Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Toxicol Appl Pharmacol ; 479: 116731, 2023 11 15.
Article En | MEDLINE | ID: mdl-37866706

The use and/or misuse of opioids by pregnant women would expose the fetuses to these drugs during critical stages of development with serious effects for the newborn, like the neonatal abstinence syndrome (NAS). We have revisited an established chicken model for NAS to describe the distribution of morphine and methadone to the brain and explore its validity as a valuable alternative to rodent models. For this purpose, chicken eggs were injected with a single dose of 10 mg/kg or 20 mg/kg morphine or 20 mg/kg methadone onto the chorioallantoic membrane (CAM) on embryonal day 13. Whole brains and lungs were harvested and the concentrations of morphine, methadone and their subsequent metabolites (morphine-3-glucuronide and EDDP, respectively) determined in the brain and lungs at different time points using LC-MS/MS. Morphine and methadone, as well as their metabolites, were detected both in the brain and lungs, with significantly higher concentrations in the lungs. Pharmacokinetic modelling showed that the distribution of morphine to the brain followed a first-order absorption with transit compartments and linear elimination, with concentrations linearly dependent on dose. Moreover, methadone, but not morphine, reduced µ receptor (the main morphine receptor) binding, which can be of relevance for opioid tolerance. The present study is the first to report the brain distribution of morphine, which can be described by standard pharmacokinetic processes, and methadone in the developing chicken embryo. The present findings supplement the already established model and support the use of this chicken model to study NAS.


Methadone , Neonatal Abstinence Syndrome , Chick Embryo , Infant, Newborn , Animals , Female , Pregnancy , Humans , Methadone/toxicity , Methadone/therapeutic use , Morphine , Analgesics, Opioid/toxicity , Chickens , Chromatography, Liquid , Drug Tolerance , Tandem Mass Spectrometry , Neonatal Abstinence Syndrome/drug therapy , Brain , Receptors, Opioid, mu
2.
Basic Clin Pharmacol Toxicol ; 133(4): 418-427, 2023 Oct.
Article En | MEDLINE | ID: mdl-37452619

Immunotherapeutic interventions that block drug effects by binding drug molecules to specific antibodies in the bloodstream have shown promising effects in animal studies. For heroin, which effects are mainly mediated by the metabolites 6-acetylmorphine (6-AM; also known as 6-monoacetylmorphine or 6-MAM) and morphine, the optimal antibody specificity has been discussed. In rodents, 6-AM specific antibodies have been recommended based on the rapid metabolism of heroin to 6-AM in the bloodstream. Since the metabolic rate of heroin in blood is unsettled in humans, we examined heroin metabolism with state-of-the-art analytical methodology (UHPLC-MS/MS) in freshly drawn human whole blood incubated with a wide range of heroin concentrations (1-500 µM). The half-life of heroin was highly concentration dependent, ranging from 1.2-1.7 min for concentrations at or above 25 µM, and gradually increasing to approximately 20 min for 1 µM heroin. At concentrations that can be attained in the bloodstream shortly after an i.v. injection, approximately 70% was transformed into 6-AM within 3 min, similar to previous observations in vivo. Our results indicate that blood enzymes play a more important role for the rapid metabolism of heroin in humans than previously assumed. This points to 6-AM as an important target for an efficient immunotherapeutic approach to block heroin effects in humans.

3.
Int J Mol Sci ; 22(21)2021 Nov 05.
Article En | MEDLINE | ID: mdl-34769427

Methiopropamine is a novel psychoactive substance (NPS) that is associated with several cases of clinical toxicity, yet little information is available regarding its neuropharmacological properties. Here, we employed in vitro and in vivo methods to compare the pharmacokinetics and neurobiological effects of methiopropamine and its structural analog methamphetamine. Methiopropamine was rapidly distributed to the blood and brain after injection in C57BL/6 mice, with a pharmacokinetic profile similar to that of methamphetamine. Methiopropamine induced psychomotor activity, but higher doses were needed (Emax 12.5 mg/kg; i.p.) compared to methamphetamine (Emax 3.75 mg/kg; i.p.). A steep increase in locomotor activity was seen after a modest increase in the methiopropamine dose from 10 to 12.5 mg/kg, suggesting that a small increase in dosage may engender unexpectedly strong effects and heighten the risk of unintended overdose in NPS users. In vitro studies revealed that methiopropamine mediates its effects through inhibition of norepinephrine and dopamine uptake into presynaptic nerve terminals (IC50 = 0.47 and 0.74 µM, respectively), while the plasmalemmal serotonin uptake and vesicular uptake are affected only at high concentrations (IC50 > 25 µM). In summary, methiopropamine closely resembles methamphetamine with regard to its pharmacokinetics, pharmacodynamic effects and mechanism of action, with a potency that is approximately five times lower than that of methamphetamine.


Brain/drug effects , Methamphetamine/analogs & derivatives , Methamphetamine/pharmacology , Methamphetamine/pharmacokinetics , Neuropharmacology/methods , Thiophenes/pharmacology , Thiophenes/pharmacokinetics , Animals , Brain/metabolism , Central Nervous System Stimulants/chemistry , Central Nervous System Stimulants/pharmacokinetics , Central Nervous System Stimulants/pharmacology , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Tissue Distribution
4.
J Pharmacol Toxicol Methods ; 112: 107105, 2021.
Article En | MEDLINE | ID: mdl-34284116

INTRODUCTION: Rodent models are routinely used to assess the safety and developmental toxicity of pharmaceuticals, along with analysis of their distribution. These models require sacrifice of parent females, have challenges in the estimation of the number of embryos and stage of development, and are expensive and time-consuming. In this study, we used fertilized chicken eggs as an alternative model to address drug distribution to the developing brain of two antiepileptic drugs, valproic acid (VPA) and lamotrigine (LTG) at two developmental stages. METHODS: VPA or LTG was injected into the allantois of the egg on embryonic day 13 (E13) or E16. Whole chicken brains were harvested at time-points of 5 min to 24 h and the concentrations of the drugs determined using GC/MS and LC-MS/MS, for VPA and LTG, respectively. RESULTS: VPA and LTG had distinct absorption and elimination phases and were found in the brain as early as 5-15 min after injection. Both drugs reached the brain in clinically relevant concentrations, with Cmax 10-30% of the calculated concentration assuming uniform distribution throughout the egg. LTG concentrations were higher when injected at E13 compared to E16. CONCLUSION: The chicken embryo model may be a suitable alternative animal model for preclinical drug distribution studies. It enables to easily approach antenatal development on an individual level, with a precise number of experimental animals, high reproducibility and low time and cost. Knowledge of the concentrations reaching the brain at different developmental stages with different drugs is important for the planning and interpretation of neurodevelopmental toxicity studies.


Epilepsy , Pharmaceutical Preparations , Animals , Anticonvulsants/therapeutic use , Anticonvulsants/toxicity , Brain , Chick Embryo , Chickens , Chromatography, Liquid , Disease Models, Animal , Drug Interactions , Epilepsy/drug therapy , Female , Pregnancy , Reproducibility of Results , Tandem Mass Spectrometry , Triazines , Valproic Acid/toxicity
5.
Int J Dev Neurosci ; 80(5): 443-453, 2020 Aug.
Article En | MEDLINE | ID: mdl-32484968

There is a growing concern related to the use of opioid maintenance treatment during pregnancy. Studies in both humans and animals have reported reduced cognitive functioning in offspring prenatally exposed to methadone or buprenorphine; however, little is known about the neurobiological mechanisms underlying these impairments. To reveal possible neurobiological effects of such in utero exposure, we examined brain tissue from methadone- and buprenorphine-exposed rat offspring previously shown to display impaired learning and memory. We studied µ-opioid receptor (MOR) and N-methyl-D-aspartate receptor (NMDAR) binding in the rat offspring cerebrum during development and in the hippocampus at young adulthood. Moreover, we examined activation of the Ca2+ /calmodulin-dependent protein kinase II (CaMKII) and the extracellular signal-regulated kinase (ERK), which are central in the downstream signaling of these receptors. The methadone- and buprenorphine-exposed rat pups displayed reduced MOR binding up to two weeks after birth, whereas the NMDAR binding was unaffected. Prenatal exposure to methadone or buprenorphine also resulted in decreased activation of CaMKII and/or ERK during development, while young adult offspring displayed increased hippocampal ERK activation. In conclusion, our findings suggest that prenatal exposure to exogenous opioids, such as methadone or buprenorphine, may disturb the endogenous opioid system during development, with long-term effects on proteins important for cognitive functioning.

6.
Drug Alcohol Depend ; 212: 108008, 2020 07 01.
Article En | MEDLINE | ID: mdl-32402939

BACKGROUND: Concerns have been raised about the use of opioid maintenance treatment (OMT) during pregnancy and negative effects for the offspring. While neonatal outcomes and short-term effects are relatively well described, studies examining long-term effects in adolescents and adults are absent. The aim of the present study was to examine effects on learning and memory in young adult rats prenatally exposed to methadone or buprenorphine. METHODS: Female rats were implanted with a 28-day osmotic minipump delivering methadone (10 mg/kg/day), buprenorphine (1 mg/kg/day) or vehicle 5 days prior to mating. To examine possible effects on cognitive functioning, young adult offspring were included in three different behavioral tests that examine recognition memory, nonspatial, and spatial learning and memory. In addition, offspring growth and maternal behavior after birh were investigated. RESULTS: Prenatal exposure to methadone or buprenorphine caused impaired recognition memory and nonspatial reference learning and memory in young adult rats compared with the vehicle-treated group. Methadone-exposed offspring, but not the buprenorphine-exposed, also showed reduced long-term spatial memory. We did not observe any changes in maternal behavior or offspring growth after prenatal exposure to methadone or buprenorphine, suggesting that the impaired cognitive functioning is due to the opioid exposure rather than reduced maternal caregiving. CONCLUSION: The present findings of long-term cognitive impairments in methadone- and buprenorphine-exposed offspring points to a negative impact of OMT on neurobiological development.


Buprenorphine/adverse effects , Cognition/drug effects , Methadone/adverse effects , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/psychology , Animals , Animals, Newborn , Buprenorphine/administration & dosage , Cognition/physiology , Female , Infusion Pumps, Implantable , Male , Memory/drug effects , Memory/physiology , Methadone/administration & dosage , Opiate Substitution Treatment/adverse effects , Pregnancy , Random Allocation , Rats , Rats, Sprague-Dawley
7.
Early Hum Dev ; 143: 104997, 2020 04.
Article En | MEDLINE | ID: mdl-32146140

AIM: To combine meta-analyses of multiple long-term outcomes in children prenatally exposed to methadone or buprenorphine through their mothers' Opioid maintenance therapy (OMT) with a systematic review of similar outcomes in experimental animals. METHOD: The Medline, Embase, Web of Science, CINAHL, Cochrane and Epistemonikos databases were searched through August 30, 2018. Clinical studies measuring effects on cognitive, behavioral or visual outcomes in 3 months or older children prenatally exposed to OMT and control group(s) were included for meta-analyses. Experimental animal studies with similar exposures and outcomes were included in a systematic review. The three authors independently performed abstract screenings and full-text reviews, and extracted the data. One author performed the meta-analyses. RESULTS: The pooled results of the meta-analyses showed worse cognitive, psychomotor, behavioral, attentional and executive functioning, and affected vision in children born to mothers who were in OMT during pregnancy compared to children without prenatal drug exposure (overall effect size = 0.49, 95% confidence interval = 0.38, 0.59, p < 0.00001). Many of the experimental animal studies showed impaired outcomes after prenatal exposure to methadone or buprenorphine. The clinical results may be biased, e.g., with the OMT group having more concurrent risk factors than the unexposed comparison group. There are few studies of older children. CONCLUSION: Children born to mothers in OMT show worse outcomes for a number of different behaviors and impaired vision compared to children born to nonusers. Experimental animal studies indicate that there might be a causal relationship between prenatal methadone or buprenorphine exposure and subsequent negative outcomes.


Analgesics, Opioid/adverse effects , Buprenorphine/adverse effects , Methadone/adverse effects , Neurodevelopmental Disorders/epidemiology , Prenatal Exposure Delayed Effects/epidemiology , Vision Disorders/epidemiology , Animals , Child , Female , Humans , Infant, Newborn , Male , Neurodevelopmental Disorders/etiology , Opiate Substitution Treatment/adverse effects , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Vision Disorders/etiology
8.
Addict Biol ; 25(2): e12727, 2020 03.
Article En | MEDLINE | ID: mdl-30788879

We have previously demonstrated that heroin's first metabolite, 6-acetylmorphine (6-AM), is an important mediator of heroin's acute effects. However, the significance of 6-AM to the rewarding properties of heroin still remains unknown. The present study therefore aimed to examine the contribution of 6-AM to heroin-induced reward and locomotor sensitization. Mice were tested for conditioned place preference (CPP) induced by equimolar doses of heroin or 6-AM (1.25-5 µmol/kg). Psychomotor activity was recorded during the CPP conditioning sessions for assessment of drug-induced locomotor sensitization. The contribution of 6-AM to heroin reward and locomotor sensitization was further examined by pretreating mice with a 6-AM specific antibody (anti-6-AM mAb) 24 hours prior to the CPP procedure. Both heroin and 6-AM induced CPP in mice, but heroin generated twice as high CPP scores compared with 6-AM. Locomotor sensitization was expressed after repeated exposure to 2.5 and 5 µmol/kg heroin or 6-AM, but not after 1.25 µmol/kg, and we found no correlation between the expression of CPP and the magnitude of locomotor sensitization for either opioid. Pretreatment with anti-6-AM mAb suppressed both heroin-induced and 6-AM-induced CPP and locomotor sensitization. These findings provide evidence that 6-AM is essential for the rewarding and sensitizing properties of heroin; however, heroin caused stronger reward compared with 6-AM. This may be explained by the higher lipophilicity of heroin, providing more efficient drug transfer to the brain, ensuring rapid increase in the brain 6-AM concentration.


Brain/drug effects , Heroin/pharmacology , Locomotion/drug effects , Morphine Derivatives/blood , Opioid-Related Disorders/physiopathology , Reward , Analgesics, Opioid/blood , Analgesics, Opioid/pharmacology , Animals , Brain/metabolism , Brain/physiopathology , Conditioning, Psychological/drug effects , Disease Models, Animal , Heroin/blood , Male , Mice , Mice, Inbred C57BL , Opioid-Related Disorders/blood , Opioid-Related Disorders/metabolism
9.
Int J Dev Neurosci ; 78: 19-27, 2019 Nov.
Article En | MEDLINE | ID: mdl-31351113

The use of opioids during pregnancy has been associated with neurodevelopmental toxicity in exposed children, leading to cognitive and behavioural deficits later in life. The N-methyl-D-aspartate receptor (NMDAR) subunit GluN2B plays critical roles in cerebellar development, and methadone has been shown to possess NMDAR antagonist effect. Consequently, we wanted to explore if prenatal opioid exposure affected GluN2B subunit expression and NMDAR function in rat and chicken cerebellum. Pregnant rats were exposed to methadone (10 mg/kg/day) or buprenorphine (1 mg/kg/day) for the whole period of gestation, using an osmotic minipump. To further examine potential effects of prenatal opioid exposure in a limited time window, chicken embryos were exposed to a 20 mg/kg dose of methadone or morphine on embryonic days 13 and 14. Western blot analysis of cerebella isolated from 14 days old rat pups exposed to buprenorphine showed significantly lower level of the GluN2B subunit, while the opioid exposed chicken embryo cerebellar GluN2B expression remained unaffected at embryonic day 17. However, we observed increased NMDA/glycine-induced calcium influx in cerebellar granule neurone cultures from opioid exposed chicken embryos. We conclude that prenatal opioid exposure leads to opioid receptor-dependent reduction in the postnatal expression of GluN2B in rat cerebella, and increase in NMDA-induced calcium influx in chicken embryo cerebella.


Analgesics, Opioid/pharmacology , Buprenorphine/pharmacology , Cerebellum/drug effects , Methadone/pharmacology , Morphine/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cerebellum/embryology , Cerebellum/metabolism , Chickens , Female , Pregnancy , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
10.
J Pharmacol Exp Ther ; 371(1): 130-137, 2019 10.
Article En | MEDLINE | ID: mdl-31358559

Experimental animal studies are valuable in revealing a causal relationship between prenatal exposure to opioid maintenance treatment (OMT) and subsequent effects; however, previous animal studies of OMT during pregnancy have been criticized for their lack of clinical relevance because of their use of high drug doses and the absence of pharmacokinetic data. Hence, the aim of this study was to determine blood and brain concentrations in rat dams, fetuses, and offspring after continuous maternal exposure to methadone or buprenorphine during gestation and to examine the offspring for neonatal outcomes and withdrawal symptoms. Female rats were implanted with a 28-day osmotic minipump delivering methadone (10 mg/kg per day), buprenorphine (1 mg/kg per day) or vehicle 5 days before mating. Continuous exposure to methadone or buprenorphine induced stable blood concentrations in the dams of 0.25 ± 0.02 µM and 5.65 ± 0.16 nM, respectively. The fetal brain concentration of methadone (1.89 ± 0.35 nmol/g) was twice as high as that in the maternal brain, whereas the fetal brain concentration of buprenorphine (20.02 ± 4.97 pmol/g) was one-third the maternal brain concentration. The opioids remained in the offspring brain several days after the exposure ceased. Offspring prenatally exposed to methadone, but not buprenorphine, displayed reduced body weight and length and increased corticosterone levels. No significant changes in ultrasonic vocalizations were revealed. Our data in rat fetuses and neonates indicate that OMT with buprenorphine may be a better choice than methadone during pregnancy. SIGNIFICANCE STATEMENT: Concern has been raised about the use of opioid maintenance treatment during pregnancy because of the important role of the endogenous opioid system in brain development. Here, we show that the methadone concentration in the fetal rat brain was twice as high as that in the maternal brain, whereas the buprenorphine concentration was one-third the maternal concentration. Furthermore, buprenorphine allowed more favorable birth outcomes, suggesting that buprenorphine may be a better choice during pregnancy.


Analgesics, Opioid/pharmacokinetics , Brain/metabolism , Buprenorphine/pharmacokinetics , Maternal-Fetal Exchange , Methadone/pharmacokinetics , Analgesics, Opioid/adverse effects , Animals , Body Weight , Brain/drug effects , Brain/embryology , Buprenorphine/adverse effects , Female , Fetus/drug effects , Fetus/metabolism , Male , Methadone/adverse effects , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Sprague-Dawley
11.
J Pharmacol Exp Ther ; 368(1): 106-115, 2019 01.
Article En | MEDLINE | ID: mdl-30361238

Escalating opioid use among fertile women has increased the number of children being exposed to opioids during fetal life. Furthermore, accumulating evidence links prenatal opioid exposure, including opioid maintenance treatment, to long-term negative effects on cognition and behavior, and presses the need to explore novel treatment strategies for pregnant opioid users. The present study examined the potential of a monoclonal antibody (mAb) targeting heroin's first metabolite, 6-acetylmorphine (6-AM), in providing fetal protection against harmful effects of prenatal heroin exposure in mice. First, we examined anti-6-AM mAb's ability to block materno-fetal transfer of active metabolites after maternal heroin administration. Next, we studied whether maternal mAb pretreatment could prevent adverse effects in neonatal and adolescent offspring exposed to intrauterine heroin (3 × 1.05 mg/kg). Anti-6-AM mAb pretreatment of pregnant dams profoundly reduced the distribution of active heroin metabolites to the fetal brain. Furthermore, maternal mAb administration prevented hyperactivity and drug sensitization in adolescent female offspring prenatally exposed to heroin. Our findings demonstrate that passive immunization with a 6-AM-specific antibody during pregnancy provides fetal neuroprotection against heroin metabolites, and thereby prevents persistent adverse behavioral effects in the offspring. An immunotherapeutic approach to protect the fetus against long-term effects of prenatal drug exposure has not been reported previously, and should be further explored as prophylactic treatment of pregnant heroin users susceptible to relapse.


Antibodies, Monoclonal/therapeutic use , Heroin/adverse effects , Locomotion/drug effects , Morphine Derivatives/antagonists & inhibitors , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/prevention & control , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Animals , Animals, Newborn , Antibodies, Monoclonal/pharmacology , Female , Heroin/administration & dosage , Locomotion/physiology , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/psychology , Random Allocation
12.
Forensic Sci Int ; 290: 121-128, 2018 Sep.
Article En | MEDLINE | ID: mdl-30015276

BACKGROUND: In heroin-related deaths, it is often of interest to determine the approximate time span between intake of heroin and death, and to decide whether heroin or other opioids have been administered. In some autopsy cases, peripheral blood cannot be sampled due to decomposition, injuries or burns. The aim of the present study was to investigate whether measurements of heroin metabolites in matrices other than peripheral blood can be used to differentiate between rapid and delayed heroin deaths, and if morphine/codeine ratios measured in other matrices can separate heroin from codeine intakes. METHODS: In this study, we included 51 forensic autopsy cases where morphine was detected in peripheral blood. Samples were collected from peripheral and cardiac blood, pericardial fluid, psoas and lateral vastus muscles, vitreous humor and urine. The opioid analysis included 6-acetylmorphine (6-AM), morphine, morphine-3-glucuronide (M3G), morphine-6-glucuronide (M6G) and codeine. Urine was only used for qualitative detection of 6-AM. 45 heroin-intake cases were divided into rapid deaths (n=24), based on the detection of 6-AM in blood, or delayed deaths (n=21), where 6-AM was detected in at least one other matrix but not in blood. An additional 6 cases were classified as codeine-intake cases, based on a morphine/codeine ratio below unity (<1) in peripheral blood, without detecting 6-AM in any matrix. RESULTS: The median morphine concentrations were significantly higher in the rapid compared with the delayed heroin deaths in all matrices (p=0.004 for vitreous humor and p<0.001 for the other matrices). In the rapid heroin deaths, the M3G/morphine concentration ratios were significantly lower than in the delayed deaths both in peripheral and cardiac blood (p<0.001), as well as in pericardial fluid (p<0.001) and vitreous humor (p=0.006), but not in muscle. The morphine/codeine ratios measured in cardiac blood, pericardial fluid and the two muscle samples resembled the ratios in peripheral blood, although codeine was less often detected in other matrices than peripheral blood. CONCLUSIONS: Measurements of heroin-metabolites in cardiac blood, pericardial fluid and vitreous humor provide information comparable to that of peripheral blood regarding rapid and delayed heroin deaths, e.g. M3G/morphine ratios <2 indicate a rapid death while ratios >3 indicate a delayed death. However, considerable overlap in results from rapid and delayed deaths was observed, and measurements in muscle appeared less useful. Furthermore, matrices other than peripheral blood can be used to investigate morphine/codeine ratios, but vitreous humor seems less suited.


Codeine/analysis , Heroin/poisoning , Morphine Derivatives/analysis , Morphine/analysis , Postmortem Changes , Drug Overdose , Forensic Toxicology , Heroin Dependence/mortality , Humans , Muscle, Skeletal/chemistry , Pericardial Fluid/chemistry , Time Factors , Vitreous Body/chemistry
13.
J Anal Toxicol ; 42(5): 311-320, 2018 Jun 01.
Article En | MEDLINE | ID: mdl-29409037

In some forensic autopsies blood is not available, and other matrices are sampled for toxicological analysis. The aims of the present study were to examine whether heroin metabolites can be detected in different post-mortem matrices, and investigate whether analyses in other matrices can give useful information about concentrations in peripheral blood. Effects of ethanol on the metabolism and distribution of heroin metabolites were also investigated. We included 45 forensic autopsies where morphine was detected in peripheral blood, concomitantly with 6-acetylmorphine (6-AM) detected in any matrix. Samples were collected from peripheral blood, cardiac blood, pericardial fluid, psoas muscle, lateral vastus muscle, vitreous humor and urine. Opioid analysis included 6-AM, morphine, codeine, and morphine glucuronides. The 6-AM was most often detected in urine (n = 39) and vitreous humor (n = 38). The median morphine concentration ratio relative to peripheral blood was 1.3 (range 0-3.6) for cardiac blood, 1.4 (range 0.07-5.3) for pericardial fluid, 1.2 (range 0-19.2) for psoas muscle, 1.1 (range 0-1.7) for lateral vastus muscle and 0.4 (range 0.2-3.2) for vitreous humor. The number of 6-AM positive cases was significantly higher (P = 0.03) in the ethanol positive group (n = 6; 86%) compared to the ethanol negative group (n = 14; 37%) in peripheral blood. The distribution of heroin metabolites to the different matrices was not significantly different between the ethanol positive and the ethanol negative group. This study shows that toxicological analyses of several matrices could be useful in heroin-related deaths. Urine and vitreous humor are superior for detection of 6-AM, while concentrations of morphine could be assessed from peripheral or cardiac blood, pericardial fluid, psoas muscle and lateral vastus muscle.


Alcohol Drinking/metabolism , Forensic Toxicology/methods , Heroin/analogs & derivatives , Morphine Derivatives/analysis , Morphine/analysis , Opioid-Related Disorders/metabolism , Substance Abuse Detection/methods , Alcohol Drinking/blood , Alcohol Drinking/urine , Cadaver , Codeine/analysis , Codeine/blood , Codeine/urine , Glucuronides/analysis , Glucuronides/blood , Glucuronides/urine , Heroin/analysis , Heroin/blood , Heroin/urine , Humans , Morphine/blood , Morphine/urine , Morphine Derivatives/blood , Morphine Derivatives/urine , Narcotics/analysis , Narcotics/blood , Narcotics/chemistry , Narcotics/urine , Norway , Opioid-Related Disorders/blood , Opioid-Related Disorders/urine , Pericardial Fluid/chemistry , Psoas Muscles/chemistry , Quadriceps Muscle/chemistry , Tissue Distribution , Toxicokinetics , Vitreous Body/chemistry
14.
Article En | MEDLINE | ID: mdl-29175696

A novel ion pair reversed phase ultra high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method for simultaneous determination of the stress hormones adrenaline, noradrenaline and corticosterone in rodent blood was developed and fully validated. Separations were performed on an Acquity HSS T3 column (2.1mm i.d.×100mm, 1.8µm) with gradient elution and a runtime of 5.5min. The retention of adrenaline and noradrenaline was substantially increased by employing the ion pair reagent heptafluorobutyric acid (HFBA). Ion pair reagents are usually added to the mobile phase only, but we demonstrate for the first time that including HFBA to the sample reconstitution solvent as well, has a major impact on the chromatography of these compounds. The stability of adrenaline and corticosterone in rodent blood was investigated using the surrogate analytes adrenaline-d3 and corticosterone-d8. The applicability of the described method was demonstrated by measuring the concentration of stress hormones in rodent blood samples.


Chromatography, Reverse-Phase/methods , Corticosterone/blood , Epinephrine/blood , Norepinephrine/blood , Tandem Mass Spectrometry/methods , Animals , Chromatography, High Pressure Liquid/methods , Drug Stability , Limit of Detection , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Reproducibility of Results
15.
J Pharmacol Exp Ther ; 358(2): 209-15, 2016 08.
Article En | MEDLINE | ID: mdl-27278234

Toll-like receptor 4 (TLR4) signaling is implied in opioid reinforcement, reward, and withdrawal. Here, we explored whether TLR4 signaling is involved in the acute psychomotor-stimulating effects of heroin, 6-acetylmorphine (6-AM), and morphine as well as whether there are differences between the three opioids regarding TLR4 signaling. To address this, we examined how pretreatment with (+)-naloxone, a TLR4 active but opioid receptor (OR) inactive antagonist, affected the acute increase in locomotor activity induced by heroin, 6-AM, or morphine in mice. We also assessed the effect of pretreatment with (-)-naloxone, a TLR4 and OR active antagonist, as well as the pharmacokinetic profiles of (+) and (-)-naloxone in the blood and brain. We found that (-)-naloxone reduced acute opioid-induced locomotor activity in a dose-dependent manner. By contrast, (+)-naloxone, administered in doses assumed to antagonize TLR4 but not ORs, did not affect acute locomotor activity induced by heroin, 6-AM, or morphine. Both naloxone isomers exhibited similar concentration versus time profiles in the blood and brain, but the brain concentrations of (-)-naloxone reached higher levels than those of (+)-naloxone. However, the discrepancies in their pharmacokinetic properties did not explain the marked difference between the two isomers' ability to affect opioid-induced locomotor activity. Our results underpin the importance of OR activation and do not indicate an apparent role of TLR4 signaling in acute opioid-induced psychomotor stimulation in mice. Furthermore, there were no marked differences between heroin, 6-AM, and morphine regarding involvement of OR or TLR4 signaling.


Central Nervous System Stimulants/pharmacology , Heroin/pharmacology , Morphine Derivatives/pharmacology , Morphine/pharmacology , Naloxone/chemistry , Naloxone/pharmacology , Animals , Brain/cytology , Brain/drug effects , Brain/metabolism , Brain/physiology , Heroin/antagonists & inhibitors , Locomotion/drug effects , Male , Mice , Morphine/antagonists & inhibitors , Morphine Derivatives/antagonists & inhibitors , Naloxone/blood , Naloxone/pharmacokinetics , Signal Transduction/drug effects , Stereoisomerism , Structure-Activity Relationship , Toll-Like Receptor 4/metabolism
16.
J Pharmacol Exp Ther ; 358(2): 181-9, 2016 08.
Article En | MEDLINE | ID: mdl-27217591

Immunotherapy can provide a supplemental treatment strategy against heroin use on the principle of sequestering the active drug in the bloodstream, thereby reducing its distribution to the brain. Previous studies have shown that heroin's first metabolite, 6-monoacetylmorphine (6-MAM), is the main mediator of acute heroin effects. The objective of the present study was to characterize the pharmacological potential of a monoclonal antibody against 6-MAM (anti-6-MAM mAb) to counteract the heroin response. The individual contributions from heroin and 6-MAM to heroin effects were also examined by pretreating mice with anti-6-MAM mAb (10-100 mg/kg) prior to either heroin or 6-MAM injection (1.25-2.5 µmol/kg). The opioid-induced behavioral response was assessed in a locomotor activity test, followed by opioid and antibody quantification in blood and brain tissue. Pretreatment with mAb caused a profound reduction of heroin- and 6-MAM-induced behavior, accompanied by correspondingly decreased levels of 6-MAM in brain tissue. mAb pretreatment was more efficient against 6-MAM injection than against heroin, leading to an almost complete blockade of 6-MAM-induced effects. mAb pretreatment was unable to block the immediate (5-minute) transport of active metabolites across the blood-brain barrier after heroin injection, indicating that heroin itself appears to enhance the immediate delivery of 6-MAM to the brain. The current study provides additional evidence that 6-MAM sequestration is crucial for counteracting the acute heroin response, and demonstrates the pharmacological potential of immunotherapy against heroin use.


Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/pharmacokinetics , Heroin/pharmacology , Locomotion/drug effects , Morphine Derivatives/immunology , Animals , Antibodies, Monoclonal/immunology , Brain/drug effects , Brain/metabolism , Brain/physiology , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Morphine Derivatives/blood , Morphine Derivatives/metabolism , Tissue Distribution
17.
J Pharmacol Exp Ther ; 349(3): 568-76, 2014 Jun.
Article En | MEDLINE | ID: mdl-24700886

Immunotherapy against drugs of abuse is being studied as an alternative treatment option in addiction medicine and is based on antibodies sequestering the drug in the bloodstream and blocking its entry into the brain. Producing an efficient vaccine against heroin has been considered particularly challenging because of the rapid metabolism of heroin to multiple psychoactive molecules. We have previously reported that heroin's first metabolite, 6-monoacetylmorphine (6-MAM), is the predominant mediator for heroin's acute behavioral effects and that heroin is metabolized to 6-MAM primarily prior to brain entry. On this basis, we hypothesized that antibody sequestration of 6-MAM is sufficient to impair heroin-induced effects and therefore examined the effects of a monoclonal antibody (mAb) specific for 6-MAM. In vitro experiments in human and rat blood revealed that the antibody was able to bind 6-MAM and block the metabolism to morphine almost completely, whereas the conversion of heroin to 6-MAM remained unaffected. Mice pretreated with the mAb toward 6-MAM displayed a reduction in heroin-induced locomotor activity that corresponded closely to the reduction in brain 6-MAM levels. Intraperitoneal and intravenous administration of the anti-6-MAM mAb gave equivalent protection against heroin effects, and the mAb was estimated to have a functional half-life of 8 to 9 days in mice. Our study implies that an antibody against 6-MAM is effective in counteracting heroin effects.


Antibodies, Monoclonal/immunology , Brain/metabolism , Heroin Dependence/drug therapy , Heroin/adverse effects , Heroin/blood , Morphine Derivatives/immunology , Adult , Animals , Antibodies, Monoclonal/administration & dosage , Binding Sites , Brain/drug effects , Brain/immunology , Heroin/chemistry , Heroin/pharmacokinetics , Heroin Dependence/immunology , Heroin Dependence/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Morphine Derivatives/blood , Morphine Derivatives/chemistry , Morphine Derivatives/pharmacokinetics , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Tissue Distribution
18.
Pharmacol Biochem Behav ; 122: 82-8, 2014 Jul.
Article En | MEDLINE | ID: mdl-24699386

The opioid receptor antagonist 3-methoxynaltrexone (3-MeONtx) has previously been shown in rodents to selectively reverse the analgesic actions of heroin and its metabolites 6-monoacetylmorphine (6-MAM), and morphine-6-glucuronide (M6G), but not that of morphine. Based on these and other results, a heroin/6-MAM/M6G µ-opioid receptor binding site or subreceptor mediating their analgesic activity has been proposed. It is however unknown whether this also accounts for the acute psychomotor stimulating properties of these opioids. The aim of the present study was therefore to explore if the acute psychomotor stimulating effects of heroin, 6-MAM, and morphine are mediated by distinct µ-opioid receptor binding sites or subreceptors. To address this aim, we examined how pretreatment with 3-MeONtx or naltrexone (NTX) affected the acute increase in locomotor activity induced by heroin, 6-MAM, or morphine in mice. The pharmacokinetic profiles of 3-MeONtx and NTX were also assessed in mouse brain. We found that 3-MeONtx similarly antagonized the acute increase in locomotor activity induced by equipotent doses of heroin, 6-MAM, or morphine. This antagonistic effect was comparable to the one observed following administration of NTX, and both antagonists gave similar pharmacokinetic profiles in mouse brain. Our findings do not support that different µ-opioid receptor subtypes or a distinct binding site at the µ-opioid receptor is involved in morphine-induced versus heroin/6-MAM-induced psychomotor activation. This might suggest that the opioid-induced psychomotor stimulation is mediated by different µ-opioid subreceptors than those responsible for their analgesic effects.


Heroin/pharmacology , Morphine Derivatives/pharmacology , Motor Activity/drug effects , Naltrexone/analogs & derivatives , Narcotic Antagonists/pharmacology , Psychomotor Performance/drug effects , Animals , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Naltrexone/pharmacology , Psychomotor Performance/physiology , Time Factors
19.
Eur J Clin Pharmacol ; 69(9): 1683-7, 2013 Sep.
Article En | MEDLINE | ID: mdl-23739999

PURPOSE: Ethanol and morphine are both substrates of uridine diphosphate glucuronosyl transferases (UGTs). A pharmacokinetic interaction between ethanol and morphine is suggested from in vitro studies, but to our knowledge not documented in vivo. The aim of this study was to compare the ratios between M6G and morphine and between M3G and morphine in blood samples from suspected drunk and drugged drivers, with and without presence of ethanol. METHODS: The data in the present study constitute all cases of suspected drunk and drugged driving positive for morphine, collected in Norway, in the period November 1st 2009 to December 1st 2012, during which all morphine positive cases were also routinely analysed for M6G and M3G. The cases were divided into two groups; one where morphine was present together with ethanol (group 1) and one where morphine was present in the absence of ethanol (group 2). RESULTS: The ratios between M3G and morphine was lower in the ethanol positive cases, i.e. mean 4.9 (95 % CI 4.03-5.79) in group 1 and mean 6.7 (95 % CI 6.35-7.00) in group 2 (p < 0.001). The ratios between M6G and morphine was also lower in the ethanol positive cases, i.e. mean 0.62 (95 % CI 0.42-0.81) in group 1 and mean 0.96 (95 % CI 0.89-1.02) in group 2 (p = 0.001). CONCLUSIONS: This study indicated that the metabolism of morphine may be changed in the presence of ethanol, resulting in less formation of the metabolites. This could lead to increased terminal half-life for morphine and also possibly more accumulation after repeated dosing.


Analgesics, Opioid/pharmacokinetics , Ethanol/administration & dosage , Morphine Derivatives/blood , Morphine/pharmacokinetics , Adult , Analgesics, Opioid/administration & dosage , Female , Humans , Male , Morphine/administration & dosage , Norway
20.
J Anal Toxicol ; 33(7): 345-50, 2009 Sep.
Article En | MEDLINE | ID: mdl-19796503

A high-performance liquid chromatography-tandem mass spectrometry (LC-MS-MS) method has been developed for the quantitative analysis of heroin and its major metabolites 6-acetylmorphine, morphine, morphine-3-glucuronide and morphine-6-glucuronide in blood and brain tissue, using 0.1-mL samples. We evaluated this method for analysis of heroin and its metabolites in samples from heroin treated mice. Ice-cold acidic buffer containing sodium fluoride was immediately added to blood and brain homogenate samples. Sample preparation was achieved by protein precipitation on ice-bath, using a mixture of ice-cold acetonitrile and methanol. The supernatant was evaporated to dryness, reconstituted with mobile phase, and injected into the chromatographic system. Separation was performed on a Xterra C18 column with gradient elution. The MS analysis was performed in positive ion mode, and multiple reaction monitoring (MRM) was used for drug quantification. The limits of quantification for the different opiates varied from 0.0007 to 0.02 mg/L in blood and from 0.002 to 0.06 microg/g in brain tissue. Day-to-day relative standard deviation ranged from 3.1 to 14.5%, and within-day variation ranged from 2.1 to 11.4%. The recoveries were between 80 and 111%. The stability of heroin was tested, and the study showed that heroin is more stable in brain tissue than in blood.


Brain/metabolism , Heroin/analysis , Narcotics/analysis , Animals , Chromatography, High Pressure Liquid , Heroin/blood , Mice , Mice, Inbred C57BL , Narcotics/blood , Reference Standards , Reproducibility of Results , Solutions , Spectrophotometry, Ultraviolet , Tandem Mass Spectrometry
...