset epilepsy. Cannabidiol was categorized as a Schedule 1 substance by the United States Drug Enforcement Administration; therefore, it was important to test CBD for human abuse potential. METHODS: This was a single-dose, randomized, double-blind, double-dummy, placebo- and active-controlled crossover trial. The abuse potential of single oral doses of plant-derived pharmaceutical formulations of highly purified CBD (Epidiolex®; 750 mg, 1500 mg, and 4500 mg) was compared with that of single oral doses of alprazolam (2 mg), dronabinol (10 mg and 30 mg), and placebo in healthy recreational polydrug users. The primary endpoint to assess abuse potential was the maximum effect (Emax) on Drug-Liking visual analog scale (VAS). Other measurements included Emax on Overall Drug-Liking VAS, Take Drug Again VAS, positive and negative effects, other subjective effects, and Drug Similarity VAS. Cognitive and psychomotor functions were assessed using the Divided Attention Test, the Hopkins Verbal Learning Test-Revised, and the Digit-Symbol Substitution Task. Pharmacokinetic parameters were determined for CBD and its major metabolites. Standard safety measures and adverse events were assessed. PRINCIPAL RESULTS: Of 95 eligible subjects, 43 qualified for the treatment phase, received at least 1 dose of investigational medicinal product, and were included in safety assessments; 35 subjects were included in the pharmacodynamic analysis. Subjects receiving alprazolam and dronabinol had significantly higher Drug-Liking Emax (P < 0.0001) compared with those receiving placebo, confirming study validity. Compared with placebo, Drug-Liking was not significantly different for subjects taking 750-mg CBD (P = 0.51). Drug-Liking Emax values for 1500-mg and 4500-mg CBD were significantly different from placebo (P = 0.04 and 0.002, respectively); however, the mean differences were <10 points on VAS compared with >18-point differences between positive controls and placebo. Alprazolam and dronabinol had significantly higher Drug-Liking, Overall-Liking, and Take Drug Again VAS Emax values compared with all doses of CBD (P ≤ 0.004). In contrast to alprazolam, CBD administration had no observable effect on cognitive/psychomotor tests. Pharmacokinetic parameters for CBD in this trial were consistent with previous studies. The majority of adverse events reported during the trial were of mild or moderate severity; no serious adverse events or deaths were reported. CONCLUSION: Administration of a therapeutic dose of CBD (750 mg) showed significantly low abuse potential in a highly sensitive population of polydrug users. Although high and supratherapeutic doses of CBD (1500 mg and 4500 mg, respectively) had detectable subjective effects compared with placebo; the effects were significantly lower than those observed with alprazolam and dronabinol.


Subject(s)
Cannabidiol/adverse effects , Illicit Drugs/adverse effects , Substance-Related Disorders/etiology , Adolescent , Adult , Cannabidiol/pharmacokinetics , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Illicit Drugs/pharmacokinetics , Male , Middle Aged , Risk Assessment , Substance-Related Disorders/prevention & control , Substance-Related Disorders/psychology , Young Adult
11.
Arch Toxicol ; 92(9): 2875-2884, 2018 09.
Article in English | MEDLINE | ID: mdl-30083819

ABSTRACT

Transporter-mediated drug-drug interactions (DDI) may induce adverse clinical events. As drugs of abuse (DOA) are marketed without preclinical safety studies, only very limited information about interplay with membrane transporters are available. Therefore, 13 DOA of various classes were tested for their in vitro affinity to the human breast cancer resistance protein (hBCRP), an important efflux transporter. As adenosine 5'-triphosphate (ATP) hydrolysis is crucial for hBCRP activity, adenosine 5'-diphosphate (ADP) formation was measured and used as in vitro marker for hBCRP ATPase activity. ADP quantification was performed by hydrophilic interaction liquid chromatography coupled to high-resolution tandem mass spectrometry and its amount in test compound incubations was compared to that in reference incubations using the hBCRP substrate sulfasalazine or the hBCRP inhibitor orthovanadate. If DOA caused stimulation or inhibition, further investigations such as Michaelis-Menten kinetic modeling or IC50 value determination were conducted. Among the tested DOA, seven compounds showed statistically significant hBCRP ATPase stimulation. The entactogen 3,4-BDB and the plant alkaloid mitragynine were identified as strongest stimulators. Their affinity to the hBCRP ATPase was lower than that of sulfasalazine but comparable to that of rosuvastatin, another hBCRP model substrate. Five DOA showed statistically significant hBCRP ATPase inhibition. Determination of IC50 values identified the synthetic cannabinoid receptor agonists JWH-200 and WIN 55,212-2 as the strongest inhibitors comparable to orthovanadate. The present study clearly demonstrated that tested DOA show in part high affinities to the hBCRP within the range of model substrates or inhibitors. Thus, there is a risk of hBCRP-mediated DDI, which needs to be considered in clinical settings.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Illicit Drugs/pharmacokinetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Drug Interactions , Humans , Rosuvastatin Calcium/pharmacokinetics , Sulfasalazine/pharmacokinetics , Vanadates/pharmacokinetics
12.
Handb Exp Pharmacol ; 252: 441-459, 2018.
Article in English | MEDLINE | ID: mdl-29476337

ABSTRACT

This summarizing and descriptive review article is an update on previously published reviews. It covers English-written and PubMed-listed review articles and original studies published between May 2016 and November 2017 on the toxicokinetics of new psychoactive substances (NPS). Compounds covered include stimulants and entactogens, synthetic cannabinoids, tryptamines, phenethylamine and phencyclidine-like drugs, benzodiazepines, and opioids. First, an overview and discussion is provided on selected review articles followed by an overview and discussion on selected original studies. Both sections are then concluded by an opinion on these latest developments. The present review shows that the NPS market is still highly dynamic and that studies regarding their toxicokinetics are necessary to understand risks associated with their consumption. Data collection and studies are encouraged to allow for detection of NPS in biological matrices in cases of acute intoxications or chronic consumption. Although some data are available, scientific papers dealing with the mechanistic reasons behind acute and chronic toxicity are still lacking.


Subject(s)
Psychotropic Drugs/pharmacokinetics , Psychotropic Drugs/toxicity , Central Nervous System Stimulants/pharmacokinetics , Central Nervous System Stimulants/toxicity , Humans , Illicit Drugs/pharmacokinetics , Illicit Drugs/toxicity , Substance-Related Disorders , Toxicokinetics
13.
J Neurovirol ; 23(4): 603-614, 2017 08.
Article in English | MEDLINE | ID: mdl-28762183

ABSTRACT

Drug abuse (e.g., methamphetamine-Meth or cocaine-Coc) is one of the major risk factors for becoming infected with HIV-1, and studies show that in combination, drug abuse and HIV-1 lead to significantly greater damage to CNS. To overcome these issues, we have developed a novel nanoformulation (NF) for drug-abusing population infected with HIV-1. In this work, a novel approach was developed for the co-encapsulation of Nelfinavir (Nel) and Rimcazole (Rico) using layer-by-layer (LbL) assembled magnetic nanoformulation for the cure of neuroAIDS. Developed NF was evaluated for blood-brain barrier (BBB) transmigration, cell uptake, cytotoxicity and efficacy (p24 assay) in HIV-1 infected primary astrocyte (HA) in presence or absence of Coc and Meth. Developed magnetic nanoformulation (NF) fabricated using the LbL approach exhibited higher amounts of drug loading (Nel and Rico) with 100% release of both the therapeutic agents in a sustained manner for 8 days. NF efficacy studies indicated a dose-dependent decrease in p24 levels in HIV-1-infected HA (~55%) compared to Coc + Meth treated (~50%). The results showed that Rico significantly subdued the effect of drugs of abuse on HIV infectivity. NF successfully transmigrated (38.8 ± 6.5%) across in vitro BBB model on the application of an external magnetic field and showed >90% of cell viability with efficient cell uptake. In conclusion, our proof of concept study revealed that sustained and concurrent release of sigma σ1 antagonist and anti-HIV drug from the developed novel sustained release NF can overcome the exacerbated effects of drugs of abuse in HIV infection and may solve the issue of medication adherence in the drug-abusing HIV-1 infected population.


Subject(s)
Carbazoles/pharmacokinetics , Cocaine/pharmacokinetics , Delayed-Action Preparations/pharmacokinetics , Illicit Drugs/pharmacokinetics , Methamphetamine/pharmacokinetics , Nelfinavir/pharmacokinetics , AIDS Dementia Complex/drug therapy , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cocaine/chemistry , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , HIV Protease Inhibitors/pharmacokinetics , HIV-1/drug effects , HIV-1/growth & development , Humans , Illicit Drugs/chemistry , Magnets/chemistry , Methamphetamine/chemistry , Nanostructures/chemistry , Neuroprotective Agents/pharmacokinetics , Primary Cell Culture , Substance Abuse, Intravenous/prevention & control
14.
Pain Med ; 18(1): 61-77, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27330154

ABSTRACT

Objective: To compare the oral abuse potential of hydrocodone extended-release (ER) tablet developed with CIMA ® Abuse-Deterrence Technology with that of hydrocodone immediate release (IR). Design: Randomized, double-blind, placebo-controlled, crossover study. Setting and Patients: One study site in the United States; adult nondependent, recreational opioid users. Methods: After confirming their ability to tolerate and discriminate hydrocodone IR 45 mg from placebo, eligible participants were randomized to receive each of the following oral treatments once: finely crushed placebo, hydrocodone IR 45-mg powder, intact hydrocodone ER 45-mg tablet, and finely crushed hydrocodone ER 45-mg tablet. Primary pharmacodynamic measure was "at the moment" drug liking. Secondary measures included overall drug liking, drug effects (e.g., balance, positive, negative, sedative), pupillometry, pharmacokinetics, and safety. Results: Mean maximum effect (E max ) for "at the moment" drug liking was significantly lower for intact (53.9) and finely crushed hydrocodone ER (66.9) vs. hydrocodone IR (85.2; P < 0.001). Drug liking for intact hydrocodone ER was comparable to placebo (E max : 53.9 vs. 53.2). Secondary measures were consistent with these results, indicating that positive, negative, and sedative drug effects were diminished with intact and crushed hydrocodone ER tablet vs. hydrocodone IR. The 72-hour plasma concentration-time profile for each treatment mimicked its respective "at the moment" drug-liking-over-time profile. Incidence of adverse events was lower with intact hydrocodone ER (53%) vs. hydrocodone IR (79%) and finely crushed hydrocodone ER (73%). Conclusions: The oral abuse potential of hydrocodone ER (intact and finely crushed) was significantly lower than hydrocodone IR in healthy, nondependent, recreational opioid users. Hydrocodone ER was generally well tolerated.


Subject(s)
Hydrocodone/administration & dosage , Hydrocodone/pharmacokinetics , Opioid-Related Disorders , Administration, Oral , Adolescent , Adult , Cross-Over Studies , Delayed-Action Preparations/administration & dosage , Double-Blind Method , Female , Humans , Illicit Drugs/pharmacokinetics , Male , Powders , Tablets , Young Adult
15.
Pain Med ; 17(5): 820-31, 2016 05.
Article in English | MEDLINE | ID: mdl-26814240

ABSTRACT

OBJECTIVES: A once-daily, extended-release hydrocodone bitartrate tablet with abuse-deterrent properties (Hysingla ER® [HYD]) is available for the treatment of chronic pain in appropriate patients. This study evaluated the intranasal abuse potential and pharmacokinetics of HYD coarse and fine particles vs hydrocodone powder or placebo. DESIGN: Single-center, double-blind, positive- and placebo-controlled, randomized, four-treatment crossover study. SUBJECTS: Healthy adult, nondependent, recreational opioid users with a history of intranasal abuse. METHODS: During four treatment periods, subjects (N = 31) received hydrocodone powder 60 mg, HYD coarse particles 60 mg, HYD fine particles 60 mg, or placebo, with five-to-seven-day washouts between treatments. Measures over 36 hours postdose included drug-liking and willingness to take drug again, assessed using visual analog scales (VASs), pupillometry, intranasal irritation, and pharmacokinetics. RESULTS: Insufflation of both HYD coarse and fine particles led to lower "At this Moment" Drug Liking VAS peak values compared with hydrocodone powder, but higher values compared with placebo (P < 0.001 for all comparisons). Similar results were observed for Overall Drug Liking VAS, Take Drug Again VAS, and Subjective Drug Value. Compared with hydrocodone, insufflation of HYD particles led to reduced miosis and increased nasal irritation. Mean hydrocodone Cmax following insufflation of HYD coarse particles, HYD fine particles, and hydrocodone powder was 27.5, 36.5, and 105.8 ng/mL, respectively; median Tmax was ≥2-fold longer with either HYD particle size than hydrocodone powder; and (Cmax/Tmax) was 9.5, 13.4, and 82.0 ng/mL/h, respectively. Safety was consistent with that of opioid agonists. CONCLUSIONS: HYD demonstrated reduced intranasal abuse potential compared with hydrocodone powder.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Hydrocodone/administration & dosage , Hydrocodone/pharmacokinetics , Illicit Drugs/pharmacokinetics , Opioid-Related Disorders/metabolism , Administration, Intranasal , Adult , Cross-Over Studies , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Double-Blind Method , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Opioid-Related Disorders/diagnosis , Powders , Young Adult
16.
Arch Toxicol ; 90(10): 2421-44, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27665567

ABSTRACT

This review article covers English-written and PubMed-listed review articles and original studies published between January 2015 and April 2016 dealing with the toxicodynamics and toxicokinetics of new psychoactive substances. Compounds covered include stimulants and entactogens, synthetic cannabinoids, tryptamines, NBOMes, phencyclidine-like drugs, benzodiazepines, and opioids. First, an overview and discussion is provided on timely review articles followed by an overview and discussion on recent original studies. Both sections are then concluded by an opinion on these latest developments. This review shows that the NPS market is still highly dynamic and that the data published on their toxicodynamics and toxicokinetics can hardly keep pace with the appearance of new entities. However, data available are very helpful to understand and predict how NPS may behave in severe intoxication. The currently best-documented parameter is the in vitro metabolism of NPS, a prerequisite to allow detection of NPS in biological matrices in cases of acute intoxications or chronic consumption. However, additional data such as their chronic toxicity are still lacking.


Subject(s)
Illicit Drugs/pharmacokinetics , Illicit Drugs/toxicity , Psychotropic Drugs/pharmacokinetics , Psychotropic Drugs/toxicity , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/toxicity , Animals , Benzodiazepines/pharmacokinetics , Benzodiazepines/toxicity , Cannabinoids/pharmacokinetics , Cannabinoids/toxicity , Central Nervous System Stimulants/pharmacokinetics , Central Nervous System Stimulants/toxicity , Humans , Immunomodulation/drug effects , Metabolic Networks and Pathways/drug effects
17.
Med Intensiva ; 40(4): 230-7, 2016 May.
Article in English, Spanish | MEDLINE | ID: mdl-26548615

ABSTRACT

PURPOSE: To evaluate the impact of a history of harmful use of alcohol (HUA) on sedoanalgesia practices and outcomes in patients on mechanical ventilation (MV). METHODS: A prospective, observational multicentre study was made of all adults consecutively admitted during 30 days to 8 Spanish ICUs. Patients on MV >24h were followed-up on until discharge from the ICU or death. Data on HUA, smoking, the use of illegal (IP) and medically prescribed psychotropics (MPP), sedoanalgesia practices and their related complications (sedative failure [SF] and sedative withdrawal [SW]), as well as outcome, were prospectively recorded. RESULTS: A total of 23.4% (119/509) of the admitted patients received MV >24h; 68.9% were males; age 57.0 (17.9) years; APACHE II score 18.8 (7.2); with a medical cause of admission in 53.9%. Half of them consumed at least one psychotropic agent (smoking 27.7%, HUA 25.2%; MPP 9.2%; and IP 7.6%). HUA patients more frequently required PS (86.7% vs. 64%; p<0.02) and the use of >2 sedatives (56.7% vs. 28.1%; p<0.02). HUA was associated to an eightfold (p<0.001) and fourfold (p<0.02) increase in SF and SW, respectively. In turn, the duration of MV and the stay in the ICU was increased by 151h (p<0.02) and 4.4 days (p<0.02), respectively, when compared with the non-HUA group. No differences were found in terms of mortality. CONCLUSIONS: HUA may be associated to a higher risk of SF and WS, and can prolong MV and the duration of stay in the ICU in critical patients. Early identification could allow the implementation of specific sedation strategies aimed at preventing these complications.


Subject(s)
Alcohol Drinking/adverse effects , Ethanol/adverse effects , Hypnotics and Sedatives/pharmacokinetics , Intensive Care Units , Respiration, Artificial , APACHE , Adult , Aged , Alcohol Drinking/epidemiology , Drug Interactions , Ethanol/pharmacokinetics , Female , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/adverse effects , Illicit Drugs/pharmacokinetics , Male , Middle Aged , Prospective Studies , Psychotropic Drugs/adverse effects , Psychotropic Drugs/pharmacokinetics , Psychotropic Drugs/therapeutic use , Smoking/epidemiology , Spain/epidemiology , Substance Withdrawal Syndrome/etiology , Substance-Related Disorders/epidemiology , Treatment Failure
18.
Drug Metab Rev ; 47(2): 124-74, 2015 May.
Article in English | MEDLINE | ID: mdl-25853390

ABSTRACT

Synthetic cannabinoids (SC), originally developed as research tools, are now highly abused novel psychoactive substances. We present a comprehensive systematic review covering in vivo and in vitro animal and human pharmacokinetics and analytical methods for identifying SC and their metabolites in biological matrices. Of two main phases of SC research, the first investigated therapeutic applications, and the second abuse-related issues. Administration studies showed high lipophilicity and distribution into brain and fat tissue. Metabolite profiling studies, mostly with human liver microsomes and human hepatocytes, structurally elucidated metabolites and identified suitable SC markers. In general, SC underwent hydroxylation at various molecular sites, defluorination of fluorinated analogs and phase II metabolites were almost exclusively glucuronides. Analytical methods are critical for documenting intake, with different strategies applied to adequately address the continuous emergence of new compounds. Immunoassays have different cross-reactivities for different SC classes, but cannot keep pace with changing analyte targets. Gas chromatography and liquid chromatography mass spectrometry assays - first for a few, then numerous analytes - are available but constrained by reference standard availability, and must be continuously updated and revalidated. In blood and oral fluid, parent compounds are frequently present, albeit in low concentrations; for urinary detection, metabolites must be identified and interpretation is complex due to shared metabolic pathways. A new approach is non-targeted HRMS screening that is more flexible and permits retrospective data analysis. We suggest that streamlined assessment of new SC's pharmacokinetics and advanced HRMS screening provide a promising strategy to maintain relevant assays.


Subject(s)
Cannabinoids/analysis , Cannabinoids/pharmacokinetics , Illicit Drugs/analysis , Illicit Drugs/pharmacokinetics , Substance Abuse Detection/methods , Animals , Biotransformation , Brain/metabolism , Cannabinoids/chemistry , Chromatography, Liquid/methods , Humans , Illicit Drugs/chemistry , Liver/metabolism , Mass Spectrometry/methods , Molecular Structure , Organ Specificity , Substance Abuse Detection/instrumentation , Tandem Mass Spectrometry/methods , Tissue Distribution
20.
Pharm Res ; 32(8): 2477-502, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25972096

ABSTRACT

Opioid-related deaths, abuse, and drug interactions are growing epidemic problems that have medical, social, and economic implications. Drug transporters play a major role in the disposition of many drugs, including opioids; hence they can modulate their pharmacokinetics, pharmacodynamics and their associated drug-drug interactions (DDIs). Our understanding of the interaction of transporters with many therapeutic agents is improving; however, investigating such interactions with opioids is progressing relatively slowly despite the alarming number of opioids-mediated DDIs that may be related to transporters. This review presents a comprehensive report of the current literature relating to opioids and their drug transporter interactions. Additionally, it highlights the emergence of transporters that are yet to be fully identified but may play prominent roles in the disposition of opioids, the growing interest in transporter genomics for opioids, and the potential implications of opioid-drug transporter interactions for cancer treatments. A better understanding of drug transporters interactions with opioids will provide greater insight into potential clinical DDIs and could help improve opioids safety and efficacy.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Carrier Proteins/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Carrier Proteins/genetics , Drug Interactions , Humans , Illicit Drugs/pharmacokinetics , Substance-Related Disorders/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL