Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 4 de 4
1.
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.

2.
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
3.
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.

4.
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
...